1
|
Gong X, Jiao Y, Hu H, Zhang R, Jia W, Zhao J, Liu Z, Xin Y, Han W. A prospective randomized controlled study of multi-intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure and reduced ejection fraction (PRIME-HFrEF) trial: Rationale and design. Contemp Clin Trials Commun 2024; 41:101350. [PMID: 39246626 PMCID: PMC11377133 DOI: 10.1016/j.conctc.2024.101350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/12/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024] Open
Abstract
Background and objective The use of mesenchymal stem cells for heart failure treatment has gained increasing interest. However, most studies have relied on a single injection approach, with no research yet confirming the effects of multiple administrations. The present trial aims to investigate the safety and efficacy of multi-intravenous infusion of umbilical cord-mesenchymal stem cells (UC-MSCs) in patients with heart failure and reduced ejection fraction (HFrEF). Methods The PRIME-HFrEF trial is a single-center, prospective, randomized, triple-blinded, placebo-controlled trial of multi-intravenous infusion of UC-MSCs in HFrEF patients. A total of 40 patients meeting the inclusion criteria for HFrEF were enrolled and randomized 1:1 to the MSC group or the placebo group. Patients enrolled will receive intravenous injections of either UC-MSCs or placebo every 6 weeks for three times. Both groups will be followed up for 12 months. The primary safety endpoint is the incidence of serious adverse events. The primary efficacy endpoint is a change in left ventricular ejection fraction (LVEF) measured by left ventricular opacification (LVO) with contrast echocardiography and magnetic resonance imaging (MRI) at 12 months. The secondary endpoints include a composite of the incidence of death and re-hospitalization caused by heart failure at the 12th month, serum NT-proBNP, growth stimulation expressed gene 2 (ST2), and a change of right ventricular structure and function. Conclusions The PRIME-HFrEF study is designed to shed new light on multiple UC-MSC administration regimens for heart failure treatment.
Collapse
Affiliation(s)
- Xin Gong
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuheng Jiao
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Hao Hu
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Rongzhen Zhang
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wenwen Jia
- Institute for Regenerative Medicine, National Stem Cell Translational Resource Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhongmin Liu
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Yuanfeng Xin
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wei Han
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| |
Collapse
|
2
|
Shan Y, Zhang M, Tao E, Wang J, Wei N, Lu Y, Liu Q, Hao K, Zhou F, Wang G. Pharmacokinetic characteristics of mesenchymal stem cells in translational challenges. Signal Transduct Target Ther 2024; 9:242. [PMID: 39271680 PMCID: PMC11399464 DOI: 10.1038/s41392-024-01936-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/04/2024] [Accepted: 07/23/2024] [Indexed: 09/15/2024] Open
Abstract
Over the past two decades, mesenchymal stem/stromal cell (MSC) therapy has made substantial strides, transitioning from experimental clinical applications to commercial products. MSC therapies hold considerable promise for treating refractory and critical conditions such as acute graft-versus-host disease, amyotrophic lateral sclerosis, and acute respiratory distress syndrome. Despite recent successes in clinical and commercial applications, MSC therapy still faces challenges when used as a commercial product. Current detection methods have limitations, leaving the dynamic biodistribution, persistence in injured tissues, and ultimate fate of MSCs in patients unclear. Clarifying the relationship between the pharmacokinetic characteristics of MSCs and their therapeutic effects is crucial for patient stratification and the formulation of precise therapeutic regimens. Moreover, the development of advanced imaging and tracking technologies is essential to address these clinical challenges. This review provides a comprehensive analysis of the kinetic properties, key regulatory molecules, different fates, and detection methods relevant to MSCs and discusses concerns in evaluating MSC druggability from the perspective of integrating pharmacokinetics and efficacy. A better understanding of these challenges could improve MSC clinical efficacy and speed up the introduction of MSC therapy products to the market.
Collapse
Affiliation(s)
- Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Mengying Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Enxiang Tao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jing Wang
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Ning Wei
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Yi Lu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qing Liu
- Jiangsu Renocell Biotech Co. Ltd., Nanjing, China
| | - Kun Hao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
3
|
Mei R, Wan Z, Yang C, Shen X, Wang R, Zhang H, Yang R, Li J, Song Y, Su H. Advances and clinical challenges of mesenchymal stem cell therapy. Front Immunol 2024; 15:1421854. [PMID: 39100671 PMCID: PMC11294097 DOI: 10.3389/fimmu.2024.1421854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/02/2024] [Indexed: 08/06/2024] Open
Abstract
In recent years, cell therapy has provided desirable properties for promising new drugs. Mesenchymal stem cells are promising candidates for developing genetic engineering and drug delivery strategies due to their inherent properties, including immune regulation, homing ability and tumor tropism. The therapeutic potential of mesenchymal stem cells is being investigated for cancer therapy, inflammatory and fibrotic diseases, among others. Mesenchymal stem cells are attractive cellular carriers for synthetic nanoparticles for drug delivery due to their inherent homing ability. In this review, we comprehensively discuss the various genetic and non-genetic strategies of mesenchymal stem cells and their derivatives in drug delivery, tumor therapy, immune regulation, tissue regeneration and other fields. In addition, we discuss the current limitations of stem cell therapy and the challenges in clinical translation, aiming to identify important development areas and potential future directions.
Collapse
Affiliation(s)
- Ruiyan Mei
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Zhuo Wan
- Department of Hematology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Cheng Yang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Xiangjing Shen
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Ronglin Wang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Haihua Zhang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Rui Yang
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Junqiang Li
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Yang Song
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Haichuan Su
- Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
4
|
Gold DA, Sandesara PB, Kindya B, Gold ME, Jain V, Vatsa N, Desai SR, Yadalam A, Razavi A, Elhage Hassan M, Ko YA, Liu C, Alkhoder A, Rahbar A, Hossain MS, Waller EK, Jaber WA, Nicholson WJ, Quyyumi AA. Circulating Progenitor Cells and Coronary Collaterals in Chronic Total Occlusion. Int J Cardiol 2024; 407:132104. [PMID: 38677332 PMCID: PMC11559591 DOI: 10.1016/j.ijcard.2024.132104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND The role of circulating progenitor cells (CPC) in collateral formation that occurs in the presence of chronic total occlusions (CTO) of a coronary artery is not well established. In stable patients with a CTO, we investigated whether CPC levels are associated with (a) collateral development and (b) ischemic burden, as measured by circulating high sensitivity troponin-I (hsTn-I) levels. METHODS CPCs were enumerated by flow cytometry as CD45med+ blood mononuclear cells expressing CD34 and both CD34 and CD133 epitopes. The association between CPC counts and both Rentrop collateral grade (0, 1, 2, or 3) and hsTn-I levels were evaluated using multivariate regression analysis, after adjusting for demographic and clinical characteristics. RESULTS In 89 patients (age 65.5, 72% male, 27% Black), a higher CPC count was positively associated with a higher Rentrop collateral grade; [CD34+ adjusted odds ratio (OR) 1.49 95% confidence interval (CI) (0.95, 2.34) P = 0.082] and [CD34+/CD133+ OR 1.57 95% CI (1.05, 2.36) P = 0.028]. Every doubling of CPC counts was also associated with lower hsTn-I levels [CD34+ β -0.35 95% CI (-0.49, -0.15) P = 0.002] and [CD34+/CD133+ β -0.27 95% CI (-0.43, -0.08) P = 0.009] after adjustment. CONCLUSION Individuals with higher CPC counts have greater collateral development and lower ischemic burden in the presence of a CTO.
Collapse
Affiliation(s)
- Daniel A Gold
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Pratik B Sandesara
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Bryan Kindya
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Matthew E Gold
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Vardhmaan Jain
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Nishant Vatsa
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Shivang R Desai
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Adithya Yadalam
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Alexander Razavi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Malika Elhage Hassan
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Chang Liu
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Ayman Alkhoder
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Alireza Rahbar
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Mohammad S Hossain
- Division of Hematology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Edmund K Waller
- Division of Hematology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Wissam A Jaber
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - William J Nicholson
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
5
|
Matta A, Ohlmann P, Nader V, Moussallem N, Carrié D, Roncalli J. A review of therapeutic approaches for post-infarction left ventricular remodeling. Curr Probl Cardiol 2024; 49:102562. [PMID: 38599556 DOI: 10.1016/j.cpcardiol.2024.102562] [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: 03/31/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Left ventricular remodeling is an adaptive process initially developed in response to acute myocardial infarction (AMI), but it ends up with negative adverse outcomes such as infarcted wall thinning, ventricular dilation, and cardiac dysfunction. A prolonged excessive inflammatory reaction to cardiomyocytes death and necrosis plays the crucial role in the pathophysiological mechanisms. The pharmacological treatment includes nitroglycerine, β-blockers, ACEi/ARBs, SGLT2i, mineralocorticoid receptor antagonists, and some miscellaneous aspects. Stem cells therapy, CD34+ cells transplantation and gene therapy constitute the promissing therapeutic approaches for post AMI cardiac remodeling, thereby enhancing angiogenesis, cardiomyocytes differenciation and left ventricular function on top of inhibiting apoptosis, inflammation, and collagen deposition. All these lead to reduce infarct size, scar formation and myocardial fibrosis.
Collapse
Affiliation(s)
- Anthony Matta
- Department of Cardiology, Civilian Hospitals of Colmar, Colmar, France; School of Medicine and Medical Sciences, Holy Spirit University of Kaslik, P.O.Box 446, Jounieh, Lebanon.
| | - Patrick Ohlmann
- Department of Cardiology, Strasbourg University Hospital, Strasbourg, France
| | - Vanessa Nader
- Department of Cardiology, Civilian Hospitals of Colmar, Colmar, France
| | - Nicolas Moussallem
- School of Medicine and Medical Sciences, Holy Spirit University of Kaslik, P.O.Box 446, Jounieh, Lebanon
| | - Didier Carrié
- Department of Cardiology, Toulouse University Hospital, Toulouse, France
| | - Jerome Roncalli
- Department of Cardiology, Toulouse University Hospital, Toulouse, France
| |
Collapse
|
6
|
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
|
7
|
Qiu W, Sun Q, Li N, Chen Z, Wu H, Chen Z, Guo X, Fang F. Superoxide dismutase 2 scavenges ROS to promote osteogenic differentiation of human periodontal ligament stem cells by regulating Smad3 in alveolar bone-defective rats. J Periodontol 2024; 95:469-482. [PMID: 37921754 DOI: 10.1002/jper.23-0469] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs) is an essential event in alveolar bone regeneration. Oxidative stress may be the main inhibiting factor of hPDLSC osteogenesis. Superoxide dismutase 2 (SOD2) is a key antioxidant enzyme, but its effect on hPDLSC osteogenic differentiation is unclear. METHODS Several surface markers were detected by flow cytometry, and the differentiation potential of hPDLSCs was validated by alkaline phosphatase (ALP), Alizarin Red S, and Oil Red O staining. Osteogenic indicators of hPDLSCs were detected by real-time quantitative polymerase chain reaction (RT-qPCR), Western blotting, and ALP staining. Furthermore, alveolar bone defect rat models were analyzed through micro-CT, hematoxylin and eosin, and Masson staining. The intracellular reactive oxygen species (ROS) level was evaluated by a ROS assay kit. Finally, the expression of SOD2, Smad3, and p-Smad3 in hPDLSCs was detected by RT-qPCR and Western blotting (WB). RESULTS SOD2 positively regulated the gene and protein expressions of ALP, BMP6, and RUNX2 in hPDLSCs (p < 0.05). Ideal bone formation and continuous cortical bone were obtained by transplanting LV-SOD2 hPDLSCs (lentivirus vector for overexpressing SOD2 in hPDLSCs) in vivo. Exogenous H2O2 downregulated osteogenic indicators (ALP, BMP6, RUNX2) in hPDLSCs (p < 0.05); this was reversed by overexpression of SOD2. WB results showed that the Smad3 and p-Smad3 signaling pathways participated in the osteogenic process of SOD2 in hPDLSCs. CONCLUSION SOD2 positively regulated hPDLSC osteogenic differentiation in vitro and in vivo. Mechanistically, SOD2 promotes hPDLSC osteogenic differentiation by regulating the phosphorylation of Smad3 to scavenge ROS. This work provides a theoretical basis for the treatment of alveolar bone regeneration.
Collapse
Affiliation(s)
- Wei Qiu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Sun
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Na Li
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zehao Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongle Wu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Zhao Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaolan Guo
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fuchun Fang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Kavand A, Noverraz F, Gerber-Lemaire S. Recent Advances in Alginate-Based Hydrogels for Cell Transplantation Applications. Pharmaceutics 2024; 16:469. [PMID: 38675129 PMCID: PMC11053880 DOI: 10.3390/pharmaceutics16040469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
With its exceptional biocompatibility, alginate emerged as a highly promising biomaterial for a large range of applications in regenerative medicine. Whether in the form of microparticles, injectable hydrogels, rigid scaffolds, or bioinks, alginate provides a versatile platform for encapsulating cells and fostering an optimal environment to enhance cell viability. This review aims to highlight recent studies utilizing alginate in diverse formulations for cell transplantation, offering insights into its efficacy in treating various diseases and injuries within the field of regenerative medicine.
Collapse
Affiliation(s)
| | | | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; (A.K.); (F.N.)
| |
Collapse
|
9
|
Cheng HY, Anggelia MR, Liu SC, Lin CF, Lin CH. Enhancing Immunomodulatory Function of Mesenchymal Stromal Cells by Hydrogel Encapsulation. Cells 2024; 13:210. [PMID: 38334602 PMCID: PMC10854565 DOI: 10.3390/cells13030210] [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] [Received: 12/04/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) showcase remarkable immunoregulatory capabilities in vitro, positioning them as promising candidates for cellular therapeutics. However, the process of administering MSCs and the dynamic in vivo environment may impact the cell-cell and cell-matrix interactions of MSCs, consequently influencing their survival, engraftment, and their immunomodulatory efficacy. Addressing these concerns, hydrogel encapsulation emerges as a promising solution to enhance the therapeutic effectiveness of MSCs in vivo. Hydrogel, a highly flexible crosslinked hydrophilic polymer with a substantial water content, serves as a versatile platform for MSC encapsulation. Demonstrating improved engraftment and heightened immunomodulatory functions in vivo, MSCs encapsulated by hydrogel are at the forefront of advancing therapeutic outcomes. This review delves into current advancements in the field, with a focus on tuning various hydrogel parameters to elucidate mechanistic insights and elevate functional outcomes. Explored parameters encompass hydrogel composition, involving monomer type, functional modification, and co-encapsulation, along with biomechanical and physical properties like stiffness, viscoelasticity, topology, and porosity. The impact of these parameters on MSC behaviors and immunomodulatory functions is examined. Additionally, we discuss potential future research directions, aiming to kindle sustained interest in the exploration of hydrogel-encapsulated MSCs in the realm of immunomodulation.
Collapse
Affiliation(s)
- Hui-Yun Cheng
- Center for Vascularized Composite Allotransplantation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (M.R.A.)
| | - Madonna Rica Anggelia
- Center for Vascularized Composite Allotransplantation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (M.R.A.)
- Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Shiao-Chin Liu
- Center for Vascularized Composite Allotransplantation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (M.R.A.)
- Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chih-Fan Lin
- Center for Vascularized Composite Allotransplantation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (M.R.A.)
| | - Cheng-Hung Lin
- Center for Vascularized Composite Allotransplantation, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (M.R.A.)
- Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
10
|
Han L, Ma C, Wu Z, Xu H, Li H, Pan G. AhR-STAT3-HO-1/COX-2 signalling pathway may restrict ferroptosis and improve hMSC accumulation and efficacy in mouse liver. Br J Pharmacol 2024; 181:125-141. [PMID: 37538043 DOI: 10.1111/bph.16208] [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] [Received: 08/06/2022] [Revised: 06/26/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND AND PURPOSE The low efficacy of mesenchymal stem cells (MSCs) has restricted their application in the treatment of liver disease. Emerging evidence suggested that ferroptosis may provoke hepatocyte dysfunction and exacerbate damage to the liver microenvironment. Here, we have investigated the contribution of liver ferroptosis to the elimination and effectiveness of human MSC (hMSC). Furthermore, potential links between liver ferroptosis and aryl hydrocarbon receptors (AhR) were explored. EXPERIMENTAL APPROACH Two mouse models, iron supplement-induced hepatic ferroptosis and hepatic ischaemia/reperfusion (I/R) injury, were used to identify effects of ferroptosis on hMSC pharmacokinetics (PK)/pharmacodynamics (PD). KEY RESULTS AhR inhibition attenuated hepatic ferroptosis and improved survival of hMSCs. hMSC viability was decreased by iron supplementation or serum from I/R mice. The AhR antagonist CH223191 reversed iron overload and oxidative stress induced by ferroptosis and increased hMSC concentration and efficacy in mouse models. Effects of CH223191 were greater than those of deferoxamine, a conventional ferroptosis inhibitor. Transcriptomic results suggested that the AhR-signal transducer and activator of transcription 3 (STAT3)-haem oxygenase 1/COX-2 signalling pathway is critical to this process. These results were confirmed in a mouse model of hepatic I/R injury. In mice pre-treated with CH223191, hMSC exhibited more potent protective effects, linked to decreased hepatic ferroptosis. CONCLUSION AND IMPLICATIONS Our findings showed that ferroptosis was a critical factor in determining the fate of hMSCs. Inhibition of AhR decreased hepatic ferroptosis, thereby increasing survival and therapeutic effects of hMSCs in mouse models of liver disease.
Collapse
Affiliation(s)
- Li Han
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chenhui Ma
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Department of Chemical and Environmental Engineering, University of Nottingham Ningbo China, Ningbo, China
| | - Zhitao Wu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Huiming Xu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai Li
- Department of Gastroenterology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoyu Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
11
|
Bui TA, Stafford N, Oceandy D. Genetic and Pharmacological YAP Activation Induces Proliferation and Improves Survival in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Cells 2023; 12:2121. [PMID: 37681853 PMCID: PMC10487209 DOI: 10.3390/cells12172121] [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] [Received: 06/13/2023] [Revised: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
Cardiomyocyte loss following myocardial infarction cannot be addressed with current clinical therapies. Cell therapy with induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) is a potential approach to replace cardiomyocyte loss. However, engraftment rates in pre-clinical studies have been low, highlighting a need to refine current iPSC-CM technology. In this study, we demonstrated that inducing Yes-associated protein (YAP) by genetic and pharmacological approaches resulted in increased iPSC-CM proliferation and reduced apoptosis in response to oxidative stress. Interestingly, iPSC-CM maturation was differently affected by each strategy, with genetic activation of YAP resulting in a more immature cardiomyocyte-like phenotype not witnessed upon pharmacological YAP activation. Overall, we conclude that YAP activation in iPSC-CMs enhances cell survival and proliferative capacity. Therefore, strategies targeting YAP, or its upstream regulator the Hippo signalling pathway, could potentially be used to improve the efficacy of iPSC-CM technology for use as a future regenerative therapy in myocardial infarction.
Collapse
Affiliation(s)
| | | | - Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (T.A.B.); (N.S.)
| |
Collapse
|
12
|
Bui TQ, Binh NT, Pham TLB, Le Van T, Truong NH, Nguyen DPH, Luu TTT, Nguyen-Xuan Pham T, Cam Tran T, Nguyen HTT, Thuy-Trinh N, Tran PA. The Efficacy of Transplanting Human Umbilical Cord Mesenchymal Stem Cell Sheets in the Treatment of Myocardial Infarction in Mice. Biomedicines 2023; 11:2187. [PMID: 37626684 PMCID: PMC10452263 DOI: 10.3390/biomedicines11082187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
The transplantation of mesenchymal stem cell (MSC) sheets derived from human umbilical cords (hUCs) was investigated in this study as a potential application in treating myocardial infarction (MI). Two groups of hUC-MSC sheets were formed by populating LunaGelTM, which are 3D scaffolds of photo-crosslinkable gelatin-based hydrogel with two different cell densities. An MI model was created by ligating the left anterior descending coronary artery of healthy BALB/c mice. After two weeks, the cell sheets were applied directly to the MI area and the efficacy of the treatment was evaluated over the next two weeks by monitoring the mice's weight, evaluating the left ventricle ejection fraction, and assessing the histology of the heart tissue at the end of the experiment. Higher cell density showed significantly greater efficiency in MI mice treatment in terms of weight gain and the recovery of ejection fraction. The heart tissue of the groups receiving cell sheets showed human-CD44-positive staining and reduced fibrosis and apoptosis. In conclusion, the hUC-MSC sheets ameliorated heart MI injury in mice and the efficacy of the cell sheets improved as the number of cells increased.
Collapse
Affiliation(s)
| | - Nguyen Trong Binh
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (T.L.-B.P.); (D.P.-H.N.); (T.N.-X.P.)
| | - Truc Le-Buu Pham
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (T.L.-B.P.); (D.P.-H.N.); (T.N.-X.P.)
- Faculty of Biotechnology, Ho Chi Minh City Open University, Ho Chi Minh City 700000, Vietnam
| | - Trinh Le Van
- Laboratory of Stem Cell Research and Application, University of Science, Ho Chi Minh City 700000, Vietnam; (T.L.V.); (N.H.T.)
- Vietnam National University, Ho Chi Minh City 700000, Vietnam;
| | - Nhung Hai Truong
- Laboratory of Stem Cell Research and Application, University of Science, Ho Chi Minh City 700000, Vietnam; (T.L.V.); (N.H.T.)
- Vietnam National University, Ho Chi Minh City 700000, Vietnam;
| | - Dang Phu-Hai Nguyen
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (T.L.-B.P.); (D.P.-H.N.); (T.N.-X.P.)
| | - Thao Thi-Thu Luu
- Histology-Embryology-Pathology Department, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam;
| | - Trang Nguyen-Xuan Pham
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (T.L.-B.P.); (D.P.-H.N.); (T.N.-X.P.)
| | - Tu Cam Tran
- Institute of Tropical Biology, Ho Chi Minh City 700000, Vietnam;
| | - Huyen Thuong-Thi Nguyen
- Divison of Human and Animal Physiology, HCMC University of Education, Ho Chi Minh City 700000, Vietnam;
| | - Nhu Thuy-Trinh
- Vietnam National University, Ho Chi Minh City 700000, Vietnam;
- School of Biomedical Engineering, International University, Ho Chi Minh City 700000, Vietnam
| | - Phong Anh Tran
- Interface Science and Materials Engineering Group, School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia;
| |
Collapse
|
13
|
Collet BC, Davis DR. Mechanisms of Cardiac Repair in Cell Therapy. Heart Lung Circ 2023; 32:825-835. [PMID: 37031061 DOI: 10.1016/j.hlc.2023.01.019] [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: 07/29/2022] [Revised: 12/10/2022] [Accepted: 01/04/2023] [Indexed: 04/08/2023]
Abstract
Heart failure is an important cause of morbidity and mortality. More than 20 years ago, special interest was drawn to cell therapy as a means of restoring damaged hearts to working condition. But progress has not been straightforward as many of our initial assumptions turned out to be wrong. In this review, we critically examine the last 20 years of progress in cardiac cell therapy and focus on several of the popular beliefs surrounding cell therapy to illustrate the mechanisms involved in restoring heart function after cardiac injury. Are they true or false?
Collapse
Affiliation(s)
- Bérénice C Collet
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Darryl R Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
14
|
Kishino Y, Fukuda K. Unlocking the Pragmatic Potential of Regenerative Therapies in Heart Failure with Next-Generation Treatments. Biomedicines 2023; 11:biomedicines11030915. [PMID: 36979894 PMCID: PMC10046277 DOI: 10.3390/biomedicines11030915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Patients with chronic heart failure (HF) have a poor prognosis due to irreversible impairment of left ventricular function, with 5-year survival rates <60%. Despite advances in conventional medicines for HF, prognosis remains poor, and there is a need to improve treatment further. Cell-based therapies to restore the myocardium offer a pragmatic approach that provides hope for the treatment of HF. Although first-generation cell-based therapies using multipotent cells (bone marrow-derived mononuclear cells, mesenchymal stem cells, adipose-derived regenerative cells, and c-kit-positive cardiac cells) demonstrated safety in preclinical models of HF, poor engraftment rates, and a limited ability to form mature cardiomyocytes (CMs) and to couple electrically with existing CMs, meant that improvements in cardiac function in double-blind clinical trials were limited and largely attributable to paracrine effects. The next generation of stem cell therapies uses CMs derived from human embryonic stem cells or, increasingly, from human-induced pluripotent stem cells (hiPSCs). These cell therapies have shown the ability to engraft more successfully and improve electromechanical function of the heart in preclinical studies, including in non-human primates. Advances in cell culture and delivery techniques promise to further improve the engraftment and integration of hiPSC-derived CMs (hiPSC-CMs), while the use of metabolic selection to eliminate undifferentiated cells will help minimize the risk of teratomas. Clinical trials of allogeneic hiPSC-CMs in HF are now ongoing, providing hope for vast numbers of patients with few other options available.
Collapse
Affiliation(s)
| | - Keiichi Fukuda
- Correspondence: ; Tel.: +81-3-5363-3874; Fax: +81-3-5363-3875
| |
Collapse
|
15
|
Aggarwal R, Potel KN, Shao A, So SW, Swingen C, Reyes CP, Rose R, Wright C, Hocum Stone LL, McFalls EO, Butterick TA, Kelly RF. An Adjuvant Stem Cell Patch with Coronary Artery Bypass Graft Surgery Improves Diastolic Recovery in Porcine Hibernating Myocardium. Int J Mol Sci 2023; 24:ijms24065475. [PMID: 36982547 PMCID: PMC10049498 DOI: 10.3390/ijms24065475] [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: 01/13/2023] [Revised: 03/02/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023] Open
Abstract
Diastolic dysfunction persists despite coronary artery bypass graft surgery (CABG) in patients with hibernating myocardium (HIB). We studied whether the adjunctive use of a mesenchymal stem cells (MSCs) patch during CABG improves diastolic function by reducing inflammation and fibrosis. HIB was induced in juvenile swine by placing a constrictor on the left anterior descending (LAD) artery, causing myocardial ischemia without infarction. At 12 weeks, CABG was performed using the left-internal-mammary-artery (LIMA)-to-LAD graft with or without placement of an epicardial vicryl patch embedded with MSCs, followed by four weeks of recovery. The animals underwent cardiac magnetic resonance imaging (MRI) prior to sacrifice, and tissue from septal and LAD regions were collected to assess for fibrosis and analyze mitochondrial and nuclear isolates. During low-dose dobutamine infusion, diastolic function was significantly reduced in HIB compared to the control, with significant improvement after CABG + MSC treatment. In HIB, we observed increased inflammation and fibrosis without transmural scarring, along with decreased peroxisome proliferator-activated receptor-gamma coactivator (PGC1α), which could be a possible mechanism underlying diastolic dysfunction. Improvement in PGC1α and diastolic function was noted with revascularization and MSCs, along with decreased inflammatory signaling and fibrosis. These findings suggest that adjuvant cell-based therapy during CABG may recover diastolic function by reducing oxidant stress–inflammatory signaling and myofibroblast presence in the myocardial tissue.
Collapse
Affiliation(s)
- Rishav Aggarwal
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (R.A.); (A.S.); (C.S.); (C.P.R.); (R.R.); (C.W.); (L.L.H.S.)
| | - Koray N. Potel
- School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK;
| | - Annie Shao
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (R.A.); (A.S.); (C.S.); (C.P.R.); (R.R.); (C.W.); (L.L.H.S.)
| | - Simon W. So
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (S.W.S.); (T.A.B.)
- Department of Research, Center for Veterans Research and Education, Minneapolis, MN 55417, USA
| | - Cory Swingen
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (R.A.); (A.S.); (C.S.); (C.P.R.); (R.R.); (C.W.); (L.L.H.S.)
| | - Christina P. Reyes
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (R.A.); (A.S.); (C.S.); (C.P.R.); (R.R.); (C.W.); (L.L.H.S.)
| | - Rebecca Rose
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (R.A.); (A.S.); (C.S.); (C.P.R.); (R.R.); (C.W.); (L.L.H.S.)
| | - Christin Wright
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (R.A.); (A.S.); (C.S.); (C.P.R.); (R.R.); (C.W.); (L.L.H.S.)
| | - Laura L. Hocum Stone
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (R.A.); (A.S.); (C.S.); (C.P.R.); (R.R.); (C.W.); (L.L.H.S.)
| | - Edward O. McFalls
- Division of Cardiology, Richmond VA Medical Center, Richmond, VA 23249, USA;
| | - Tammy A. Butterick
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (S.W.S.); (T.A.B.)
- Department of Research, Center for Veterans Research and Education, Minneapolis, MN 55417, USA
| | - Rosemary F. Kelly
- Division of Cardiothoracic Surgery, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (R.A.); (A.S.); (C.S.); (C.P.R.); (R.R.); (C.W.); (L.L.H.S.)
- Correspondence: ; Tel.: +1-612-625-3902
| |
Collapse
|
16
|
Thummarati P, Laiwattanapaisal W, Nitta R, Fukuda M, Hassametto A, Kino-oka M. Recent Advances in Cell Sheet Engineering: From Fabrication to Clinical Translation. Bioengineering (Basel) 2023; 10:211. [PMID: 36829705 PMCID: PMC9952256 DOI: 10.3390/bioengineering10020211] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/26/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Cell sheet engineering, a scaffold-free tissue fabrication technique, has proven to be an important breakthrough technology in regenerative medicine. Over the past two decades, the field has developed rapidly in terms of investigating fabrication techniques and multipurpose applications in regenerative medicine and biological research. This review highlights the most important achievements in cell sheet engineering to date. We first discuss cell sheet harvesting systems, which have been introduced in temperature-responsive surfaces and other systems to overcome the limitations of conventional cell harvesting methods. In addition, we describe several techniques of cell sheet transfer for preclinical (in vitro and in vivo) and clinical trials. This review also covers cell sheet cryopreservation, which allows short- and long-term storage of cells. Subsequently, we discuss the cell sheet properties of angiogenic cytokines and vasculogenesis. Finally, we discuss updates to various applications, from biological research to clinical translation. We believe that the present review, which shows and compares fundamental technologies and recent advances in cell engineering, can potentially be helpful for new and experienced researchers to promote the further development of tissue engineering in different applications.
Collapse
Affiliation(s)
- Parichut Thummarati
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
- Biosensors and Bioanalytical Technology for Cells and Innovative Testing Device Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wanida Laiwattanapaisal
- Biosensors and Bioanalytical Technology for Cells and Innovative Testing Device Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Rikiya Nitta
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Megumi Fukuda
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Artchaya Hassametto
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Masahiro Kino-oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
17
|
Zuo X, Jiang X, Zhang Y, Huang Y, Wang N, Zhu P, Kang YJ. A clinical feasible stem cell encapsulation ensures an improved wound healing. Biomed Mater 2023; 18. [PMID: 36701809 DOI: 10.1088/1748-605x/acb67a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/26/2023] [Indexed: 01/28/2023]
Abstract
Cell encapsulation has proven to be promising in stem cell therapy. However, there are issues needed to be addressed, including unsatisfied yield, unmet clinically friendly formulation, and unacceptable viability of stem cells after cryopreservation and thawing. We developed a novel biosynsphere technology to encapsulate stem cells in clinically-ready biomaterials with controlled microsphere size. We demonstrated that biosynspheres ensure the bioviability and functionality of adipose-derived stromal cells (ADSCs) encapsulated, as delineated by a series of testing procedures. We further demonstrated that biosynspheres protect ADSCs from the hardness of clinically handling such as cryopreservation, thawing, high-speed centrifugation and syringe/nozzle injection. In a swine full skin defect model, we showed that biosynspheres were integrated to the destined tissues and promoted the repair of injured tissues with an accelerating healing process, less scar tissue formation and normalized deposition of collagen type I and type III, the ratio similar to that found in normal skin. These findings underscore the potential of biosynsphere as an improved biofabrication technology for tissue regeneration in clinical setting.
Collapse
Affiliation(s)
- Xiao Zuo
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, People's Republic of China.,Sichuan 3D Bio-Printing Institute, Chengdu, Sichuan 611731, People's Republic of China.,Revotek Co., Ltd, Chengdu, Sichuan 611731, People's Republic of China
| | - Xia Jiang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, People's Republic of China
| | - Yaya Zhang
- Sichuan 3D Bio-Printing Institute, Chengdu, Sichuan 611731, People's Republic of China.,Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, United States of America
| | - Yushi Huang
- Revotek Co., Ltd, Chengdu, Sichuan 611731, People's Republic of China
| | - Ning Wang
- Revotek Co., Ltd, Chengdu, Sichuan 611731, People's Republic of China
| | - Ping Zhu
- Revotek Co., Ltd, Chengdu, Sichuan 611731, People's Republic of China
| | - Y James Kang
- Regenerative Medicine Research Center, Sichuan University West China Hospital, Chengdu, Sichuan 610041, People's Republic of China.,Sichuan 3D Bio-Printing Institute, Chengdu, Sichuan 611731, People's Republic of China.,Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, United States of America
| |
Collapse
|
18
|
Ding Z, Tan K, Alter C, Temme S, Bouvain P, Owenier C, Hänsch S, Wesselborg S, Peter C, Weidtkamp-Peters S, Flögel U, Schira-Heinen J, Stühler K, Hesse J, Kögler G, Schrader J. Cardiac injection of USSC boosts remuscularization of the infarcted heart by shaping the T-cell response. J Mol Cell Cardiol 2023; 175:29-43. [PMID: 36493853 DOI: 10.1016/j.yjmcc.2022.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/20/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022]
Abstract
Regenerating the injured heart remains one of the most vexing challenges in cardiovascular medicine. Cell therapy has shown potential for treatment of myocardial infarction, but low cell retention so far has limited its success. Here we show that intramyocardial injection of highly apoptosis-resistant unrestricted somatic stem cells (USSC) into infarcted rat hearts resulted in an unprecedented thickening of the left ventricular wall with cTnT+/BrdU+ cardiomyocytes that was paralleled by progressively restored ejection fraction. USSC induced significant T-cell enrichment in ischemic tissue with enhanced expression of T-cell related cytokines. Inhibition of T-cell activation by anti-CD28 monoclonal antibody, fully abolished the regenerative response which was restored by adoptive T-cell transfer. Secretome analysis of USSC and lineage tracing studies suggest that USSC secrete paracrine factors over an extended period of time which boosts a T-cell driven endogenous regenerative response mainly from adult cardiomyocytes.
Collapse
Affiliation(s)
- Zhaoping Ding
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Kezhe Tan
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Christina Alter
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Sebastian Temme
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Pascal Bouvain
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Christoph Owenier
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Sebastian Hänsch
- Center for Advanced Imaging, Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Sebastian Wesselborg
- Institute of Molecular Medicine I, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Christoph Peter
- Institute of Molecular Medicine I, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | | | - Ulrich Flögel
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Jessica Schira-Heinen
- Molecular Proteomics Laboratory (MPL), Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory (MPL), Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Julia Hesse
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Gesine Kögler
- Jose Carreras Stem Cell Bank, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany
| | - Jürgen Schrader
- Department of Molecular Cardiology, Heinrich Heine University of Duesseldorf, Duesseldorf, Germany.
| |
Collapse
|
19
|
Pruller J, Pham TT, Blower JE, Charoenphun P, Volpe A, Sunassee K, Mullen GED, Blower PJ, Smith RAG, Ma MT. An indium-111-labelled membrane-targeted peptide for cell tracking with radionuclide imaging. RSC Chem Biol 2023; 4:65-73. [PMID: 36685254 PMCID: PMC9811519 DOI: 10.1039/d2cb00164k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022] Open
Abstract
Cell labelling agents that enable longitudinal in vivo tracking of administered cells will support the clinical development of cell-based therapies. Radionuclide imaging with gamma and positron-emitting radioisotopes can provide quantitative and longitudinal mapping of cells in vivo. To make this widely accessible and adaptable to a range of cell types, new, versatile and simple methods for directly radiolabelling cells are required. We have developed [111In]In-DTPA-CTP, the first example of a radiolabelled peptide that binds to the extracellular membrane of cells, for tracking cell distribution in vivo using Single Photon Emission Computed Tomography (SPECT). [111In]In-DTPA-CTP consists of (i) myristoyl groups for insertion into the phospholipid bilayer, (ii) positively charged lysine residues for electrostatic association with negatively charged phospholipid groups at the cell surface and (iii) a diethylenetriamine pentaacetate derivative that coordinates the γ-emitting radiometal, [111In]In3+. [111In]In-DTPA-CTP binds to 5T33 murine myeloma cells, enabling qualitative SPECT tracking of myeloma cells' accumulation in lungs immediately after intravenous administration. This is the first report of a radiolabelled cell-membrane binding peptide for use in cell tracking.
Collapse
Affiliation(s)
- Johanna Pruller
- Randall Division of Cell and Molecular Biophysics, King's College London UK
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Truc Thuy Pham
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Julia E Blower
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Putthiporn Charoenphun
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
- Department of Diagnostic and Therapeutic Radiology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University Bangkok Thailand
| | - Alessia Volpe
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Kavitha Sunassee
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Gregory E D Mullen
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Philip J Blower
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| | - Richard A G Smith
- MRC Centre for Transplantation, King's College London, Guy's Hospital London UK
| | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital London UK
| |
Collapse
|
20
|
Chauhan A, Alam MA, Kaur A, Malviya R. Advancements and Utilizations of Scaffolds in Tissue Engineering and Drug Delivery. Curr Drug Targets 2023; 24:13-40. [PMID: 36221880 DOI: 10.2174/1389450123666221011100235] [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: 01/05/2022] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 11/22/2022]
Abstract
The drug development process requires a thorough understanding of the scaffold and its three-dimensional structure. Scaffolding is a technique for tissue engineering and the formation of contemporary functioning tissues. Tissue engineering is sometimes referred to as regenerative medicine. They also ensure that drugs are delivered with precision. Information regarding scaffolding techniques, scaffolding kinds, and other relevant facts, such as 3D nanostructuring, are discussed in depth in this literature. They are specific and demonstrate localized action for a specific reason. Scaffold's acquisition nature and flexibility make it a new drug delivery technology with good availability and structural parameter management.
Collapse
Affiliation(s)
- Akash Chauhan
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Md Aftab Alam
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Awaneet Kaur
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
21
|
Garay RP. Recent clinical trials with stem cells to slow or reverse normal aging processes. FRONTIERS IN AGING 2023; 4:1148926. [PMID: 37090485 PMCID: PMC10116573 DOI: 10.3389/fragi.2023.1148926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/24/2023] [Indexed: 04/25/2023]
Abstract
Aging is associated with a decline in the regenerative potential of stem cells. In recent years, several clinical trials have been launched in order to evaluate the efficacy of mesenchymal stem cell interventions to slow or reverse normal aging processes (aging conditions). Information concerning those clinical trials was extracted from national and international databases (United States, EU, China, Japan, and World Health Organization). Mesenchymal stem cell preparations were in development for two main aging conditions: physical frailty and facial skin aging. With regard to physical frailty, positive results have been obtained in phase II studies with intravenous Lomecel-B (an allogeneic bone marrow stem cell preparation), and a phase I/II study with an allogeneic preparation of umbilical cord-derived stem cells was recently completed. With regard to facial skin aging, positive results have been obtained with an autologous preparation of adipose-derived stem cells. A further sixteen clinical trials for physical frailty and facial skin aging are currently underway. Reducing physical frailty with intravenous mesenchymal stem cell administration can increase healthy life expectancy and decrease costs to the public health system. However, intravenous administration runs the risk of entrapment of the stem cells in the lungs (and could raise safety concerns). In addition to aesthetic purposes, clinical research on facial skin aging allows direct evaluation of tissue regeneration using sophisticated and precise methods. Therefore, research on both conditions is complementary, which facilitates a global vision.
Collapse
Affiliation(s)
- Ricardo P. Garay
- Pharmacology and Therapeutics, Craven, 91360 Villemoisson-sur-Orge, France
- CNRS, National Centre of Scientific Research, Paris, France
- *Correspondence: Ricardo P. Garay,
| |
Collapse
|
22
|
Hassanpour M, Salybekov AA, Kobayashi S, Asahara T. CD34 positive cells as endothelial progenitor cells in biology and medicine. Front Cell Dev Biol 2023; 11:1128134. [PMID: 37138792 PMCID: PMC10150654 DOI: 10.3389/fcell.2023.1128134] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/03/2023] [Indexed: 05/05/2023] Open
Abstract
CD34 is a cell surface antigen expressed in numerous stem/progenitor cells including hematopoietic stem cells (HSCs) and endothelial progenitor cells (EPCs), which are known to be rich sources of EPCs. Therefore, regenerative therapy using CD34+ cells has attracted interest for application in patients with various vascular, ischemic, and inflammatory diseases. CD34+ cells have recently been reported to improve therapeutic angiogenesis in a variety of diseases. Mechanistically, CD34+ cells are involved in both direct incorporation into the expanding vasculature and paracrine activity through angiogenesis, anti-inflammatory, immunomodulatory, and anti-apoptosis/fibrosis roles, which support the developing microvasculature. Preclinical, pilot, and clinical trials have well documented a track record of safety, practicality, and validity of CD34+ cell therapy in various diseases. However, the clinical application of CD34+ cell therapy has triggered scientific debates and controversies in last decade. This review covers all preexisting scientific literature and prepares an overview of the comprehensive biology of CD34+ cells as well as the preclinical/clinical details of CD34+ cell therapy for regenerative medicine.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell Therapy and Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Amankeldi A. Salybekov
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell Therapy and Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Shuzo Kobayashi
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
| | - Takayuki Asahara
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- Center for Cell Therapy and Regenerative Medicine, Shonan Kamakura General Hospital, Kamakura, Kanagawa, Japan
- *Correspondence: Takayuki Asahara,
| |
Collapse
|
23
|
Kojima H, Kushige H, Yagi H, Nishijima T, Moritoki N, Nagoshi N, Nakano Y, Tanaka M, Hori S, Hasegawa Y, Abe Y, Kitago M, Nakamura M, Kitagawa Y. Combinational Treatment Involving Decellularized Extracellular Matrix Hydrogels With Mesenchymal Stem Cells Increased the Efficacy of Cell Therapy in Pancreatitis. Cell Transplant 2023; 32:9636897231170437. [PMID: 37191199 PMCID: PMC10192953 DOI: 10.1177/09636897231170437] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 05/17/2023] Open
Abstract
Cell transplantation using mesenchymal stem cells (MSCs) has emerged as a promising approach to repairing and regenerating injured or impaired organs. However, the survival and retention of MSCs following transplantation remain a challenge. Therefore, we investigated the efficacy of co-transplantation of MSCs and decellularized extracellular matrix (dECM) hydrogels, which have high cytocompatibility and biocompatibility. The dECM solution was prepared by enzymatic digestion of an acellular porcine liver scaffold. It could be gelled and formed into porous fibrillar microstructures at physiological temperatures. MSCs expanded three-dimensionally in the hydrogel without cell death. Compared to the 2-dimensional cell culture, MSCs cultured in the hydrogel showed increased secretion of hepatocyte growth factor (HGF) and tumor necrosis factor-inducible gene 6 protein (TSG-6), both of which are major anti-inflammatory and anti-fibrotic paracrine factors of MSCs, under TNFα stimulation. In vivo experiments showed that the co-transplantation of MSCs with dECM hydrogel improved the survival rate of engrafted cells compared to those administered without the hydrogel. MSCs also demonstrated therapeutic effects in improving inflammation and fibrosis of pancreatic tissue in a dibutyltin dichloride (DBTC)-induced rat pancreatitis model. Combinational use of dECM hydrogel with MSCs is a new strategy to overcome the challenges of cell therapy using MSCs and can be used for treating chronic inflammatory diseases in clinical settings.
Collapse
Affiliation(s)
- Hideaki Kojima
- Department of Surgery, Keio University
School of Medicine, Tokyo, Japan
| | - Hiroko Kushige
- Department of Surgery, Keio University
School of Medicine, Tokyo, Japan
| | - Hiroshi Yagi
- Department of Surgery, Keio University
School of Medicine, Tokyo, Japan
| | - Takayuki Nishijima
- Department of Orthopaedic Surgery, Keio
University School of Medicine, Tokyo, Japan
| | - Nobuko Moritoki
- Electron Microscope Laboratory, Keio
University School of Medicine, Tokyo, Japan
| | - Narihito Nagoshi
- Department of Orthopaedic Surgery, Keio
University School of Medicine, Tokyo, Japan
| | - Yutaka Nakano
- Department of Surgery, Keio University
School of Medicine, Tokyo, Japan
| | - Masayuki Tanaka
- Department of Surgery, Keio University
School of Medicine, Tokyo, Japan
| | - Shutaro Hori
- Department of Surgery, Keio University
School of Medicine, Tokyo, Japan
| | - Yasushi Hasegawa
- Department of Surgery, Keio University
School of Medicine, Tokyo, Japan
| | - Yuta Abe
- Department of Surgery, Keio University
School of Medicine, Tokyo, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University
School of Medicine, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio
University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University
School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Fujita Y, Kawamoto A. Therapeutic Angiogenesis Using Autologous CD34-Positive Cells for Vascular Diseases. Ann Vasc Dis 2022; 15:241-252. [PMID: 36644256 PMCID: PMC9816028 DOI: 10.3400/avd.ra.22-00086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/13/2022] [Indexed: 12/25/2022] Open
Abstract
CD34 is a cell surface marker, which is expressed in various somatic stem/progenitor cells such as bone marrow (BM)-derived hematopoietic stem cells and endothelial progenitor cells (EPCs), skeletal muscle satellite cells, epithelial hair follicle stem cells, and adipose tissue mesenchymal stem cells. CD34+ cells in BM and peripheral blood are known as a rich source of EPCs. Thus, vascular regeneration therapy using granulocyte colony stimulating factor (G-CSF) mobilized- or BM CD34+ cells has been carried out in patients with various vascular diseases such as chronic severe lower limb ischemia, acute myocardial infarction, refractory angina, ischemic cardiomyopathy, and dilated cardiomyopathy as well as ischemic stroke. Pilot and randomized clinical trials demonstrated the safety, feasibility, and effectiveness of the CD34+ cell therapy in peripheral arterial, cardiovascular, and cerebrovascular diseases. This review provides an overview of the preclinical and clinical reports of CD34+ cell therapy for vascular regeneration.
Collapse
Affiliation(s)
- Yasuyuki Fujita
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Hyogo, Japan
| | - Atsuhiko Kawamoto
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Hyogo, Japan,Corresponding author: Atsuhiko Kawamoto, MD, PhD. Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, 1-5-4 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan Tel: +81-78-304-5772, Fax: +81-78-304-5263, E-mail:
| |
Collapse
|
25
|
Höving AL, Schmidt KE, Kaltschmidt B, Kaltschmidt C, Knabbe C. The Role of Blood-Derived Factors in Protection and Regeneration of Aged Tissues. Int J Mol Sci 2022; 23:ijms23179626. [PMID: 36077021 PMCID: PMC9455681 DOI: 10.3390/ijms23179626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 12/02/2022] Open
Abstract
Tissue regeneration substantially relies on the functionality of tissue-resident endogenous adult stem cell populations. However, during aging, a progressive decline in organ function and regenerative capacities impedes endogenous repair processes. Especially the adult human heart is considered as an organ with generally low regenerative capacities. Interestingly, beneficial effects of systemic factors carried by young blood have been described in diverse organs including the heart, brain and skeletal muscle of the murine system. Thus, the interest in young blood or blood components as potential therapeutic agents to target age-associated malignancies led to a wide range of preclinical and clinical research. However, the translation of promising results from the murine to the human system remains difficult. Likewise, the establishment of adequate cellular models could help to study the effects of human blood plasma on the regeneration of human tissues and particularly the heart. Facing this challenge, this review describes the current knowledge of blood plasma-mediated protection and regeneration of aging tissues. The current status of preclinical and clinical research examining blood borne factors that act in stem cell-based tissue maintenance and regeneration is summarized. Further, examples of cellular model systems for a more detailed examination of selected regulatory pathways are presented.
Collapse
Affiliation(s)
- Anna L. Höving
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
- Correspondence:
| | - Kazuko E. Schmidt
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Barbara Kaltschmidt
- AG Molecular Neurobiology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Cornelius Knabbe
- Heart and Diabetes Centre NRW, Institute for Laboratory and Transfusion Medicine, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| |
Collapse
|
26
|
CXCR4+ Sorted Adipose-Derived Stem Cells Enhance Their Functional Benefits and Improve Cardiac Function after Myocardial Infarction. Stem Cells Int 2022; 2022:6714765. [PMID: 36051532 PMCID: PMC9427246 DOI: 10.1155/2022/6714765] [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] [Received: 11/15/2021] [Revised: 07/25/2022] [Accepted: 08/08/2022] [Indexed: 11/18/2022] Open
Abstract
Objective The homing of adipose-derived stem cells (ASCs) to infarcted myocardium, which is important for improved cardiac function, has been investigated previously, but with poor efficiency. Substantial improvements in engraftments are required to optimize ASC treatment. Stromal derived factor-1α (SDF-1α) is upregulated early after MI, and its endogenous receptor, chemokine receptor 4 (CXCR4), is pivotal in stem cell survival, migration, and engraftment. We examined whether CXCR4+ ASCs enhance their efficacy of migration and engraftment posttransplantation and improve heart function following myocardial infarction (MI). Methods and Results CXCR4+ ASC subpopulations were sorted by fluorescence-activated cell sorting. CXCR4+ sorted ASCs exhibited the stronger cell viability, the faster proliferation rate, and the better migration capability in comparison with unfractionated ASCs. CXCR4+ sorted ASCs secreted a higher level of angiogenic growth factors including VEGF, HGF, and IGF-1 relative to unfractionated ASCs. Fewer apoptotic cells under oxygen-glucose deprivation were detected in CXCR4+ sorted ASCs than in unfractionated ASCs. Osteogenic and angiogenic differentiation were more pronounced in CXCR4+ sorted ASCs than in unfractionated ASCs. At 3 days after acute MI, rats were randomly allocated to receive intramyocardial injection of cell culture medium, CXCR4+ sorted ASCs, and unfractionated ASCs. Left ventricular function was assessed echocardiographically 4 weeks thereafter. Explanted hearts were then processed for the immunofluorescence detection of survived cells, quantification of angiogenesis, and cell engraftment. CXCR4+ sorted ASCs more obviously engrafted into infarcted myocardium, more markedly inhibited collagen remodeling, and more effectively improved heart function and promoted capillary formation than did unfractionated ASCs. Conclusion CXCR4+ sorted ASCs are superior to unfractionated ASCs due to better viability, faster proliferation, more cytokine secretion, and stronger migration. CXCR4+ sorted ASCs provide better curative benefits on MI than do unfractionated ASCs and can be efficiently harvested and purified from adipose tissue, they may serve as a promising candidate for MI.
Collapse
|
27
|
Patrick MD, Annamalai RT. Licensing microgels prolong the immunomodulatory phenotype of mesenchymal stromal cells. Front Immunol 2022; 13:987032. [PMID: 36059508 PMCID: PMC9433901 DOI: 10.3389/fimmu.2022.987032] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stromal cells (MSC) are sensors of inflammation, and they exert immunomodulatory properties through the secretion of cytokines and exosomes and direct cell-cell interactions. MSC are routinely used in clinical trials and effectively resolve inflammatory conditions. Nevertheless, inconsistent clinical outcomes necessitate the need for more robust therapeutic phenotypes. The immunomodulatory properties of MSC can be enhanced and protracted by priming (aka licensing) them with IFNγ and TNFα. Yet these enhanced properties rapidly diminish, and prolonged stimulation could tolerize their response. Hence a balanced approach is needed to enhance the therapeutic potential of the MSC for consistent clinical performance. Here, we investigated the concentration-dependent effects of IFNγ and TNFα and developed gelatin-based microgels to sustain a licensed MSC phenotype. We show that IFNγ treatment is more beneficial than TNFα in promoting an immunomodulatory MSC phenotype. We also show that the microgels possess integrin-binding sites to support adipose tissue-derived MSC (AD-MSC) attachment and a net positive charge to sequester the licensing cytokines electrostatically. Microgels are enzymatically degradable, and the rate is dependent on the enzyme concentration and matrix density. Our studies show that one milligram of microgels by dry mass can sequester up to 641 ± 81 ng of IFNγ. Upon enzymatic degradation, microgels exhibited a sustained release of IFNγ that linearly correlated with their degradation rate. The AD-MSC cultured on the IFNγ sequestered microgels displayed efficient licensing potential comparable to or exceeding the effects of bolus IFNγ treatment. When cultured with proinflammatory M1-like macrophages, the AD-MSC-seeded on licensing microgel showed an enhanced immunomodulatory potential compared to untreated AD-MSC and AD-MSC treated with bolus IFNγ treatment. Specifically, the AD-MSC seeded on licensing microgels significantly upregulated Arg1, Mrc1, and Igf1, and downregulated Tnfα in M1-like macrophages compared to other treatment conditions. These licensing microgels are a potent immunomodulatory approach that shows substantial promise in elevating the efficacy of current MSC therapies and may find utility in treating chronic inflammatory conditions.
Collapse
|
28
|
Mao Q, Shao W, Lv S, Tong P, He B. Case report: Directional infusion of peripheral blood stem cells into the necrotic zone in femoral heads through the medial circumflex femoral artery: A tracing study. Front Med (Lausanne) 2022; 9:945268. [PMID: 36059815 PMCID: PMC9433798 DOI: 10.3389/fmed.2022.945268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Objective This study aimed to explore whether peripheral blood stem cells (PBSCs) infused through the medial circumflex femoral artery to treat osteonecrosis of the femoral head (ONFH) could migrate into the necrotic area of femoral head. Methods We collected PBSCs from a patient who had bilateral ONFH by apheresis technique using COBE spectra apheresis system (COBE BCT Inc, Lakewood, CO, USA) after subcutaneous injections of granulocyte-colony stimulating factor (G-CSF) at a dosage of 10 μg/kg for 4 days to mobilize PBSCs. After that, 100 MBq 2-[18F]-fluoro-2-deoxy-D-glucose (18F-FDG) was used to label PBSCs. 18F-FDG labeled PBSCs were infused into the left femoral head via the medial circumflex femoral artery to treat ONFH. Then the patient was underwent three-dimensional positron emission tomography (3D-PET) examination 60 min after cell infusion to monitor the biological distribution of 18F-FDG-labeled PBSCs, and to observe whether the transplanted PBSCs could migrate into the necrotic area of femoral head. Results The total number of monouclear cells in the peripheral blood stem cell suspension was 1.95 × 108 which contained 2.20 × 106 CD34+ cells. The activity of 18F-FDG in the labeled cells was 1.8Bq/103 monouclear cells. 3D-PET imaging showed that 18F-FDG radioactivity was detected in the necrotic area of femoral head, acetabulum and femoral bone marrow cavity after transplantation of 18F-FDG-labeled PBSCs via the medial circumflex femoral artery. It is worth noting that although PBSCs labeled with 18F-FDG were widely distributed around the hip, such as femoral bone marrow cavity, femoral head and acetabulum, PBSCs were generally located in the necrotic area of femoral head. Conclusions PBSCs could enter into the femoral head and migrate into the necrotic field of femoral head participating in the repair of osteonecrosis after infusion through the medial circumflex femoral artery.
Collapse
Affiliation(s)
- Qiang Mao
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Weijie Shao
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuaijie Lv
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Peijian Tong
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Bangjian He
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Bangjian He
| |
Collapse
|
29
|
Liu J, Liang X, Li M, Lin F, Ma X, Xin Y, Meng Q, Zhuang R, Zhang Q, Han W, Gao L, He Z, Zhou X, Liu Z. Intramyocardial injected human umbilical cord-derived mesenchymal stem cells (HucMSCs) contribute to the recovery of cardiac function and the migration of CD4 + T cells into the infarcted heart via CCL5/CCR5 signaling. Stem Cell Res Ther 2022; 13:247. [PMID: 35690805 PMCID: PMC9188247 DOI: 10.1186/s13287-022-02914-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 05/25/2022] [Indexed: 12/14/2022] Open
Abstract
Background Human umbilical cord-derived mesenchymal stem cells (HucMSCs) have been recognized as a promising cell for treating myocardial infarction (MI). Inflammatory response post MI is critical in determining the cardiac function and subsequent adverse left ventricular remodeling. However, the local inflammatory effect of HucMSCs after intramyocardial injection in murine remains unclear. Methods HucMSCs were cultured and transplanted into the mice after MI surgery. Cardiac function of mice were analyzed among MI-N.S, MI-HucMSC and MI-HucMSC-C–C Motif Chemokine receptor 5 (CCR5) antagonist groups, and angiogenesis, fibrosis and hypertrophy, and immune cells infiltration of murine hearts were evaluated between MI-N.S and MI-HucMSC groups. We detected the expression of inflammatory cytokines and their effects on CD4+ T cells migration. Results HucMSCs treatment can significantly improve the cardiac function and some cells can survive at least 28 days after MI. Intramyocardial administration of HucMSCs also improved angiogenesis and alleviated cardiac fibrosis and hypertrophy. Moreover, we found the much higher numbers of CD4+ T cells and CD4+FoxP3+ regulatory T cells (Tregs) in the heart with HucMSCs than that with N.S treatment on day 7 post MI. In addition, the protein level of C–C Motif Chemokine Ligand 5 (CCL5) greatly increased in HucMSCs treated heart compared to MI-N.S group. In vitro, HucMSCs inhibited CD4+ T cells migration and addition of CCL5 antibody or CCR5 antagonist significantly reversed this effect. In vivo results further showed that addition of CCR5 antagonist can reduce the cardioprotective effect of HucMSCs administration on day 7 post MI injury. Conclusion These findings indicated that HucMSCs contributed to cardiac functional recovery and attenuated cardiac remodeling post MI. Intramyocardial injection of HucMSCs upregulated the CD4+FoxP3+ Tregs and contributed to the migration of CD4+ T cells into the injured heart via CCL5/CCR5 pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02914-z.
Collapse
Affiliation(s)
- Jing Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China.,Department of Burn and Plastic Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, People's Republic of China
| | - Xiaoting Liang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China.,Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, People's Republic of China
| | - Mimi Li
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China.,Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China
| | - Fang Lin
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China.,Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China
| | - Xiaoxue Ma
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China.,Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China
| | - Yuanfeng Xin
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China.,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China
| | - Qingshu Meng
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China.,Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China
| | - Rulin Zhuang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China.,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China
| | - Qingliu Zhang
- Department of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, People's Republic of China
| | - Wei Han
- Department of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, People's Republic of China
| | - Ling Gao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, People's Republic of China
| | - Zhiying He
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, People's Republic of China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, People's Republic of China.,Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, 200335, People's Republic of China
| | - Xiaohui Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China. .,Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China.
| | - Zhongmin Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China. .,Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, People's Republic of China. .,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Rd, Pudong, Shanghai, 200120, People's Republic of China. .,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, People's Republic of China.
| |
Collapse
|
30
|
Gawne P, Man F, Blower PJ, T. M. de Rosales R. Direct Cell Radiolabeling for in Vivo Cell Tracking with PET and SPECT Imaging. Chem Rev 2022; 122:10266-10318. [PMID: 35549242 PMCID: PMC9185691 DOI: 10.1021/acs.chemrev.1c00767] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Indexed: 02/07/2023]
Abstract
The arrival of cell-based therapies is a revolution in medicine. However, its safe clinical application in a rational manner depends on reliable, clinically applicable methods for determining the fate and trafficking of therapeutic cells in vivo using medical imaging techniques─known as in vivo cell tracking. Radionuclide imaging using single photon emission computed tomography (SPECT) or positron emission tomography (PET) has several advantages over other imaging modalities for cell tracking because of its high sensitivity (requiring low amounts of probe per cell for imaging) and whole-body quantitative imaging capability using clinically available scanners. For cell tracking with radionuclides, ex vivo direct cell radiolabeling, that is, radiolabeling cells before their administration, is the simplest and most robust method, allowing labeling of any cell type without the need for genetic modification. This Review covers the development and application of direct cell radiolabeling probes utilizing a variety of chemical approaches: organic and inorganic/coordination (radio)chemistry, nanomaterials, and biochemistry. We describe the key early developments and the most recent advances in the field, identifying advantages and disadvantages of the different approaches and informing future development and choice of methods for clinical and preclinical application.
Collapse
Affiliation(s)
- Peter
J. Gawne
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Francis Man
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
- Institute
of Pharmaceutical Science, School of Cancer
and Pharmaceutical Sciences, King’s College London, London, SE1 9NH, U.K.
| | - Philip J. Blower
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| | - Rafael T. M. de Rosales
- School
of Biomedical Engineering & Imaging Sciences, King’s College London, St Thomas’ Hospital, London, SE1 7EH, U.K.
| |
Collapse
|
31
|
Calzetta L, Aiello M, Frizzelli A, Camardelli F, Cazzola M, Rogliani P, Chetta A. Stem Cell-Based Regenerative Therapy and Derived Products in COPD: A Systematic Review and Meta-Analysis. Cells 2022; 11:cells11111797. [PMID: 35681492 PMCID: PMC9180461 DOI: 10.3390/cells11111797] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
COPD is an incurable disorder, characterized by a progressive alveolar tissue destruction and defective mechanisms of repair and defense leading to emphysema. Currently, treatment for COPD is exclusively symptomatic; therefore, stem cell-based therapies represent a promising therapeutic approach to regenerate damaged structures of the respiratory system and restore lung function. The aim of this study was to provide a quantitative synthesis of the efficacy profile of stem cell-based regenerative therapies and derived products in COPD patients. A systematic review and meta-analysis was performed according to PRISMA-P. Data from 371 COPD patients were extracted from 11 studies. Active treatments elicited a strong tendency towards significance in FEV1 improvement (+71 mL 95% CI -2−145; p = 0.056) and significantly increased 6MWT (52 m 95% CI 18−87; p < 0.05) vs. baseline or control. Active treatments did not reduce the risk of hospitalization due to acute exacerbations (RR 0.77 95% CI 0.40−1.49; p > 0.05). This study suggests that stem cell-based regenerative therapies and derived products may be effective to treat COPD patients, but the current evidence comes from small clinical trials. Large and well-designed randomized controlled trials are needed to really quantify the beneficial impact of stem cell-based regenerative therapy and derived products in COPD.
Collapse
Affiliation(s)
- Luigino Calzetta
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.A.); (A.F.); (A.C.)
- Correspondence:
| | - Marina Aiello
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.A.); (A.F.); (A.C.)
| | - Annalisa Frizzelli
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.A.); (A.F.); (A.C.)
| | - Francesca Camardelli
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (F.C.); (M.C.); (P.R.)
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (F.C.); (M.C.); (P.R.)
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (F.C.); (M.C.); (P.R.)
| | - Alfredo Chetta
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (M.A.); (A.F.); (A.C.)
| |
Collapse
|
32
|
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
|
33
|
Yan S, Zhang C, Ji X, Wu G, Huang X, Zhang Y, Zhang Y. MSC-ACE2 Ameliorates Streptococcus uberis-Induced Inflammatory Injury in Mammary Epithelial Cells by Upregulating the IL-10/STAT3/SOCS3 Pathway. Front Immunol 2022; 13:870780. [PMID: 35677060 PMCID: PMC9167935 DOI: 10.3389/fimmu.2022.870780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
In the dairy industry, Streptococcus uberis (S. uberis) is one of the most important pathogenic bacteria associated with mastitis in milk-producing cows, causing vast economic loss. To date, the only real effective method of treating and preventing streptococcal mastitis is antimicrobial therapy. In many inflammatory diseases, mesenchymal stem cells (MSCs) and angiotensin-converting enzyme 2 (ACE2) play an anti-inflammatory and anti-injurious role. Accordingly, we hypothesized that MSCs overexpressing ACE2 (MSC-ACE2) would ameliorate the inflammatory injury caused by S. uberis in mammary epithelial cells more efficiently than MSC alone. By activating the transcription 3/suppressor of cytokine signaling 3 (IL-10/STAT3/SOCS3) signaling pathway, MSC-ACE2 inhibited the NF-κB, MAPKs, apoptosis, and pyroptosis passways. Moreover, MSC-ACE2 overturned the downregulation of Occludin, Zonula occludens 1 (ZO-1), and Claudin-3 expression levels caused by S. uberis, suggesting that MSC-ACE2 promotes the repair of the blood-milk barrier. MSC-ACE2 demonstrated greater effectiveness than MSC alone, as expected. Based on these results, MSC-ACE2 effectively inhibits EpH4-Ev cell's inflammatory responses induced by S. uberis, and would be an effective therapeutic tool for treating streptococcal mastitis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuanshu Zhang
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
34
|
Suhar RA, Doulames VM, Liu Y, Hefferon ME, Figueroa O, Buabbas H, Heilshorn SC. Hyaluronan and elastin-like protein (HELP) gels significantly improve microsphere retention in the myocardium. Biomater Sci 2022; 10:2590-2608. [PMID: 35411353 PMCID: PMC9123900 DOI: 10.1039/d1bm01890f] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Heart disease is the leading cause of death globally, and delivery of therapeutic cargo (e.g., particles loaded with proteins, drugs, or genes and cells) through direct injection into the myocardium is a promising clinical intervention. However, retention of deliverables to the contracting myocardium is low, with as much as 60-90% of payload being lost within 24 hr. Commercially-available injectable hydrogels, including Matrigel, have been hypothesized to increase payload retention but have not yielded significant improvements in quantified analyses. Here, we assess a recombinant hydrogel composed of chemically modified hyaluronan and elastin-like protein (HELP) as an alternative injectable carrier to increase cargo retention. HELP is crosslinked using dynamic covalent bonds, and tuning the hyaluronan chemistry significantly alters hydrogel mechanical properties including stiffness, stress relaxation rate, and ease of injectability through a needle or catheter. These materials can be injected even after complete crosslinking, extending the time window for surgical delivery. We show that HELP gels significantly improve in vivo retention of microsphere cargo compared to Matrigel, both 1 day and 7 days post-injection directly into the rat myocardium. These data suggest that HELP gels may assist with the clinical translation of therapeutic cargo designed for delivery into the contracting myocardium by preventing acute cargo loss.
Collapse
Affiliation(s)
- Riley A Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
| | - Vanessa M Doulames
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Yueming Liu
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
| | - Meghan E Hefferon
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | - Hana Buabbas
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
- Department of Biology, Stanford University, Stanford, California, 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
| |
Collapse
|
35
|
Holthaus M, Santhakumar N, Wahlers T, Paunel-Görgülü A. The Secretome of Preconditioned Mesenchymal Stem Cells Drives Polarization and Reprogramming of M2a Macrophages toward an IL-10-Producing Phenotype. Int J Mol Sci 2022; 23:ijms23084104. [PMID: 35456922 PMCID: PMC9024470 DOI: 10.3390/ijms23084104] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 12/13/2022] Open
Abstract
The preconditioning of mesenchymal stem cells (MSCs) has been recognized as an attractive tool to improve their regenerative and immunomodulatory capacities based on their paracrine effects. In this study, we examined the potential of an MSC-conditioned medium (MSC-CM) to alter the phenotype of murine macrophages and to drive reprogramming toward an anti-inflammatory, M2-like state in vitro. We further explored the impact of MSC cytokine preconditioning on the immunosuppressive properties of the MSC secretome. The MSC-CM suppressed the expression of proinflammatory genes in murine M1 macrophages, but only the CM from preconditioned MSCs (preMSC-CM) downregulated their expression during M1 polarization. Remarkably, only the preMSC-CM significantly increased the expression of M2a-, M2b- and M2c-specific genes and proteins during M2a polarization. Further, macrophages were found to secrete high levels of anti-inflammatory IL-10. Similarly, M2a macrophages cultured in the presence of the preMSC-CM displayed an enhanced expression of M2b/M2c-specific markers, suggesting that the secretome of preMSC promotes the repolarization of M2a-like macrophages to M2b/M2c subtypes. The preMSC-CM was found to be enriched in molecules involved in M2 polarization. Additionally, a unique downregulation of extracellular matrix components was observed. Altogether, the preMSC-CM may provide an attractive strategy to dampen inflammation by suppressing the expression of proinflammatory mediators and promoting the polarization and phenotype switch of M2a cells to IL-10-secreting M2b/M2c-like macrophages.
Collapse
Affiliation(s)
- Michelle Holthaus
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, 50937 Cologne, Germany
| | - Nivethiha Santhakumar
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, 50937 Cologne, Germany
| | - Thorsten Wahlers
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, 50937 Cologne, Germany
| | - Adnana Paunel-Görgülü
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
36
|
Shalaby N, Kelly J, Martinez F, Fox M, Qi Q, Thiessen J, Hicks J, Scholl TJ, Ronald JA. A Human-derived Dual MRI/PET Reporter Gene System with High Translational Potential for Cell Tracking. Mol Imaging Biol 2022; 24:341-351. [PMID: 35146614 PMCID: PMC9235057 DOI: 10.1007/s11307-021-01697-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Reporter gene imaging has been extensively used to longitudinally report on whole-body distribution and viability of transplanted engineered cells. Multi-modal cell tracking can provide complementary information on cell fate. Typical multi-modal reporter gene systems often combine clinical and preclinical modalities. A multi-modal reporter gene system for magnetic resonance imaging (MRI) and positron emission tomography (PET), two clinical modalities, would be advantageous by combining the sensitivity of PET with the high-resolution morphology and non-ionizing nature of MRI. PROCEDURES We developed and evaluated a dual MRI/PET reporter gene system composed of two human-derived reporter genes that utilize clinical reporter probes for engineered cell detection. As a proof-of-concept, breast cancer cells were engineered to co-express the human organic anion transporter polypeptide 1B3 (OATP1B3) that uptakes the clinical MRI contrast agent gadolinium ethoxybenzyl-diethylenetriaminepentaacetic acid (Gd-EOB-DTPA), and the human sodium iodide symporter (NIS) which uptakes the PET tracer, [18F] tetrafluoroborate ([18F] TFB). RESULTS T1-weighted MRI results in mice exhibited significantly higher MRI signals in reporter-gene-engineered mammary fat pad tumors versus contralateral naïve tumors (p < 0.05). No differences in contrast enhancement were observed at 5 h after Gd-EOB-DTPA administration using either intravenous or intraperitoneal injection. We also found significantly higher standard uptake values (SUV) in engineered tumors in comparison to the naïve tumors in [18F]TFB PET images (p < 0.001). Intratumoral heterogeneity in signal enhancement was more conspicuous in relatively higher resolution MR images compared to PET images. CONCLUSIONS Our study demonstrates the ability to noninvasively track cells engineered with our human-derived dual MRI/PET reporter system, enabling a more comprehensive evaluation of transplanted cells. Future work is focused on applying this tool to track therapeutic cells, which may one day enable the broader application of cell tracking within the healthcare system.
Collapse
Affiliation(s)
- Nourhan Shalaby
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada.
| | - John Kelly
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Francisco Martinez
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Mathew Fox
- Lawson Health Research Institute, London, Canada
- Saint Joseph's Health Care, Toronto, Canada
| | - Qi Qi
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Jonathan Thiessen
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Saint Joseph's Health Care, Toronto, Canada
- Lawson Cyclotron and Radiochemistry Facility, London, Canada
| | - Justin Hicks
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
- Lawson Cyclotron and Radiochemistry Facility, London, Canada
| | - Timothy J Scholl
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Ontario Institute for Cancer Research, Toronto, Canada
| | - John A Ronald
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Lawson Health Research Institute, London, Canada
| |
Collapse
|
37
|
Harvell-Smith S, Tung LD, Thanh NTK. Magnetic particle imaging: tracer development and the biomedical applications of a radiation-free, sensitive, and quantitative imaging modality. NANOSCALE 2022; 14:3658-3697. [PMID: 35080544 DOI: 10.1039/d1nr05670k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Magnetic particle imaging (MPI) is an emerging tracer-based modality that enables real-time three-dimensional imaging of the non-linear magnetisation produced by superparamagnetic iron oxide nanoparticles (SPIONs), in the presence of an external oscillating magnetic field. As a technique, it produces highly sensitive radiation-free tomographic images with absolute quantitation. Coupled with a high contrast, as well as zero signal attenuation at-depth, there are essentially no limitations to where that can be imaged within the body. These characteristics enable various biomedical applications of clinical interest. In the opening sections of this review, the principles of image generation are introduced, along with a detailed comparison of the fundamental properties of this technique with other common imaging modalities. The main feature is a presentation on the up-to-date literature for the development of SPIONs tailored for improved imaging performance, and developments in the current and promising biomedical applications of this emerging technique, with a specific focus on theranostics, cell tracking and perfusion imaging. Finally, we will discuss recent progress in the clinical translation of MPI. As signal detection in MPI is almost entirely dependent on the properties of the SPION employed, this work emphasises the importance of tailoring the synthetic process to produce SPIONs demonstrating specific properties and how this impacts imaging in particular applications and MPI's overall performance.
Collapse
Affiliation(s)
- Stanley Harvell-Smith
- Biophysics Group, Department of Physics and Astronomy, University College London, Gower Street, London WC1E 6BT, UK.
- UCL Healthcare Biomagnetic and Nanomaterials Laboratories, University College London, 21 Albemarle Street, London W1S 4BS, UK
| | - Le Duc Tung
- Biophysics Group, Department of Physics and Astronomy, University College London, Gower Street, London WC1E 6BT, UK.
- UCL Healthcare Biomagnetic and Nanomaterials Laboratories, University College London, 21 Albemarle Street, London W1S 4BS, UK
| | - Nguyen Thi Kim Thanh
- Biophysics Group, Department of Physics and Astronomy, University College London, Gower Street, London WC1E 6BT, UK.
- UCL Healthcare Biomagnetic and Nanomaterials Laboratories, University College London, 21 Albemarle Street, London W1S 4BS, UK
| |
Collapse
|
38
|
Dethe MR, A P, Ahmed H, Agrawal M, Roy U, Alexander A. PCL-PEG copolymer based injectable thermosensitive hydrogels. J Control Release 2022; 343:217-236. [PMID: 35090961 PMCID: PMC9134269 DOI: 10.1016/j.jconrel.2022.01.035] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 01/09/2023]
Abstract
A number of stimuli-responsive-based hydrogels has been widely explored in biomedical applications in the last few decades because of their excellent biodegradability and biocompatibility. The development of synthetic chemistry and materials science leads to the emergence of in situ stimuli-responsive hydrogels. In this regard, several synthetic and natural polymers have been synthesized and utilized to prepare temperature-sensitive in situ forming hydrogels. This could be best used via injections as temperature stimulus could trigger in situ hydrogels gelation and swelling behaviors. There are many smart polymers available for the formulation of the in situ based thermoresponsive injectable hydrogel. Among these, poly (ε-caprolactone) (PCL) polymer has been recognized and approved by the FDA for numerous biomedical applications. More specifically, the PCL is coupled with polyethylene glycol (PEG) to obtain amphiphilic thermosensitive "smart" copolymers (PCL-PEG), to form rapid and reversible physical gelation behavior. However, the chemical structure of the copolymer is a critical aspect in determining water solubility, thermo-gelation behavior, drug release rate, degradation rate, and the possibility to deliver a diverse range of drugs. In this review, we have highlighted the typical PCL-PEG-based thermosensitive injectable hydrogels progress in the last decade for tissue engineering and localized drug delivery applications to treat various diseases. Additionally, the impact of molecular weight of PCL-PEG upon gelling behavior has also been critically highlighted for optimum hydrogels properties for potential pharmaceutical and biomedical applications.
Collapse
Affiliation(s)
- Mithun Rajendra Dethe
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati 781101, India
| | - Prabakaran A
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati 781101, India
| | - Hafiz Ahmed
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati 781101, India
| | - Mukta Agrawal
- School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Polepally SEZ, TSIIC Jadcherla, Hyderabad 509301, India
| | - Upal Roy
- Department of Health and Biomedical Sciences, College of Health Affairs, One West University Blvd., Brownsville, TX 78520, United States of America
| | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati 781101, India.
| |
Collapse
|
39
|
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: 1.5] [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
|
40
|
Angiogenic Effects and Crosstalk of Adipose-Derived Mesenchymal Stem/Stromal Cells and Their Extracellular Vesicles with Endothelial Cells. Int J Mol Sci 2021; 22:ijms221910890. [PMID: 34639228 PMCID: PMC8509224 DOI: 10.3390/ijms221910890] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived mesenchymal stem/stromal cells (ASCs) are an adult stem cell population able to self-renew and differentiate into numerous cell lineages. ASCs provide a promising future for therapeutic angiogenesis due to their ability to promote blood vessel formation. Specifically, their ability to differentiate into endothelial cells (ECs) and pericyte-like cells and to secrete angiogenesis-promoting growth factors and extracellular vesicles (EVs) makes them an ideal option in cell therapy and in regenerative medicine in conditions including tissue ischemia. In recent angiogenesis research, ASCs have often been co-cultured with an endothelial cell (EC) type in order to form mature vessel-like networks in specific culture conditions. In this review, we introduce co-culture systems and co-transplantation studies between ASCs and ECs. In co-cultures, the cells communicate via direct cell-cell contact or via paracrine signaling. Most often, ASCs are found in the perivascular niche lining the vessels, where they stabilize the vascular structures and express common pericyte surface proteins. In co-cultures, ASCs modulate endothelial cells and induce angiogenesis by promoting tube formation, partly via secretion of EVs. In vivo co-transplantation of ASCs and ECs showed improved formation of functional vessels over a single cell type transplantation. Adipose tissue as a cell source for both mesenchymal stem cells and ECs for co-transplantation serves as a prominent option for therapeutic angiogenesis and blood perfusion in vivo.
Collapse
|
41
|
Kim SJ, Lee S, Kim C, Shin H. One-step harvest and delivery of micropatterned cell sheets mimicking the multi-cellular microenvironment of vascularized tissue. Acta Biomater 2021; 132:176-187. [PMID: 33571713 DOI: 10.1016/j.actbio.2021.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/15/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022]
Abstract
Techniques for harvest and delivery of cell sheets have been improving for decades. However, cell sheets with complicated patterns closely related to natural tissue architecture were hardly achieved. Here, we developed an efficient method to culture and harvest cell sheets with complex shape (noted as microtissues) using temperature-responsive hydrogel consisting of expandable polyethylene oxide polymer at low temperature. Firstly, a temperature-responsive hydrogel surface with honeycomb patterns (50 and 100 µm in width) were developed through microcontact printing of polydopamine (PD). The human dermal fibroblasts (HDFBs) and human umbilical vein endothelial cells (HUVECs) spontaneously formed honeycomb-shaped microtissues on the patterned hydrogel surface. The microtissues on the hydrogel were able to be harvested and directly delivered to the desired target through thermal expansion of the hydrogel at 4 °C with an efficiency close to 80% within 10 min which is faster than conventional method based on poly(N-isopropylacrylamide). The microtissues maintained their original honeycomb network and intact structures. Honeycomb-patterned cell sheets also were fabricated through serial seeding of various cell lines, including HDFBs, HUVECs, and human adipose-derived stem cells, in which cells were attached along the honeycomb pattern. The underlying honeycomb patterns in the cell sheets were successfully maintained for 3 days, even after delivery. In addition, patterned cell sheets were successfully delivered in vivo while maintaining an intact structure for 7 days. Together, our findings demonstrate that micropatterned temperature-responsive hydrogel is an efficient method of one-step culturing and delivery of complex microtissues and should prove useful in various tissue engineering applications. STATEMENT OF SIGNIFICANCE: Scaffold-free cell delivery techniques, including cell sheet engineering, have been developed for decades. However, there is limited research regarding culture and delivery of microtissues with complex architecture mimicking natural tissue. Herein, we developed a micro-patterned hydrogel platform for the culture and delivery of honeycomb-shaped microtissues. Honeycomb patterns were chemically engineered on the temperature-responsive hydrogel through microcontact printing of polydopamine to selectively allow for human dermal fibroblast or human umbilical vein endothelial cell adhesion. They spontaneously formed honeycomb-shaped microtissues within 24 hr upon cell seeding and directly delivered to various target area including in vivo via thermal expansion of the hydrogel at 4 °C, suggesting that the micro-patterned hydrogel can be an efficient tool for culture and delivery of complex microtissue.
Collapse
Affiliation(s)
- Se-Jeong Kim
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Sangmin Lee
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Chunggoo Kim
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; BK21 FOUR, Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea; Institute of Nano Science and Technology (INST), Hanyang University, 222 Wangsimri-ro, Seongdong-gu, Seoul 04763, Republic of Korea.
| |
Collapse
|
42
|
Helfer BM, Ponomarev V, Patrick PS, Blower PJ, Feitel A, Fruhwirth GO, Jackman S, Pereira Mouriès L, Park MVDZ, Srinivas M, Stuckey DJ, Thu MS, van den Hoorn T, Herberts CA, Shingleton WD. Options for imaging cellular therapeutics in vivo: a multi-stakeholder perspective. Cytotherapy 2021; 23:757-773. [PMID: 33832818 PMCID: PMC9344904 DOI: 10.1016/j.jcyt.2021.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/01/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022]
Abstract
Cell-based therapies have been making great advances toward clinical reality. Despite the increase in trial activity, few therapies have successfully navigated late-phase clinical trials and received market authorization. One possible explanation for this is that additional tools and technologies to enable their development have only recently become available. To support the safety evaluation of cell therapies, the Health and Environmental Sciences Institute Cell Therapy-Tracking, Circulation and Safety Committee, a multisector collaborative committee, polled the attendees of the 2017 International Society for Cell & Gene Therapy conference in London, UK, to understand the gaps and needs that cell therapy developers have encountered regarding safety evaluations in vivo. The goal of the survey was to collect information to inform stakeholders of areas of interest that can help ensure the safe use of cellular therapeutics in the clinic. This review is a response to the cellular imaging interests of those respondents. The authors offer a brief overview of available technologies and then highlight the areas of interest from the survey by describing how imaging technologies can meet those needs. The areas of interest include imaging of cells over time, sensitivity of imaging modalities, ability to quantify cells, imaging cellular survival and differentiation and safety concerns around adding imaging agents to cellular therapy protocols. The Health and Environmental Sciences Institute Cell Therapy-Tracking, Circulation and Safety Committee believes that the ability to understand therapeutic cell fate is vital for determining and understanding cell therapy efficacy and safety and offers this review to aid in those needs. An aim of this article is to share the available imaging technologies with the cell therapy community to demonstrate how these technologies can accomplish unmet needs throughout the translational process and strengthen the understanding of cellular therapeutics.
Collapse
Affiliation(s)
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - P Stephen Patrick
- Department of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Philip J Blower
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Alexandra Feitel
- Formerly, Health and Environmental Sciences Institute, US Environmental Protection Agency, Washington, DC, USA
| | - Gilbert O Fruhwirth
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Shawna Jackman
- Charles River Laboratories, Shrewsbury, Massachusetts, USA
| | | | - Margriet V D Z Park
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Mangala Srinivas
- Department of Tumor Immunology, Radboud University Medical Center, Nijmegen, the Netherlands; Cenya Imaging BV, Amsterdam, the Netherlands
| | - Daniel J Stuckey
- Department of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Mya S Thu
- Visicell Medical Inc, La Jolla, California, USA
| | | | | | | |
Collapse
|
43
|
Lechermann LM, Lau D, Attili B, Aloj L, Gallagher FA. In Vivo Cell Tracking Using PET: Opportunities and Challenges for Clinical Translation in Oncology. Cancers (Basel) 2021; 13:4042. [PMID: 34439195 PMCID: PMC8392745 DOI: 10.3390/cancers13164042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/21/2022] Open
Abstract
Cell therapy is a rapidly evolving field involving a wide spectrum of therapeutic cells for personalised medicine in cancer. In vivo imaging and tracking of cells can provide useful information for improving the accuracy, efficacy, and safety of cell therapies. This review focuses on radiopharmaceuticals for the non-invasive detection and tracking of therapeutic cells using positron emission tomography (PET). A range of approaches for imaging therapeutic cells is discussed: Direct ex vivo labelling of cells, in vivo indirect labelling of cells by utilising gene reporters, and detection of specific antigens expressed on the target cells using antibody-based radiopharmaceuticals (immuno-PET). This review examines the evaluation of PET imaging methods for therapeutic cell tracking in preclinical cancer models, their role in the translation into patients, first-in-human studies, as well as the translational challenges involved and how they can be overcome.
Collapse
Affiliation(s)
- Laura M. Lechermann
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (B.A.); (L.A.); (F.A.G.)
- Cancer Research UK Cambridge Centre, Cambridge CB2 0RE, UK
| | - Doreen Lau
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (B.A.); (L.A.); (F.A.G.)
- Cancer Research UK Cambridge Centre, Cambridge CB2 0RE, UK
| | - Bala Attili
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (B.A.); (L.A.); (F.A.G.)
- Cancer Research UK Cambridge Centre, Cambridge CB2 0RE, UK
| | - Luigi Aloj
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (B.A.); (L.A.); (F.A.G.)
- Cancer Research UK Cambridge Centre, Cambridge CB2 0RE, UK
- Department of Nuclear Medicine, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Ferdia A. Gallagher
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (B.A.); (L.A.); (F.A.G.)
- Cancer Research UK Cambridge Centre, Cambridge CB2 0RE, UK
| |
Collapse
|
44
|
Yang N, Moore MJ, Michael PL, Santos M, Lam YT, Bao S, Ng MKC, Rnjak‐Kovacina J, Tan RP, Wise SG. Silk Fibroin Scaffold Architecture Regulates Inflammatory Responses and Engraftment of Bone Marrow-Mononuclear Cells. Adv Healthc Mater 2021; 10:e2100615. [PMID: 33963682 DOI: 10.1002/adhm.202100615] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Indexed: 12/13/2022]
Abstract
Despite being one of the most clinically trialed cell therapies, bone marrow-mononuclear cell (BM-MNC) infusion has largely failed to fulfill its clinical promise. Implanting biomimetic scaffolds at sites of injury prior to BM-MNC infusion is a promising approach to enhance BM-MNC engraftment and therapeutic function. Here, it is demonstrated that scaffold architecture can be leveraged to regulate the immune responses that drive BM-MNC engraftment. Silk scaffolds with thin fibers and low porosity (LP) impairs immune activation in vitro compared with thicker fiber, high porosity (HP) scaffolds. Using the authors' established in vivo bioluminescent BM-MNC tracking model, they showed that BM-MNCs home to and engraft in greater numbers in HP scaffolds over 14 days. Histological analysis reveals thicker fibrous capsule formation, with enhanced collagen deposition in HP compared to LP scaffolds consistent with substantially more native CD68+ macrophages and CD4+ T cells, driven by their elevated pro-inflammatory M1 and Th1 phenotypes, respectively. These results suggest that implant architecture impacts local inflammation that drives differential engraftment and remodeling behavior of infused BM-MNC. These findings inform the future design of biomimetic scaffolds that may better enhance the clinical effectiveness of BM-MNC infusion therapy.
Collapse
Affiliation(s)
- Nianji Yang
- School of Medical Sciences Faculty of Health and Medicine The University of Sydney Sydney NSW 2006 Australia
- Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
| | - Matthew J. Moore
- School of Medical Sciences Faculty of Health and Medicine The University of Sydney Sydney NSW 2006 Australia
- Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
| | - Praveesuda L. Michael
- School of Medical Sciences Faculty of Health and Medicine The University of Sydney Sydney NSW 2006 Australia
- Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
| | - Miguel Santos
- School of Medical Sciences Faculty of Health and Medicine The University of Sydney Sydney NSW 2006 Australia
- Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
| | - Yuen Ting Lam
- School of Medical Sciences Faculty of Health and Medicine The University of Sydney Sydney NSW 2006 Australia
- Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
| | - Shisan Bao
- School of Medical Sciences Faculty of Health and Medicine The University of Sydney Sydney NSW 2006 Australia
- Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
| | - Martin K. C. Ng
- Sydney Medical School The University of Sydney Sydney NSW 2006 Australia
- Department of Cardiology Royal Prince Alfred Hospital Sydney NSW 2042 Australia
| | - Jelena Rnjak‐Kovacina
- Graduate School of Biomedical Engineering University of New South Wales Sydney NSW 2052 Australia
| | - Richard P. Tan
- School of Medical Sciences Faculty of Health and Medicine The University of Sydney Sydney NSW 2006 Australia
- Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
| | - Steven G. Wise
- School of Medical Sciences Faculty of Health and Medicine The University of Sydney Sydney NSW 2006 Australia
- Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
- The University of Sydney Nano Institute (Sydney Nano) The University of Sydney Sydney NSW 2006 Australia
| |
Collapse
|
45
|
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: 0.7] [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
|
46
|
He X, Hong W, Yang J, Lei H, Lu T, He C, Bi Z, Pan X, Liu Y, Dai L, Wang W, Huang C, Deng H, Wei X. Spontaneous apoptosis of cells in therapeutic stem cell preparation exert immunomodulatory effects through release of phosphatidylserine. Signal Transduct Target Ther 2021; 6:270. [PMID: 34262012 PMCID: PMC8280232 DOI: 10.1038/s41392-021-00688-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/08/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cell (MSC)-mediated immunomodulation has been harnessed for the treatment of human diseases, but its underlying mechanism has not been fully understood. Dead cells, including apoptotic cells have immunomodulatory properties. It has been repeatedly reported that the proportion of nonviable MSCs in a MSC therapeutic preparation varied from 5~50% in the ongoing clinical trials. It is conceivable that the nonviable cells in a MSC therapeutic preparation may play a role in the therapeutic effects of MSCs. We found that the MSC therapeutic preparation in the present study had about 5% dead MSCs (DMSCs), characterized by apoptotic cells. Namely, 1 × 106 MSCs in the preparation contained about 5 × 104 DMSCs. We found that the treatment with even 5 × 104 DMSCs alone had the equal therapeutic effects as with 1 × 106 MSCs. This protective effect of the dead MSCs alone was confirmed in four mouse models, including concanavalin A (ConA)- and carbon tetrachloride (CCl4)-induced acute liver injury, LPS-induced lung injury and spinal cord injury. We also found that the infused MSCs died by apoptosis in vivo. Furthermore, the therapeutic effect was attributed to the elevated level of phosphatidylserine (PS) upon the injection of MSCs or DMSCs. The direct administration of PS liposomes (PSLs) mimic apoptotic cell fragments also exerted the protective effects as MSCs and DMSCs. The Mer tyrosine kinase (MerTK) deficiency or the knockout of chemokine receptor C-C motif chemokine receptor 2 (CCR2) reversed these protective effects of MSCs or DMSCs. These results revealed that DMSCs alone in the therapeutic stem cell preparation or the apoptotic cells induced in vivo may exert the same immunomodulatory property as the "living MSCs preparation" through releasing PS, which was further recognized by MerTK and participated in modulating immune cells.
Collapse
Affiliation(s)
- Xuemei He
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China ,grid.488387.8Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan People’s Republic of China
| | - Weiqi Hong
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Jingyun Yang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Hong Lei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Tianqi Lu
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Cai He
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Zhenfei Bi
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Xiangyu Pan
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Yu Liu
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Lunzhi Dai
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Wei Wang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Canhua Huang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Hongxin Deng
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Xiawei Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan People’s Republic of China
| |
Collapse
|
47
|
Abstract
The role of stem cells in augmenting reparative processes in the heart after ischemic injury has been successfully demonstrated in small and large animal models. However, the outcomes of cell therapy in clinical trials have been somewhat variable, with overall effects of autologous stem cell therapies demonstrating a modest improvement in cardiac structure and function. How stem cells repair the heart after cardiac injury is still not well understood. Most recent studies suggest that adult derived stem cells act primarily through paracrine signaling to exert beneficial effects, including modulation of immune response, stimulation of new blood vessel formation, or by inducing mature myocytes to transiently reenter the cell cycle, rather than robust direct differentiation of the transplanted cells into myocytes. In addition, data from multiple laboratory results confirmed clearance of stem cells themselves within a few days still leading to functional benefits further confirming the role of paracrine signaling in augmenting cardiac reparative processes rather than direct differentiation of cells. These findings rapidly evolved the field of extracellular vesicles specifically microvesicles (MVs) as they are active hubs of autocrine, paracrine, and endocrine signaling targeting different biological processes. The beneficial effects seen after stem cell transplantation could be linked to the cardioprotective factors packaged in the MVs secreted from stem cells. Therefore, stem cell MVs provide a new avenue for the treatment of cardiovascular disease through a multitude of mechanisms including cellular communication within the stem cell niches, delivery of genetic information, regulation of the immune system in the heart, and stimulation of angiogenesis which will be discussed in this review.
Collapse
|
48
|
Tripathi H, Peng H, Donahue R, Chelvarajan L, Gottipati A, Levitan B, Al-Darraji A, Gao E, Abdel-Latif A, Berron BJ. Isolation Methods for Human CD34 Subsets Using Fluorescent and Magnetic Activated Cell Sorting: an In Vivo Comparative Study. Stem Cell Rev Rep 2021; 16:413-423. [PMID: 31953639 DOI: 10.1007/s12015-019-09939-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Acute myocardial infarction (AMI) and resulting cardiac damage and heart failure are leading causes of morbidity and mortality worldwide. Multiple studies have examined the utility of CD34+ cells for the treatment of acute and ischemic heart disease. However, the optimal strategy to enrich CD34 cells from clinical sources is not known. We examined the efficacy of fluorescence activated cell sorting (FACS) and magnetic beads cell sorting (MACS) methods for CD34 cell isolation from mobilized human mononuclear peripheral blood cells (mhPBMNCs). METHODS mhPBCs were processed following acquisition using FACS or MACS according to clinically established protocols. Cell viability, CD34 cell purity and characterization of surface marker expression were assessed using a flow cytometer. For in vivo characterization of cardiac repair, we conducted LAD ligation surgery on 8-10 weeks female NOD/SCID mice followed by intramyocardial transplantation of unselected mhPBMNCs, FACS or MACS enriched CD34+ cells. RESULTS Both MACS and FACS isolation methods achieved high purity rates, viability, and enrichment of CD34+ cells. In vivo studies following myocardial infarction demonstrated retention of CD34+ in the peri-infarct region for up to 30 days after transplantation. Retained CD34+ cells were associated with enhanced angiogenesis and reduced inflammation compared to unselected mhPBMNCs or PBS treatment arms. Cardiac scar and fibrosis as assessed by immunohistochemistry were reduced in FACS and MACS CD34+ treatment groups. Finally, reduced scar and augmented angiogenesis resulted in improved cardiac functional recovery, both on the global and regional function and remodeling assessments by echocardiography. CONCLUSION Cell based therapy using enriched CD34+ cells sorted by FACS or MACS result in better cardiac recovery after ischemic injury compared to unselected mhPBMNCs. Both enrichment techniques offer excellent recovery and purity and can be equally used for clinical applications.
Collapse
Affiliation(s)
- Himi Tripathi
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Hsuan Peng
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Renee Donahue
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Lakshman Chelvarajan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Anuhya Gottipati
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY, USA
| | - Bryana Levitan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Ahmed Al-Darraji
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Erhe Gao
- The Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Ahmed Abdel-Latif
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, KY, USA
| | - Bradley J Berron
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
49
|
Therapies to prevent post-infarction remodelling: From repair to regeneration. Biomaterials 2021; 275:120906. [PMID: 34139506 DOI: 10.1016/j.biomaterials.2021.120906] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/02/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
Myocardial infarction is the first cause of worldwide mortality, with an increasing incidence also reported in developing countries. Over the past decades, preclinical research and clinical trials continually tested the efficacy of cellular and acellular-based treatments. However, none of them resulted in a drug or device currently used in combination with either percutaneous coronary intervention or coronary artery bypass graft. Inflammatory, proliferation and remodelling phases follow the ischaemic event in the myocardial tissue. Only recently, single-cell sequencing analyses provided insights into the specific cell populations which determine the final fibrotic deposition in the affected region. In this review, ischaemia, inflammation, fibrosis, angiogenesis, cellular stress and fundamental cellular and molecular components are evaluated as therapeutic targets. Given the emerging evidence of biomaterial-based systems, the increasing use of injectable hydrogels/scaffolds and epicardial patches is reported both as acellular and cellularised/functionalised treatments. Since several variables influence the outcome of any experimented treatment, we return to the pathological basis with an unbiased view towards any specific process or cellular component. Thus, by evaluating the benefits and limitations of the approaches based on these targets, the reader can weigh the rationale of each of the strategies that reached the clinical trials stage. As recent studies focused on the relevance of the extracellular matrix in modulating ischaemic remodelling and enhancing myocardial regeneration, we aim to portray current trends in the field with this review. Finally, approaches towards feasible translational studies that are as yet unexplored are also suggested.
Collapse
|
50
|
Ghanta RK, Aghlara-Fotovat S, Pugazenthi A, Ryan CT, Singh VP, Mathison M, Jarvis MI, Mukherjee S, Hernandez A, Veiseh O. Immune-modulatory alginate protects mesenchymal stem cells for sustained delivery of reparative factors to ischemic myocardium. Biomater Sci 2021; 8:5061-5070. [PMID: 32797143 DOI: 10.1039/d0bm00855a] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Paracrine factors secreted by mesenchymal stem cells (MSCs) have been previously shown to improve cardiac function following acute myocardial infarction (MI). However, cell therapy activates the innate immune response, leading to the rapid elimination of transplanted cells and only short-term therapeutic delivery. Herein, we describe a new strategy to deliver sustained paracrine-mediated MSC therapy to ischemic myocardium. Using an immune evasive, small molecule modified alginate, we encapsulated rat MSC cells in a core-shell hydrogel capsule and implanted them in the pericardial sac of post-MI rats. Encapsulated cells allowed diffusion of reparative paracrine factors at levels similar to non-encapsulated cells in vitro. Encapsulation enabled sustained cell survival with localization over the heart for 2 weeks. The effect of the experimental group on ventricular function and fibrosis was compared with blank (cell free) capsules and unencapsulated MSCs injected into infarcted myocardium. MSC capsules improved post-MI ventricular function ∼2.5× greater than MSC injection. After 4 weeks, post-MI fibrosis was reduced ∼2/3 with MSC capsules, but unchanged with MSC injection. MSC encapsulation with alginate core-shell capsules sustains cell survival and potentiates efficacy of therapy.
Collapse
Affiliation(s)
- Ravi K Ghanta
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | | | - Aarthi Pugazenthi
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Christopher T Ryan
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Vivek P Singh
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Megumi Mathison
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Maria I Jarvis
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Andrea Hernandez
- Department of Bioengineering, Rice University, Houston, TX, USA.
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|