1
|
Chi A, Yang B, Dai H, Li X, Mo J, Gao Y, Chen Z, Feng X, Ma M, Li Y, Yang C, Liu J, Liu H, Wang Z, Gao F, Liao Y, Shi X, Deng C, Zhang M. Stem Leydig cells support macrophage immunological homeostasis through mitochondrial transfer in mice. Nat Commun 2024; 15:2120. [PMID: 38459012 PMCID: PMC10924100 DOI: 10.1038/s41467-024-46190-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 02/16/2024] [Indexed: 03/10/2024] Open
Abstract
As testicular mesenchymal stromal cells, stem Leydig cells (SLCs) show great promise in the treatment of male hypogonadism. The therapeutic functions of mesenchymal stromal cells are largely determined by their reciprocal regulation by immune responses. However, the immunoregulatory properties of SLCs remain unclear. Here, we observe that SLCs transplantation restore male fertility and testosterone production in an ischemia‒reperfusion injury mouse model. SLCs prevent inflammatory cascades through mitochondrial transfer to macrophages. Reactive oxygen species (ROS) released from activated macrophages inducing mitochondrial transfer from SLCs to macrophages in a transient receptor potential cation channel subfamily member 7 (TRPM7)-mediated manner. Notably, knockdown of TRPM7 in transplanted SLCs compromised therapeutic outcomes in both testicular ischemia‒reperfusion and testicular aging mouse models. These findings reveal a new mechanism of SLCs transplantation that may contribute to preserve testis function in male patients with hypogonadism related to immune disorders.
Collapse
Affiliation(s)
- Ani Chi
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Bicheng Yang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hao Dai
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Xinyu Li
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiahui Mo
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yong Gao
- Reproductive Medicine Center, The Key Laboratory for Reproductive Medicine of Guangdong Province, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zhihong Chen
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xin Feng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Menghui Ma
- Center of Reproductive Medicine, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Yanqing Li
- Center of Reproductive Medicine, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Chao Yang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Jie Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Hanchao Liu
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhenqing Wang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Feng Gao
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Reproductive Medicine Center, The Key Laboratory for Reproductive Medicine of Guangdong Province, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Yan Liao
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Xuetao Shi
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
- National Engineering Research Centre for Tissue Restoration and Reconstruction and Key Laboratory of Biomedical Engineering of Guangdong Province South China University of Technology, Guangzhou, 510640, China.
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, 518054, China.
| | - Chunhua Deng
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Min Zhang
- Department of Andrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
2
|
Lin YH, Lehle JD, McCarrey JR. Source cell-type epigenetic memory persists in induced pluripotent cells but is lost in subsequently derived germline cells. Front Cell Dev Biol 2024; 12:1306530. [PMID: 38410371 PMCID: PMC10895008 DOI: 10.3389/fcell.2024.1306530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/24/2024] [Indexed: 02/28/2024] Open
Abstract
Introduction: Retention of source cell-type epigenetic memory may mitigate the potential for induced pluripotent stem cells (iPSCs) to fully achieve transitions in cell fate in vitro. While this may not preclude the use of iPSC-derived somatic cell types for therapeutic applications, it becomes a major concern impacting the potential use of iPSC-derived germline cell types for reproductive applications. The transition from a source somatic cell type to iPSCs and then on to germ-cell like cells (GCLCs) recapitulates two major epigenetic reprogramming events that normally occur during development in vivo-embryonic reprogramming in the epiblast and germline reprogramming in primordial germ cells (PGCs). We examined the extent of epigenetic and transcriptomic memory persisting first during the transition from differentiated source cell types to iPSCs, and then during the transition from iPSCs to PGC-like cells (PGCLCs). Methods: We derived iPSCs from four differentiated mouse cell types including two somatic and two germ cell types and tested the extent to which each resulting iPSC line resembled a) a validated ES cell reference line, and b) their respective source cell types, on the basis of genome-wide gene expression and DNA methylation patterns. We then induced each iPSC line to form PGCLCs, and assessed epigenomic and transcriptomic memory in each compared to endogenous PGCs/M-prospermatogonia. Results: In each iPSC line, we found residual gene expression and epigenetic programming patterns characteristic of the corresponding source differentiated cell type from which each was derived. However, upon deriving PGCLCs, we found very little evidence of lingering epigenetic or transcriptomic memory of the original source cell type. Discussion: This result indicates that derivation of iPSCs and then GCLCs from differentiated source cell types in vitro recapitulates the two-phase epigenetic reprogramming that normally occurs in vivo, and that, to a significant extent, germline cell types derived in vitro from pluripotent cells accurately recapitulate epigenetic programming and gene expression patterns corresponding to equivalent endogenous germ cell types, suggesting that they have the potential to form the basis of in vitro gametogenesis as a useful therapeutic strategy for treatment of infertility.
Collapse
Affiliation(s)
- Yu-Huey Lin
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Jake D Lehle
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
| | - John R McCarrey
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
3
|
Cardiac Cell Therapy with Pluripotent Stem Cell-Derived Cardiomyocytes: What Has Been Done and What Remains to Do? Curr Cardiol Rep 2022; 24:445-461. [PMID: 35275365 PMCID: PMC9068652 DOI: 10.1007/s11886-022-01666-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/05/2022] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW Exciting pre-clinical data presents pluripotent stem cell-derived cardiomyocytes (PSC-CM) as a novel therapeutic prospect following myocardial infarction, and worldwide clinical trials are imminent. However, despite notable advances, several challenges remain. Here, we review PSC-CM pre-clinical studies, identifying key translational hurdles. We further discuss cell production and characterization strategies, identifying markers that may help generate cells which overcome these barriers. RECENT FINDINGS PSC-CMs can robustly repopulate infarcted myocardium with functional, force generating cardiomyocytes. However, current differentiation protocols produce immature and heterogenous cardiomyocytes, creating related issues such as arrhythmogenicity, immunogenicity and poor engraftment. Recent efforts have enhanced our understanding of cardiovascular developmental biology. This knowledge may help implement novel differentiation or gene editing strategies that could overcome these limitations. PSC-CMs are an exciting therapeutic prospect. Despite substantial recent advances, limitations of the technology remain. However, with our continued and increasing biological understanding, these issues are addressable, with several worldwide clinical trials anticipated in the coming years.
Collapse
|
4
|
Akter F, Araf Y, Naser IB, Promon SK. Prospect of 3D bioprinting over cardiac cell therapy and conventional tissue engineering in the treatment of COVID-19 patients with myocardial injury. Regen Ther 2021; 18:447-456. [PMID: 34608441 PMCID: PMC8481096 DOI: 10.1016/j.reth.2021.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 09/01/2021] [Accepted: 09/23/2021] [Indexed: 01/08/2023] Open
Abstract
Due to multiple mutations of SARS-CoV-2, the mystery of defeating the virus is still unknown. Cardiovascular complications are one of the most concerning effects of COVID-19 recently, originating from direct and indirect mechanisms. These complications are associated with long-term Cardio-vascular diseases and can induce sudden cardiac death in both infected and recovered COVID-19 patients. The purpose of this research is to do a competitive analysis between conventional techniques with the upgraded alternative 3D bioprinting to replace the damaged portion of the myocardium. Additionally, this study focuses on the potential of 3D bioprinting to be a novel alternative. Finally, current challenges and future perspective of 3D bioprinting technique is briefly discussed.
Collapse
Affiliation(s)
- Fariya Akter
- Biotechnology Program, Department of Mathematics and Natural Sciences, Brac University, Dhaka, Bangladesh
| | - Yusha Araf
- Department of Genetic Engineering and Biotechnology, School of Life Sciences, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Iftekhar Bin Naser
- Biotechnology Program, Department of Mathematics and Natural Sciences, Brac University, Dhaka, Bangladesh
| | - Salman Khan Promon
- Department of Life Sciences, School of Environment and Life Sciences, Independent University, Bangladesh (IUB), Bashundhara, Dhaka, Bangladesh
| |
Collapse
|
5
|
Regenerating Damaged Myocardium: A Review of Stem-Cell Therapies for Heart Failure. Cells 2021; 10:cells10113125. [PMID: 34831347 PMCID: PMC8625160 DOI: 10.3390/cells10113125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease (CVD) is one of the contributing factors to more than one-third of human mortality and the leading cause of death worldwide. The death of cardiac myocyte is a fundamental pathological process in cardiac pathologies caused by various heart diseases, including myocardial infarction. Thus, strategies for replacing fibrotic tissue in the infarcted region with functional myocardium have long been a goal of cardiovascular research. This review begins by briefly discussing a variety of somatic stem- and progenitor-cell populations that were frequently studied in early investigations of regenerative myocardial therapy and then focuses primarily on pluripotent stem cells (PSCs), especially induced-pluripotent stem cells (iPSCs), which have emerged as perhaps the most promising source of cardiomyocytes for both therapeutic applications and drug testing. We also describe attempts to generate cardiomyocytes directly from cardiac fibroblasts (i.e., transdifferentiation), which, if successful, may enable the pool of endogenous cardiac fibroblasts to be used as an in-situ source of cardiomyocytes for myocardial repair.
Collapse
|
6
|
Gao Y, Pu J. Differentiation and Application of Human Pluripotent Stem Cells Derived Cardiovascular Cells for Treatment of Heart Diseases: Promises and Challenges. Front Cell Dev Biol 2021; 9:658088. [PMID: 34055788 PMCID: PMC8149736 DOI: 10.3389/fcell.2021.658088] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/25/2021] [Indexed: 12/15/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) are derived from human embryos (human embryonic stem cells) or reprogrammed from human somatic cells (human induced pluripotent stem cells). They can differentiate into cardiovascular cells, which have great potential as exogenous cell resources for restoring cardiac structure and function in patients with heart disease or heart failure. A variety of protocols have been developed to generate and expand cardiovascular cells derived from hPSCs in vitro. Precisely and spatiotemporally activating or inhibiting various pathways in hPSCs is required to obtain cardiovascular lineages with high differentiation efficiency. In this concise review, we summarize the protocols of differentiating hPSCs into cardiovascular cells, highlight their therapeutic application for treatment of cardiac diseases in large animal models, and discuss the challenges and limitations in the use of cardiac cells generated from hPSCs for a better clinical application of hPSC-based cardiac cell therapy.
Collapse
Affiliation(s)
- Yu Gao
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
7
|
Liu Y, Wang M, Liang Y, Wang C, Naruse K, Takahashi K. Treatment of Oxidative Stress with Exosomes in Myocardial Ischemia. Int J Mol Sci 2021; 22:ijms22041729. [PMID: 33572188 PMCID: PMC7915208 DOI: 10.3390/ijms22041729] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
A thrombus in a coronary artery causes ischemia, which eventually leads to myocardial infarction (MI) if not removed. However, removal generates reactive oxygen species (ROS), which causes ischemia–reperfusion (I/R) injury that damages the tissue and exacerbates the resulting MI. The mechanism of I/R injury is currently extensively understood. However, supplementation of exogenous antioxidants is ineffective against oxidative stress (OS). Enhancing the ability of endogenous antioxidants may be a more effective way to treat OS, and exosomes may play a role as targeted carriers. Exosomes are nanosized vesicles wrapped in biofilms which contain various complex RNAs and proteins. They are important intermediate carriers of intercellular communication and material exchange. In recent years, diagnosis and treatment with exosomes in cardiovascular diseases have gained considerable attention. Herein, we review the new findings of exosomes in the regulation of OS in coronary heart disease, discuss the possibility of exosomes as carriers for the targeted regulation of endogenous ROS generation, and compare the advantages of exosome therapy with those of stem-cell therapy. Finally, we explore several miRNAs found in exosomes against OS.
Collapse
|
8
|
Sareen N, Srivastava A, Dhingra S. Role of prostaglandin E2 in allogeneic mesenchymal stem cell therapy for cardiac repair. Can J Physiol Pharmacol 2021; 99:140-150. [PMID: 33559528 DOI: 10.1139/cjpp-2020-0413] [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] [Indexed: 11/22/2022]
Abstract
Ischemic heart disease is among the primary causes of cardiovascular-related deaths worldwide. Conventional treatments including surgical interventions and medical therapies aid in preventing further damage to heart muscle but are unable to provide a permanent solution. In recent years, stem cell therapy has emerged as an attractive alternative to restore damaged myocardium after myocardial injury. Allogeneic (donor-derived) mesenchymal stem cells (MSCs) have shown great promise in preclinical and clinical studies, making them the most widely accepted candidates for cardiac cell therapy. MSCs promote cardiac repair by modulating host immune system and secreting various soluble factors, of which prostaglandin E2 (PGE2) is an important one. PGE2 plays a significant role in regulating cardiac remodeling following myocardial injury. In this review, we provide an overview of allogeneic MSCs as candidates for myocardial regeneration with a focus on the role of the PGE2/cyclooxygenase-2 (COX2) pathway in mediating these effects.
Collapse
Affiliation(s)
- Niketa Sareen
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Abhay Srivastava
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sanjiv Dhingra
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
9
|
Regulation of cardiomyocyte fate plasticity: a key strategy for cardiac regeneration. Signal Transduct Target Ther 2021; 6:31. [PMID: 33500391 PMCID: PMC7838318 DOI: 10.1038/s41392-020-00413-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/11/2020] [Accepted: 10/26/2020] [Indexed: 01/14/2023] Open
Abstract
With the high morbidity and mortality rates, cardiovascular diseases have become one of the most concerning diseases worldwide. The heart of adult mammals can hardly regenerate naturally after injury because adult cardiomyocytes have already exited the cell cycle, which subseqently triggers cardiac remodeling and heart failure. Although a series of pharmacological treatments and surgical methods have been utilized to improve heart functions, they cannot replenish the massive loss of beating cardiomyocytes after injury. Here, we summarize the latest research progress in cardiac regeneration and heart repair through altering cardiomyocyte fate plasticity, which is emerging as an effective strategy to compensate for the loss of functional cardiomyocytes and improve the impaired heart functions. First, residual cardiomyocytes in damaged hearts re-enter the cell cycle to acquire the proliferative capacity by the modifications of cell cycle-related genes or regulation of growth-related signals. Additionally, non-cardiomyocytes such as cardiac fibroblasts, were shown to be reprogrammed into cardiomyocytes and thus favor the repair of damaged hearts. Moreover, pluripotent stem cells have been shown to transform into cardiomyocytes to promote heart healing after myocardial infarction (MI). Furthermore, in vitro and in vivo studies demonstrated that environmental oxygen, energy metabolism, extracellular factors, nerves, non-coding RNAs, etc. play the key regulatory functions in cardiac regeneration. These findings provide the theoretical basis of targeting cellular fate plasticity to induce cardiomyocyte proliferation or formation, and also provide the clues for stimulating heart repair after injury.
Collapse
|
10
|
Dessouki FBA, Kukreja RC, Singla DK. Stem Cell-Derived Exosomes Ameliorate Doxorubicin-Induced Muscle Toxicity through Counteracting Pyroptosis. Pharmaceuticals (Basel) 2020; 13:ph13120450. [PMID: 33316945 PMCID: PMC7764639 DOI: 10.3390/ph13120450] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/30/2020] [Accepted: 12/04/2020] [Indexed: 12/21/2022] Open
Abstract
Doxorubicin (Dox)-induced muscle toxicity (DIMT) is a common occurrence in cancer patients; however, the cause of its development and progression is not established. We tested whether inflammation-triggered cell death, “pyroptosis” plays a role in DIMT. We also examined the potential role of exosomes derived from embryonic stem cells (ES-Exos) in attenuating DIMT. C57BL/6J mice (10 ± 2 wks age) underwent the following treatments: Control (saline), Dox, Dox+ES-Exos, and Dox+MEF-Exos (mouse-embryonic fibroblast-derived exosomes, negative control). Our results demonstrated that Dox significantly reduced muscle function in mice, which was associated with a significant increase in NLRP3 inflammasome and initiation marker TLR4 as compared with controls. Pyroptosis activator, ASC, was significantly increased compared to controls with an upregulation of specific markers (caspase-1, IL-1β, and IL-18). Treatment with ES-Exos but not MEF-Exos showed a significant reduction in inflammasome and pyroptosis along with improved muscle function. Additionally, we detected a significant increase in pro-inflammatory cytokines (TNF-α and IL-6) and inflammatory M1 macrophages in Dox-treated animals. Treatment with ES-Exos decreased M1 macrophages and upregulated anti-inflammatory M2 macrophages. Furthermore, ES-Exos showed a significant reduction in muscular atrophy and fibrosis. In conclusion, these results suggest that DIMT is mediated through inflammation and pyroptosis, which is attenuated following treatment with ES-Exos.
Collapse
Affiliation(s)
- Fatima Bianca A. Dessouki
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA;
| | - Rakesh C. Kukreja
- Pauley Heart Center, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Dinender K. Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA;
- Correspondence: ; Tel.: +1-401-823-0953
| |
Collapse
|
11
|
Strategies for Cancer Immunotherapy Using Induced Pluripotency Stem Cells-Based Vaccines. Cancers (Basel) 2020; 12:cancers12123581. [PMID: 33266109 PMCID: PMC7760556 DOI: 10.3390/cancers12123581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 12/14/2022] Open
Abstract
Despite improvements in cancer therapy, metastatic solid tumors remain largely incurable. Immunotherapy has emerged as a pioneering and promising approach for cancer therapy and management, and in particular intended for advanced tumors unresponsive to current therapeutics. In cancer immunotherapy, components of the immune system are exploited to eliminate cancer cells and treat patients. The recent clinical successes of immune checkpoint blockade and chimeric antigen receptor T cell therapies represent a turning point in cancer treatment. Despite their potential success, current approaches depend on efficient tumor antigen presentation which are often inaccessible, and most tumors turn refractory to current immunotherapy. Patient-derived induced pluripotent stem cells (iPSCs) have been shown to share several characteristics with cancer (stem) cells (CSCs), eliciting a specific anti-tumoral response when injected in rodent cancer models. Indeed, artificial cellular reprogramming has been widely compared to the biogenesis of CSCs. Here, we will discuss the state-of-the-art on the potential implication of cellular reprogramming and iPSCs for the design of patient-specific immunotherapeutic strategies, debating the similarities between iPSCs and cancer cells and introducing potential strategies that could enhance the efficiency and therapeutic potential of iPSCs-based cancer vaccines.
Collapse
|
12
|
Wagner MJ, Khan M, Mohsin S. Healing the Broken Heart; The Immunomodulatory Effects of Stem Cell Therapy. Front Immunol 2020; 11:639. [PMID: 32328072 PMCID: PMC7160320 DOI: 10.3389/fimmu.2020.00639] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/20/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular Disease (CVD) is a leading cause of mortality within the United States. Current treatments being administered to patients who suffered a myocardial infarction (MI) have increased patient survival, but do not facilitate the replacement of damaged myocardium. Recent studies demonstrate that stem cell-based therapies promote myocardial repair; however, the poor engraftment of the transferred stem cell populations within the infarcted myocardium is a major limitation, regardless of the cell type. One explanation for poor cell retention is attributed to the harsh inflammatory response mounted following MI. The inflammatory response coupled to cardiac repair processes is divided into two distinct phases. The first phase is initiated during ischemic injury when necrosed myocardium releases Danger Associated Molecular Patterns (DAMPs) and chemokines/cytokines to induce the activation and recruitment of neutrophils and pro-inflammatory M1 macrophages (MΦs); in turn, facilitating necrotic tissue clearance. During the second phase, a shift from the M1 inflammatory functional phenotype to the M2 anti-inflammatory and pro-reparative functional phenotype, permits the resolution of inflammation and the establishment of tissue repair. T-regulatory cells (Tregs) are also influential in mediating the establishment of the pro-reparative phase by directly regulating M1 to M2 MΦ differentiation. Current studies suggest CD4+ T-lymphocyte populations become activated when presented with autoantigens released from the injured myocardium. The identity of the cardiac autoantigens or paracrine signaling molecules released from the ischemic tissue that directly mediate the phenotypic plasticity of T-lymphocyte populations in the post-MI heart are just beginning to be elucidated. Stem cells are enriched centers that contain a diverse paracrine secretome that can directly regulate responses within neighboring cell populations. Previous studies identify that stem cell mediated paracrine signaling can influence the phenotype and function of immune cell populations in vitro, but how stem cells directly mediate the inflammatory microenvironment of the ischemic heart is poorly characterized and is a topic of extensive investigation. In this review, we summarize the complex literature that details the inflammatory microenvironment of the ischemic heart and provide novel insights regarding how paracrine mediated signaling produced by stem cell-based therapies can regulate immune cell subsets to facilitate pro-reparative myocardial wound healing.
Collapse
Affiliation(s)
- Marcus J Wagner
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Mohsin Khan
- Center for Metabolic Disease, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Sadia Mohsin
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
13
|
Shao L, Zhang Y, Pan X, Liu B, Liang C, Zhang Y, Wang Y, Yan B, Xie W, Sun Y, Shen Z, Yu XY, Li Y. Knockout of beta-2 microglobulin enhances cardiac repair by modulating exosome imprinting and inhibiting stem cell-induced immune rejection. Cell Mol Life Sci 2020; 77:937-952. [PMID: 31312880 PMCID: PMC11104803 DOI: 10.1007/s00018-019-03220-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/26/2019] [Accepted: 07/05/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Allogeneic human umbilical mesenchymal stem cells (alloUMSC) are convenient cell source for stem cell-based therapy. However, immune rejection is a major obstacle for clinical application of alloUMSC for cardiac repair after myocardial infarction (MI). The immune rejection is due to the presence of human leukocyte antigen (HLA) class I molecule which is increased during MI. The aim of this study was to knockout HLA light chain β2-microglobulin (B2M) in UMSC to enhance stem cell engraftment and survival after transplantation. METHODS AND RESULTS We developed an innovative strategy using CRISPR/Cas9 to generate UMSC with B2M deletion (B2M-UMSC). AlloUMSC injection induced CD8+ T cell-mediated immune rejection in immune competent rats, whereas no CD8+ T cell-mediated killing against B2M-UMSC was observed even when the cells were treated with IFN-γ. Moreover, we demonstrate that UMSC-derived exosomes can inhibit cardiac fibrosis and restore cardiac function, and exosomes derived from B2M-UMSC are more efficient than those derived from UMSC, indicating that the beneficial effect of exosomes can be enhanced by modulating exosome's imprinting. Mechanistically, microRNA sequencing identifies miR-24 as a major component of the exosomes from B2M-UMSCs. Bioinformatics analysis identifies Bim as a putative target of miR-24. Loss-of-function studies at the cellular level and gain-of-function approaches in exosomes show that the beneficial effects of B2M-UMSCs are mediated by the exosome/miR-24/Bim pathway. CONCLUSION Our findings demonstrate that modulation of exosome's imprinting via B2M knockout is an efficient strategy to prevent the immune rejection of alloUMSCs. This study paved the way to the development of new strategies for tissue repair and regeneration without the need for HLA matching.
Collapse
Affiliation(s)
- Lianbo Shao
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yu Zhang
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Xiangbin Pan
- Department of Cardiac Surgery, Fuwai Hospital, Beijing, 100037, People's Republic of China
| | - Bin Liu
- Department of Cardiology, The First Hospital of Jilin University, Changchun, 130041, Jilin, People's Republic of China
| | - Chun Liang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Yuqing Zhang
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yanli Wang
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Bing Yan
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Wenping Xie
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yi Sun
- Fuwai Yunnan Cardiovascular Hospital, Kunming, 650302, Yunnan, People's Republic of China
| | - Zhenya Shen
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Xi-Yong Yu
- Guangzhou Medical University, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Yangxin Li
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China.
| |
Collapse
|
14
|
Abstract
The investment of nearly 2 decades of clinical investigation into cardiac cell therapy has yet to change cardiovascular practice. Recent insights into the mechanism of cardiac regeneration help explain these results and provide important context in which we can develop next-generation therapies. Non-contractile cells such as bone marrow or adult heart derivatives neither engraft long-term nor induce new muscle formation. Correspondingly, these cells offer little functional benefit to infarct patients. In contrast, preclinical data indicate that transplantation of bona fide cardiomyocytes derived from pluripotent stem cells induces direct remuscularization. This new myocardium beats synchronously with the host heart and induces substantial contractile benefits in macaque monkeys, suggesting that regeneration of contractile myocardium is required to fully recover function. Through a review of the preclinical and clinical trials of cardiac cell therapy, distinguishing the primary mechanism of benefit as either contractile or non-contractile helps appreciate the barriers to cardiac repair and establishes a rational path to optimizing therapeutic benefit.
Collapse
Affiliation(s)
- Kenta Nakamura
- Institute for Stem Cell and Regenerative Medicine, University of Washington
- Center for Cardiovascular Biology, University of Washington
- Department of Medicine/Cardiology, University of Washington
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington
- Center for Cardiovascular Biology, University of Washington
- Department of Medicine/Cardiology, University of Washington
- Department of Pathology, University of Washington
- Department of Bioengineering, University of Washington
| |
Collapse
|
15
|
Kot M, Baj-Krzyworzeka M, Szatanek R, Musiał-Wysocka A, Suda-Szczurek M, Majka M. The Importance of HLA Assessment in "Off-the-Shelf" Allogeneic Mesenchymal Stem Cells Based-Therapies. Int J Mol Sci 2019; 20:E5680. [PMID: 31766164 PMCID: PMC6888380 DOI: 10.3390/ijms20225680] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
The need for more effective therapies of chronic and acute diseases has led to the attempts of developing more adequate and less invasive treatment methods. Regenerative medicine relies mainly on the therapeutic potential of stem cells. Mesenchymal stem cells (MSCs), due to their immunosuppressive properties and tissue repair abilities, seem to be an ideal tool for cell-based therapies. Taking into account all available sources of MSCs, perinatal tissues become an attractive source of allogeneic MSCs. The allogeneic MSCs provide "off-the-shelf" cellular therapy, however, their allogenicity may be viewed as a limitation for their use. Moreover, some evidence suggests that MSCs are not as immune-privileged as it was previously reported. Therefore, understanding their interactions with the recipient's immune system is crucial for their successful clinical application. In this review, we discuss both autologous and allogeneic application of MSCs, focusing on current approaches to allogeneic MSCs therapies, with a particular interest in the role of human leukocyte antigens (HLA) and HLA-matching in allogeneic MSCs transplantation. Importantly, the evidence from the currently completed and ongoing clinical trials demonstrates that allogeneic MSCs transplantation is safe and seems to cause no major side-effects to the patient. These findings strongly support the case for MSCs efficacy in treatment of a variety of diseases and their use as an "off-the-shelf" medical product.
Collapse
Affiliation(s)
- Marta Kot
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| | - Monika Baj-Krzyworzeka
- Department of Clinical Immunology, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.B.-K.); (R.S.)
| | - Rafał Szatanek
- Department of Clinical Immunology, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.B.-K.); (R.S.)
| | - Aleksandra Musiał-Wysocka
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| | - Magdalena Suda-Szczurek
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| | - Marcin Majka
- Department of Transplantation, Faculty of Medicine, Medical College, Jagiellonian University, Wielicka 265, 30-663 Kraków, Poland; (M.K.); (A.M.-W.); (M.S.-S.)
| |
Collapse
|
16
|
Exosome Treatment Enhances Anti-Inflammatory M2 Macrophages and Reduces Inflammation-Induced Pyroptosis in Doxorubicin-Induced Cardiomyopathy. Cells 2019; 8:cells8101224. [PMID: 31600901 PMCID: PMC6830113 DOI: 10.3390/cells8101224] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/18/2019] [Accepted: 09/26/2019] [Indexed: 12/20/2022] Open
Abstract
Doxorubicin (Dox) is an effective antineoplastic agent used to treat cancers, but its use is limited as Dox induces adverse cardiotoxic effects. Dox-induced cardiotoxicity (DIC) can lead to heart failure and death. There is no study that investigates whether embryonic stem cell-derived exosomes (ES-Exos) in DIC can attenuate inflammation-induced pyroptosis, pro-inflammatory M1 macrophages, inflammatory cell signaling, and adverse cardiac remodeling. For this purpose, we transplanted ES-Exos and compared with ES-cells (ESCs) to examine pyroptosis, inflammation, cell signaling, adverse cardiac remodeling, and their influence on DIC induced cardiac dysfunction. Therefore, we used C57BL/6J mice ages 10 ± 2 weeks and divided them into four groups (n = 6–8/group): Control, Dox, Dox + ESCs, and Dox + ES-Exos. Our data shows that the Dox treatment significantly increased expression of inflammasome markers (TLR4 and NLRP3), pyroptotic markers (caspase-1, IL1-β, and IL-18), cell signaling proteins (MyD88, p-P38, and p-JNK), pro-inflammatory M1 macrophages, and TNF-α cytokine. This increased pyroptosis, inflammation, and cell signaling proteins were inhibited with ES-Exos or ESCs. Moreover, ES-Exos or ESCs increased M2 macrophages and anti-inflammatory cytokine, IL-10. Additionally, ES-Exos or ESCs treatment inhibited significantly cytoplasmic vacuolization, myofibril loss, hypertrophy, and improved heart function. In conclusion, for the first time we demonstrated that Dox-induced pyroptosis and cardiac remodeling are ameliorated by ES-Exos or ESCs.
Collapse
|
17
|
Winnier GE, Valenzuela N, Peters-Hall J, Kellner J, Alt C, Alt EU. Isolation of adipose tissue derived regenerative cells from human subcutaneous tissue with or without the use of an enzymatic reagent. PLoS One 2019; 14:e0221457. [PMID: 31479463 PMCID: PMC6719836 DOI: 10.1371/journal.pone.0221457] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 08/07/2019] [Indexed: 12/15/2022] Open
Abstract
Freshly isolated, uncultured, autologous adipose derived regenerative cells (ADRCs) have emerged as a promising tool for regenerative cell therapy. The Transpose RT system (InGeneron, Inc., Houston, TX, USA) is a system for isolating ADRCs from adipose tissue, commercially available in Europe as a CE-marked medical device and under clinical evaluation in the United States. This system makes use of the proprietary, enzymatic Matrase Reagent for isolating cells. The present study addressed the question whether the use of Matrase Reagent influences cell yield, cell viability, live cell yield, biological characteristics, physiological functions or structural properties of the ADRCs in final cell suspension. Identical samples of subcutaneous adipose tissue from 12 subjects undergoing elective lipoplasty were processed either with or without the use of Matrase Reagent. Then, characteristics of the ADRCs in the respective final cell suspensions were evaluated. Compared to non-enzymatic isolation, enzymatic isolation resulted in approximately twelve times higher mean cell yield (i.e., numbers of viable cells/ml lipoaspirate) and approximately 16 times more colony forming units. Despite these differences, cells isolated from lipoaspirate both with and without the use of Matrase Reagent were independently able to differentiate into cells of all three germ layers. This indicates that biological characteristics, physiological functions or structural properties relevant for the intended use were not altered or induced using Matrase Reagent. A comprehensive literature review demonstrated that isolation of ADRCs from lipoaspirate using the Transpose RT system and the Matrase Reagent results in the highest viable cell yield among published data regarding isolation of ADRCs from lipoaspirate.
Collapse
Affiliation(s)
| | | | | | | | | | - Eckhard U. Alt
- InGeneron, Inc., Houston, TX, United States of America
- Heart and Vascular Institute, Department of Medicine, Tulane University Health Science Center, New Orleans, LA, United States of America
- Sanford Health, University of South Dakota, Sioux Falls, SD, United States of America
- Isar Klinikum Munich, Munich, Germany
- * E-mail: ,
| |
Collapse
|
18
|
Tavakoli Dargani Z, Singla DK. Embryonic stem cell-derived exosomes inhibit doxorubicin-induced TLR4-NLRP3-mediated cell death-pyroptosis. Am J Physiol Heart Circ Physiol 2019; 317:H460-H471. [PMID: 31172809 PMCID: PMC6732475 DOI: 10.1152/ajpheart.00056.2019] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/04/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023]
Abstract
Doxorubicin (Dox)-induced cardiac side effects are regulated through increased oxidative stress and apoptosis. However, it remains unknown whether Dox induces the specific inflammatory-mediated form of cell death called pyroptosis. The current study is undertaken to determine whether Dox induces pyroptosis in an in vitro model and to test the potential of exosomes derived from embryonic stem cells (ES-Exos) in inhibiting pyroptosis. H9c2 cells were exposed to Dox to generate pyroptosis and then subsequently treated with exosomes to investigate the protective effects of ES-Exos. Mouse embryonic fibroblast-exosomes (MEF-Exos) were used as a cell line control. We confirmed pyroptosis by analyzing the presence of Toll-like receptor 4 (TLR4)-pyrin domain containing-3 (NLRP3) inflammasome that initiates pyroptosis, which was further confirmed with pyroptotic markers caspase-1, IL-1β, caspase-11, and gasdermin-D. The presence of inflammation was confirmed for proinflammatory cytokines, TNF-α, and IL-6. Our data show that Dox exposure significantly (P < 0.05) increases expression of TLR4, NLRP3, pyroptotic markers (caspase-1, IL-1β, caspase-11, and gasdermin-D), and proinflammatory cytokines (TNF-α and IL-6) in H9c2 cells. The increased expression of inflammasome, pyroptosis, and inflammation was significantly (P < 0.05) inhibited by ES-Exos. Interestingly, our cell line control, MEF-Exos, did not show any protective effects. Furthermore, our cytokine array data suggest increased anti-inflammatory (IL-4, IL-9, and IL-13) and decreased proinflammatory cytokines (Fas ligand, IL-12, and TNF-α) in ES-Exos, suggesting that anti-inflammatory cytokines might be mediating the protective effects of ES-Exos. In conclusion, our data show that Dox induces pyroptotic cell death in the H9c2 cell culture model and is attenuated via treatment with ES-Exos.NEW & NOTEWORTHY Doxorubicin (Dox)-induced cardiotoxicity is mediated through increased oxidative stress, apoptosis, and necrosis. We report for the first time as per the best of our knowledge that Dox initiates Toll-like receptor 4 and pyrin domain containing-3 inflammasome formation and induces caspase-1-mediated inflammatory pyroptotic cell death in H9c2 cells. Moreover, we establish that inflammation and pyroptosis is inhibited by embryonic stem cell-derived exosomes that could be used as a future therapeutic option to treat Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zahra Tavakoli Dargani
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| |
Collapse
|
19
|
Nejadnik H, Tseng J, Daldrup-Link H. Magnetic resonance imaging of stem cell-macrophage interactions with ferumoxytol and ferumoxytol-derived nanoparticles. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1552. [PMID: 30734542 PMCID: PMC6579657 DOI: 10.1002/wnan.1552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/13/2018] [Accepted: 12/19/2018] [Indexed: 01/07/2023]
Abstract
"Off the shelf" allogeneic stem cell transplants and stem cell nano-composites are being used for the treatment of degenerative bone diseases. However, major and minor histocompatibility antigens of therapeutic cell transplants can be recognized as foreign and lead to their rejection by the host immune system. If a host immune response is identified within the first week post-transplant, immune modulating therapies could be applied to prevent graft failure and support engraftment. Ferumoxytol (Feraheme™) is an FDA approved iron oxide nanoparticle preparation for the treatment of anemia in patients. Ferumoxytol can be used "off label" as an magnetic resonance (MR) contrast agent, as these nanoparticles provide measurable signal changes on magnetic resonance imaging (MRI). In this focused review article, we will discuss three methods to localize and identify innate immune responses to stem cell transplants using ferumoxytol-enhanced MRI, which are based on tracking stem cells, tracking macrophages or detecting mediators of cell death: (a) monitor MRI signal changes of ferumoxytol-labeled stem cells in the presence or absence of innate immune responses, (b) monitor influx of ferumoxytol-labeled macrophages into stem cell implants, and (c) monitor apoptosis of stem cell implants with caspase-3 activatable nanoparticles. These techniques can detect transplant failure at an early stage, when immune-modulating interventions can potentially preserve the viability of the cell transplants and thereby improve bone and cartilage repair outcomes. Approaches 1 and 2 are immediately translatable to clinical practice. This article is categorized under: Diagnostic Tools > in vivo Nanodiagnostics and Imaging Nanotechnology Approaches to Biology > Cells at the Nanoscale Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Hossein Nejadnik
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Jessica Tseng
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| | - Heike Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California
| |
Collapse
|
20
|
Feridooni T, Pasumarthi KBS. Fractionation of embryonic cardiac progenitor cells and evaluation of their differentiation potential. Differentiation 2018; 105:1-13. [PMID: 30530197 DOI: 10.1016/j.diff.2018.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/24/2018] [Accepted: 11/22/2018] [Indexed: 12/19/2022]
Abstract
Mid-gestation mouse ventricles (E11.5) contain a larger number of Nkx2.5+ cardiac progenitor cells (CPCs). The proliferation rates are consistently higher in CPCs compared to myocyte population of developing ventricles. Recent studies suggested that CPCs are an ideal donor cell type for replacing damaged tissue in diseased hearts. Thus, the ability to isolate and expand CPCs from embryos or stem cell cultures could be useful for cell fate studies and regenerative therapies. Since embryonic CPCs possess fewer mitochondria compared to cardiomyocytes, we reasoned that CPCs can be fractionated using a fluorescent mitochondrial membrane potential dye (TMRM) and these cells may retain cardiomyogenic potential even in the absence of cardiomyocytes (CMs). FACS sorting of TMRM stained embryonic ventricular cells indicated that over 99% of cells in TMRM high fraction stained positive for sarcomeric myosin (MF20) and all of them expressed Nkx2.5. Although majority of cells present in TMRM low fraction expressed Nkx2.5, very few cells (~1%) stained positive for MF20. Further culturing of TMRM low cells over a period of 48 h showed a progressive increase in MF20 positive cells. Additional analyses revealed that MF20 negative cells in TMRM low fraction do not express markers for endothelial cells (vWF, CD31) or smooth muscle cells (SM myosin). Treatment of TMRM low cells with known cardiogenic factors DMSO and dynorphin B significantly increased the percentage of MF20+ cells compared to untreated cultures. Collectively, these studies suggest that embryonic CPCs can be separated as a TMRM low fraction and their differentiation potential can be enhanced by exogenous addition of known cardiomyogenic factors.
Collapse
Affiliation(s)
- Tiam Feridooni
- Department of Pharmacology, Dalhousie University, Sir Charles Tupper Building, 5850 College Street, P.O. Box 15000, Halifax, Nova Scotia, Canada B3H 4R2
| | - Kishore B S Pasumarthi
- Department of Pharmacology, Dalhousie University, Sir Charles Tupper Building, 5850 College Street, P.O. Box 15000, Halifax, Nova Scotia, Canada B3H 4R2.
| |
Collapse
|
21
|
Rodrigues ICP, Kaasi A, Maciel Filho R, Jardini AL, Gabriel LP. Cardiac tissue engineering: current state-of-the-art materials, cells and tissue formation. ACTA ACUST UNITED AC 2018; 16:eRB4538. [PMID: 30281764 PMCID: PMC6178861 DOI: 10.1590/s1679-45082018rb4538] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 07/24/2018] [Indexed: 12/23/2022]
Abstract
Cardiovascular diseases are the major cause of death worldwide. The heart has limited capacity of regeneration, therefore, transplantation is the only solution in some cases despite presenting many disadvantages. Tissue engineering has been considered the ideal strategy for regenerative medicine in cardiology. It is an interdisciplinary field combining many techniques that aim to maintain, regenerate or replace a tissue or organ. The main approach of cardiac tissue engineering is to create cardiac grafts, either whole heart substitutes or tissues that can be efficiently implanted in the organism, regenerating the tissue and giving rise to a fully functional heart, without causing side effects, such as immunogenicity. In this review, we systematically present and compare the techniques that have drawn the most attention in this field and that generally have focused on four important issues: the scaffold material selection, the scaffold material production, cellular selection and in vitro cell culture. Many studies used several techniques that are herein presented, including biopolymers, decellularization and bioreactors, and made significant advances, either seeking a graft or an entire bioartificial heart. However, much work remains to better understand and improve existing techniques, to develop robust, efficient and efficacious methods.
Collapse
Affiliation(s)
| | | | - Rubens Maciel Filho
- Instituto Nacional de Ciência e Tecnologia em Biofabricação, Campinas, SP, Brazil
| | - André Luiz Jardini
- Instituto Nacional de Ciência e Tecnologia em Biofabricação, Campinas, SP, Brazil
| | | |
Collapse
|
22
|
Macrin D, Joseph JP, Pillai AA, Devi A. Eminent Sources of Adult Mesenchymal Stem Cells and Their Therapeutic Imminence. Stem Cell Rev Rep 2018; 13:741-756. [PMID: 28812219 DOI: 10.1007/s12015-017-9759-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the recent times, stem cell biology has garnered the attention of the scientific fraternity and the general public alike due to the immense therapeutic potential that it holds in the field of regenerative medicine. A breakthrough in this direction came with the isolation of stem cells from human embryo and their differentiation into cell types of all three germ layers. However, the isolation of mesenchymal stem cells from adult tissues proved to be advantageous over embryonic stem cells due to the ethical and immunological naivety. Mesenchymal Stem Cells (MSCs) isolated from the bone marrow were found to differentiate into multiple cell lineages with the help of appropriate differentiation factors. Furthermore, other sources of stem cells including adipose tissue, dental pulp, and breast milk have been identified. Newer sources of stem cells have been emerging recently and their clinical applications are also being studied. In this review, we examine the eminent sources of Mesenchymal Stem Cells (MSCs), their immunophenotypes, and therapeutic imminence.
Collapse
Affiliation(s)
- Dannie Macrin
- Department of Genetic Engineering, SRM University, Kattankulathur, Tamil Nadu, India
| | - Joel P Joseph
- Department of Genetic Engineering, SRM University, Kattankulathur, Tamil Nadu, India
| | | | - Arikketh Devi
- Department of Genetic Engineering, SRM University, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
23
|
Human embryonic stem cell-derived cardiomyocytes restore function in infarcted hearts of non-human primates. Nat Biotechnol 2018; 36:597-605. [PMID: 29969440 PMCID: PMC6329375 DOI: 10.1038/nbt.4162] [Citation(s) in RCA: 411] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 05/06/2018] [Indexed: 02/07/2023]
Abstract
Pluripotent stem cell–derived cardiomyocyte grafts can remuscularize substantial amounts of infarcted myocardium and beat in synchrony with the heart, but in some settings cause ventricular arrhythmias. It is unknown whether human cardiomyocytes can restore cardiac function in a physiologically relevant large animal model. Here we show that transplantation of ~750 million cryopreserved human embryonic stem cell–derived cardiomyocytes (hESC-CMs) enhances cardiac function in macaque monkeys with large myocardial infarctions. One month after hESC-CM transplantation, global left ventricular ejection fraction improved 10.6±0.9% vs. 2.5±0.8% in controls, and by 3 months there was an additional 12.4% improvement in treated vs. a 3.5% decline in controls. Grafts averaged 11.6% of infarct size, formed electromechanical junctions with the host heart and by 3 months contained ~99% ventricular myocytes. A subset of animals experienced graft-associated ventricular arrhythmias, shown by electrical mapping to originate from a point-source acting as an ectopic pacemaker. Our data demonstrate that remuscularization of the infarcted macaque heart with human myocardium provides durable improvement in left ventricular function.
Collapse
|
24
|
Li K, Chan CT, Nejadnik H, Lenkov OD, Wolterman C, Paulmurugan R, Yang H, Gambhir SS, Daldrup-Link HE. Ferumoxytol-based Dual-modality Imaging Probe for Detection of Stem Cell Transplant Rejection. Nanotheranostics 2018; 2:306-319. [PMID: 29977742 PMCID: PMC6030766 DOI: 10.7150/ntno.26389] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/14/2018] [Indexed: 12/24/2022] Open
Abstract
Purpose: Stem cell transplants are an effective approach to repair large bone defects. However, comprehensive techniques to monitor the fate of transplanted stem cells in vivo are lacking. Such strategies would enable corrective interventions at an early stage and greatly benefit the development of more successful tissue regeneration approaches. In this study, we designed and synthesized a dual-modality imaging probe (Feru-AFC) that can simultaneously localize transplanted stem cells and diagnose immune rejection-induced apoptosis at an early stage in vivo. Methods: We used a customized caspase-3 cleavable peptide-dye conjugate to modify the surface of clinically approved ferumoxytol nanoparticles (NPs) to generate the dual-modality imaging probe with fluorescence "light-up" feature. We labeled both mouse mesenchymal stem cells (mMSCs, matched) and pig mesenchymal stem cells (pMSCs, mismatched) with the probe and transplanted the labeled cells with biocompatible scaffold at the calvarial defects in mice. We then employed intravital microscopy (IVM) and magnetic resonance imaging (MRI) to investigate the localization, engraftment, and viability of matched and mismatched stem cells, followed by histological analyses to evaluate the results obtained from in vivo studies. Results: The Feru-AFC NPs showed good cellular uptake efficiency in the presence of lipofectin without cytotoxicity to mMSCs and pMSCs. The fluorescence of Feru-AFC NPs was turned on inside apoptotic cells due to the cleavage of peptide by activated caspase-3 and subsequent release of fluorescence dye molecules. Upon transplantation at the calvarial defects in mice, the intense fluorescence from the cleaved Feru-AFC NPs in apoptotic pMSCs was observed with a concomitant decrease in the overall cell number from days 1 to 6. In contrast, the Feru-AFC NP-treated mMSCs exhibited minimum fluorescence and the cell number also remained similar. Furthermore, in vivo MRI of the Feru-AFC NP-treated mMSC and pMSCs transplants could clearly indicate the localization of matched and mismatched cells, respectively. Conclusions: We successfully developed a dual-modality imaging probe for evaluation of the localization and viability of transplanted stem cells in mouse calvarial defects. Using ferumoxytol NPs as the platform, our Feru-AFC NPs are superparamagnetic and display a fluorescence "light-up" signature upon exposure to activated caspase-3. The results show that the probe is a promising tool for long-term stem cell tracking through MRI and early diagnosis of immune rejection-induced apoptosis through longitudinal fluorescence imaging.
Collapse
Affiliation(s)
- Kai Li
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305.,Institute of Materials Research and Engineering, ASTAR, Singapore, 138634
| | - Carmel T Chan
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Hossein Nejadnik
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Olga D Lenkov
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Cody Wolterman
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Ramasamy Paulmurugan
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Huaxiao Yang
- Stanford Cardiovascular Institute, Stanford, CA 94305
| | - Sanjiv Sam Gambhir
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| | - Heike E Daldrup-Link
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, CA 94305
| |
Collapse
|
25
|
Tachibana A. [6. Application of MRI Technology to the Regenerative Medicine in Heart Disease Using Induced Pluripotent Stem Cells]. Nihon Hoshasen Gijutsu Gakkai Zasshi 2018; 74:491-498. [PMID: 29780049 DOI: 10.6009/jjrt.2018_jsrt_74.5.491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
26
|
HLA and Histo-Blood Group Antigen Expression in Human Pluripotent Stem Cells and their Derivatives. Sci Rep 2017; 7:13072. [PMID: 29026098 PMCID: PMC5638960 DOI: 10.1038/s41598-017-12231-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/06/2017] [Indexed: 01/03/2023] Open
Abstract
One prerequisite for a successful clinical outcome of human pluripotent stem cell (hPSC) based therapies is immune compatibility between grafted cells/tissue and recipient. This study explores immune determinants of human embryonic stem cell lines (hESC) and induced human pluripotent stem cell (hiPSC) lines and hepatocyte- and cardiomyocyte-like cells derived from these cells. HLA class I was expressed on all pluripotent hPSC lines which upon differentiation into hepatocyte-like cells was considerably reduced in contrast to cardiomyocyte-like cells which retained class I antigens. No HLA class II antigens were found in the pluripotent or differentiated cells. Histo-blood group carbohydrate antigens SSEA-3/SSEA-4/SSEA-5, Globo H, A, Lex/Ley and sialyl-lactotetra were expressed on all hPSC lines. Blood group AB(O)H antigen expression was in accordance with ABO genotype. Interestingly, only a subpopulation of A1O1 cells expressed A. During differentiation of hPSC, some histo-blood group antigens showed congruent alteration patterns while expression of other antigens differed between the cell lines. No systematic difference in the hPSC cell surface tissue antigen expression was detected. In conclusion, hPSC and their derivatives express cell surface antigens that may cause an immune rejection. Furthermore, tissue antigen expression must be established for each individual stem cell line prior to clinical application.
Collapse
|
27
|
Didié M, Galla S, Muppala V, Dressel R, Zimmermann WH. Immunological Properties of Murine Parthenogenetic Stem Cell-Derived Cardiomyocytes and Engineered Heart Muscle. Front Immunol 2017; 8:955. [PMID: 28855904 PMCID: PMC5557729 DOI: 10.3389/fimmu.2017.00955] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/26/2017] [Indexed: 12/27/2022] Open
Abstract
Pluripotent parthenogenetic stem cells (pSCs) can be derived by pharmacological activation of unfertilized oocytes. Homozygosity of the major histocompatibility complex (MHC) in pSCs makes them an attractive cell source for applications in allogeneic tissue repair. This was recently demonstrated for pSC-based tissue-engineered heart repair. A detailed analysis of immunological properties of pSC-derived cardiomyocytes and engineered heart muscle (EHM) thereof is, however, lacking. The aim of this study was to determine baseline and cytokine-inducible MHC class I and MHC class II as well as programmed death ligand-1 (PDL-1) and co-stimulatory protein (CD40, CD80, CD86) expression in pSC-derived cardiomyocytes and pSC-EHM in vitro and in vivo. Cardiomyocytes from an MHC-homologous (H2d/d) pSC-line were enriched to ~90% by making use of a recently developed cardiomyocyte-specific genetic selection protocol. MHC class I and MHC class II expression in cardiomyocytes could only be observed after stimulation with interferon gamma (IFN-γ). PDL-1 was markedly upregulated under IFN-γ. CD40, CD80, and CD86 were expressed at low levels and not upregulated by IFN-γ. EHM constructed from H2d/d cardiomyocytes expressed similarly low levels of MHC class I, MHC class II, and costimulatory molecules under basal conditions. However, in EHM only MHC class I, but not MHC class II, molecules were upregulated after IFN-γ-stimulation. We next employed a cocultivation system with MHC-matched and MHC-mismatched splenocytes and T-cells to analyze the immune stimulatory properties of EHMs. Despite MHC-mismatched conditions, EHM did not induce splenocyte or T-cell proliferation in vitro. To evaluate the immunogenicity of pSC-derived cardiomyocytes in vivo, we implanted pSC-derived embryoid bodies after elimination of non-cardiomyocytes (cardiac bodies) under the kidney capsules of MHC-matched and -mismatched mice. Spontaneous beating of cardiac bodies could be observed for 28 days in the matched and for 7 days in the mismatched conditions. Teratomas formed after 28 days only in the MHC-matched conditions. Immunohistochemistry revealed single clusters of CD3-positive cells in the border zone of the implant in the mismatched conditions with few CD3-positive cells infiltrating the implant. Taken together, MHC-matched pSC-cardiomyocyte allografts show little immune cell activation, offering an explanation for the observed long-term retention of pSC-EHM allografts in the absence of immunosuppression.
Collapse
Affiliation(s)
- Michael Didié
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Satish Galla
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Vijayakumar Muppala
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
| | - Ralf Dressel
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.,Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| |
Collapse
|
28
|
Gröschel C, Hübscher D, Nolte J, Monecke S, Sasse A, Elsner L, Paulus W, Trenkwalder C, Polić B, Mansouri A, Guan K, Dressel R. Efficient Killing of Murine Pluripotent Stem Cells by Natural Killer (NK) Cells Requires Activation by Cytokines and Partly Depends on the Activating NK Receptor NKG2D. Front Immunol 2017; 8:870. [PMID: 28890717 PMCID: PMC5582315 DOI: 10.3389/fimmu.2017.00870] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 07/10/2017] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells play an important role as cytotoxic effector cells, which scan the organism for infected or tumorigenic cells. Conflicting data have been published whether NK cells can also kill allogeneic or even autologous pluripotent stem cells (PSCs) and which receptors are involved. A clarification of this question is relevant since an activity of NK cells against PSCs could reduce the risk of teratoma growth after transplantation of PSC-derived grafts. Therefore, the hypothesis has been tested that the activity of NK cells against PSCs depends on cytokine activation and specifically on the activating NK receptor NKG2D. It is shown that a subcutaneous injection of autologous iPSCs failed to activate NK cells against these iPSCs and can give rise to teratomas. In agreement with this result, several PSC lines, including two iPSC, two embryonic stem cell (ESC), and two so-called multipotent adult germline stem cell (maGSC) lines, were largely resistant against resting NK cells although differences in killing were found at low level. All PSC lines were killed by interleukin (IL)-2-activated NK cells, and maGSCs were better killed than the other PSC types. The PSCs expressed ligands of the activating NK receptor NKG2D and NKG2D-deficient NK cells from Klrk1-/- mice were impaired in their cytotoxic activity against PSCs. The low-cytotoxic activity of resting NK cells was almost completely dependent on NKG2D. The cytotoxic activity of IL-2-activated NKG2D-deficient NK cells against PSCs was reduced, indicating that also other activating receptors on cytokine-activated NK cells must be engaged by ligands on PSCs. Thus, NKG2D is an important activating receptor involved in killing of murine PSCs. However, NK cells need to be activated by cytokines before they efficiently target PSCs and then also other NK receptors become relevant. These features of NK cells might be relevant for transplantation of PSC-derived grafts since NK cells have the capability to kill undifferentiated cells, which might be present in grafts in trace amounts.
Collapse
Affiliation(s)
- Carina Gröschel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Göttingen, Germany
| | - Daniela Hübscher
- DZHK (German Center for Cardiovascular Research), Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Jessica Nolte
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Sebastian Monecke
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Göttingen, Germany
| | - André Sasse
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Leslie Elsner
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Walter Paulus
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Bojan Polić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Ahmed Mansouri
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany.,Department of Molecular Cell Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Kaomei Guan
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,Institute of Pharmacology and Toxicology, Technische Universität Dresden, Dresden, Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Göttingen, Germany
| |
Collapse
|
29
|
Rao KS, Spees JL. Harnessing Epicardial Progenitor Cells and Their Derivatives for Rescue and Repair of Cardiac Tissue After Myocardial Infarction. ACTA ACUST UNITED AC 2017; 3:149-158. [PMID: 29057207 DOI: 10.1007/s40610-017-0066-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Ischemic heart disease and stroke lead to the greatest number of deaths worldwide. Despite decreased time to intervention and improvements in the standard of care, 1 out of 5 patients that survive a myocardial infarction (MI) still face long-term chronic heart failure and a 5-year mortality rate of about 50%. Based on their multi-potency for differentiation and paracrine activity, epicardial cells and their derivatives have potential to rescue jeopardized tissue and/or promote cardiac regeneration. Here we review the diagnosis and treatment of MI, basic epicardial cell biology, and potential treatment strategies designed to harness the reparative properties of epicardial cells. RECENT FINDINGS During cardiac development, epicardial cells covering the surface of the heart generate migratory progenitor cells that contribute to the coronary vasculature and the interstitial fibroblasts. Epicardial cells also produce paracrine signals required for myocardial expansion and cardiac growth. In adults with myocardial infarction, epicardial cells and their derivatives provide paracrine factors that affect myocardial remodeling and repair. At present, the intrinsic mechanisms and extrinsic signals that regulate epicardial cell fate and paracrine activity in adults remain poorly understood. SUMMARY Human diseases that result in heart failure due to negative remodeling or extensive loss of viable cardiac tissue require new, effective treatments. Improved understanding of epicardial cell function(s) and epicardial-mediated secretion of growth factors, cytokines and hormones during cardiac growth, homeostasis and injury may lead to new ways to treat patients with myocardial infarction.
Collapse
Affiliation(s)
- Krithika S Rao
- Department of Medicine, Stem Cell Core, University of Vermont, Colchester, VT 05446
- Cardiovascular Research Institute, University of Vermont, Colchester, VT 05446
| | - Jeffrey L Spees
- Department of Medicine, Stem Cell Core, University of Vermont, Colchester, VT 05446
- Cardiovascular Research Institute, University of Vermont, Colchester, VT 05446
| |
Collapse
|
30
|
Abstract
Human embryonic stem cells (hESCs) can undergo unlimited self-renewal and differentiate into all cell types in human body, and therefore hold great potential for cell therapy of currently incurable diseases including neural degenerative diseases, heart failure, and macular degeneration. This potential is further underscored by the promising safety and efficacy data from the ongoing clinical trials of hESC-based therapy of macular degeneration. However, one main challenge for the clinical application of hESC-based therapy is the allogeneic immune rejection of hESC-derived cells by the recipient. The breakthrough of the technology to generate autologous-induced pluripotent stem cells (iPSCs) by nuclear reprogramming of patient’s somatic cells raised the possibility that autologous iPSC-derived cells can be transplanted into the patients without the concern of immune rejection. However, accumulating data indicate that certain iPSC-derived cells can be immunogenic. In addition, the genomic instability associated with iPSCs raises additional safety concern to use iPSC-derived cells in human cell therapy. In this review, we will discuss the mechanism underlying the immunogenicity of the pluripotent stem cells and recent progress in developing immune tolerance strategies of human pluripotent stem cell (hPSC)-derived allografts. The successful development of safe and effective immune tolerance strategy will greatly facilitate the clinical development of hPSC-based cell therapy.
Collapse
Affiliation(s)
- Xin Liu
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Wenjuan Li
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuemei Fu
- The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yang Xu
- Center for Regenerative and Translational Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
31
|
Daldrup-Link HE, Chan C, Lenkov O, Taghavigarmestani S, Nazekati T, Nejadnik H, Chapelin F, Khurana A, Tong X, Yang F, Pisani L, Longaker M, Gambhir SS. Detection of Stem Cell Transplant Rejection with Ferumoxytol MR Imaging: Correlation of MR Imaging Findings with Those at Intravital Microscopy. Radiology 2017; 284:495-507. [PMID: 28128708 DOI: 10.1148/radiol.2017161139] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Purpose To determine whether endogenous labeling of macrophages with clinically applicable nanoparticles enables noninvasive detection of innate immune responses to stem cell transplants with magnetic resonance (MR) imaging. Materials and Methods Work with human stem cells was approved by the institutional review board and the stem cell research oversight committee, and animal experiments were approved by the administrative panel on laboratory animal care. Nine immunocompetent Sprague-Dawley rats received intravenous injection of ferumoxytol, and 18 Jax C57BL/6-Tg (Csf1r-EGFP-NGFR/FKBP1A/TNFRSF6) 2Bck/J mice received rhodamine-conjugated ferumoxytol. Then, 48 hours later, immune-matched or mismatched stem cells were implanted into osteochondral defects of the knee joints of experimental rats and calvarial defects of Jax mice. All animals underwent serial MR imaging and intravital microscopy (IVM) up to 4 weeks after surgery. Macrophages of Jax C57BL/6-Tg (Csf1r-EGFP-NGFR/FKBP1A/TNFRSF6) 2Bck/J mice express enhanced green fluorescent protein (GFP), which enables in vivo correlation of ferumoxytol enhancement at MR imaging with macrophage quantities at IVM. All quantitative data were compared between experimental groups by using a mixed linear model and t tests. Results Immune-mismatched stem cell implants demonstrated stronger ferumoxytol enhancement than did matched stem cell implants. At 4 weeks, T2 values of mismatched implants were significantly lower than those of matched implants in osteochondral defects of female rats (mean, 10.72 msec for human stem cells and 11.55 msec for male rat stem cells vs 15.45 msec for sex-matched rat stem cells; P = .02 and P = .04, respectively) and calvarial defects of recipient mice (mean, 21.7 msec vs 27.1 msec, respectively; P = .0444). This corresponded to increased recruitment of enhanced GFP- and rhodamine-ferumoxytol-positive macrophages into stem cell transplants, as visualized with IVM and histopathologic examination. Conclusion Endogenous labeling of macrophages with ferumoxytol enables noninvasive detection of innate immune responses to stem cell transplants with MR imaging. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Heike E Daldrup-Link
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Carmel Chan
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Olga Lenkov
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Seyedmeghdad Taghavigarmestani
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Toktam Nazekati
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Hossein Nejadnik
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Fanny Chapelin
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Aman Khurana
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Xinming Tong
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Fan Yang
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Laura Pisani
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Michael Longaker
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| | - Sanjiv Sam Gambhir
- From the Department of Radiology, Molecular Imaging Program at Stanford (MIPS) (H.E.D.L., C.C., O.L., S.T., T.N., H.N., F.C., A.K., F.Y., L.P., M.L., S.S.G.), Department of Pediatrics (H.E.D.L.), Institute for Stem Cell Biology and Regenerative Medicine (H.E.D.L.), Department of Orthopaedic Surgery (X.T., F.Y.), Department of Bioengineering (F.Y., S.S.G.), Department of Surgery, Division of Plastic and Reconstructive Surgery (M.L.), and Department of Materials Science and Engineering (M.L., S.S.G.), Stanford University, 725 Welch Rd, Room 1665, Stanford, CA 94305-5614
| |
Collapse
|
32
|
F4/80 + Host Macrophages Are a Barrier to Murine Embryonic Stem Cell-Derived Hematopoietic Progenitor Engraftment In Vivo. J Immunol Res 2016; 2016:2414906. [PMID: 27872864 PMCID: PMC5107259 DOI: 10.1155/2016/2414906] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 09/03/2016] [Accepted: 10/04/2016] [Indexed: 01/13/2023] Open
Abstract
Understanding how embryonic stem cells and their derivatives interact with the adult host immune system is critical to developing their therapeutic potential. Murine embryonic stem cell-derived hematopoietic progenitors (ESHPs) were generated via coculture with the bone marrow stromal cell line, OP9, and then transplanted into NOD.SCID.Common Gamma Chain (NSG) knockout mice, which lack B, T, and natural killer cells. Compared to control mice transplanted with adult lineage-negative bone marrow (Lin− BM) progenitors, ESHP-transplanted mice attained a low but significant level of donor hematopoietic chimerism. Based on our previous studies, we hypothesized that macrophages might contribute to the low engraftment of ESHPs in vivo. Enlarged spleens were observed in ESHP-transplanted mice and found to contain higher numbers of host F4/80+ macrophages compared to BM-transplanted controls. In vivo depletion of host macrophages using clodronate-loaded liposomes improved the ESHP-derived hematopoietic chimerism in the spleen but not in the BM. F4/80+ macrophages demonstrated a striking propensity to phagocytose ESHP targets in vitro. Taken together, these results suggest that macrophages are a barrier to both syngeneic and allogeneic ESHP engraftment in vivo.
Collapse
|
33
|
Doiron B, Hu W, DeFronzo RA. Beta Cell Formation in vivo Through Cellular Networking, Integration and Processing (CNIP) in Wild Type Adult Mice. Curr Pharm Biotechnol 2016; 17:376-88. [PMID: 26696016 PMCID: PMC5421132 DOI: 10.2174/1389201017666151223124031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 11/18/2015] [Accepted: 11/26/2015] [Indexed: 11/22/2022]
Abstract
Insulin replacement therapy is essential in type 1 diabetic individuals and is required in ~40-50% of type 2 diabetics during their lifetime. Prior attempts at beta cell regeneration have relied upon pancreatic injury to induce beta cell proliferation, dedifferentiation and activation of the embryonic pathway, or stem cell replacement. We report an alternative method to transform adult non-stem (somatic) cells into pancreatic beta cells. The Cellular Networking, Integration and Processing (CNIP) approach targets cellular mechanisms involved in pancreatic function in the organ’s adult state and utilizes a synergistic mechanism that integrates three important levels of cellular regulation to induce beta cell formation: (i) glucose metabolism, (ii) membrane receptor function, and (iii) gene transcription. The aim of the present study was to induce pancreatic beta cell formation in vivo in adult animals without stem cells and without dedifferentiating cells to recapitulate the embryonic pathway as previously published (1-3). Our results employing CNIP demonstrate that: (i) insulin secreting cells can be generated in adult pancreatic tissue in vivo and circumvent the problem of generating endocrine (glucagon and somatostatin) cells that exert deleterious effects on glucose homeostasis, and (ii) long-term normalization of glucose tolerance and insulin secretion can be achieved in a wild type diabetic mouse model. The CNIP cocktail has the potential to be used as a preventative or therapeutic treatment or cure for both type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Bruno Doiron
- Diabetes Division University of Texas Health Science Center 7703 Floyd Curl Drive San Antonio, Texas, 78231.
| | | | | |
Collapse
|
34
|
Sheyn D, Ben-David S, Shapiro G, De Mel S, Bez M, Ornelas L, Sahabian A, Sareen D, Da X, Pelled G, Tawackoli W, Liu Z, Gazit D, Gazit Z. Human Induced Pluripotent Stem Cells Differentiate Into Functional Mesenchymal Stem Cells and Repair Bone Defects. Stem Cells Transl Med 2016; 5:1447-1460. [PMID: 27400789 PMCID: PMC5070500 DOI: 10.5966/sctm.2015-0311] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/08/2016] [Indexed: 12/19/2022] Open
Abstract
Using short-term exposure of embryoid bodies to transforming growth factor-β, the authors directed induced pluripotent stem cells (iPSCs) toward mesenchymal stem cell (MSC) differentiation. Two types of iPSC-derived MSCs were identified: early (aiMSCs) and late (tiMSCs) outgrowing cells. Both types differentiated in vitro in response to osteogenic or adipogenic supplements; aiMSCs demonstrated higher osteogenic potential than tiMSCs. Upon orthotopic injection into radial defects, both types regenerated bone and contributed to defect repair. Mesenchymal stem cells (MSCs) are currently the most established cells for skeletal tissue engineering and regeneration; however, their availability and capability of self-renewal are limited. Recent discoveries of somatic cell reprogramming may be used to overcome these challenges. We hypothesized that induced pluripotent stem cells (iPSCs) that were differentiated into MSCs could be used for bone regeneration. Short-term exposure of embryoid bodies to transforming growth factor-β was used to direct iPSCs toward MSC differentiation. During this process, two types of iPSC-derived MSCs (iMSCs) were identified: early (aiMSCs) and late (tiMSCs) outgrowing cells. The transition of iPSCs toward MSCs was documented using MSC marker flow cytometry. Both types of iMSCs differentiated in vitro in response to osteogenic or adipogenic supplements. The results of quantitative assays showed that both cell types retained their multidifferentiation potential, although aiMSCs demonstrated higher osteogenic potential than tiMSCs and bone marrow-derived MSCs (BM-MSCs). Ectopic injections of BMP6-overexpressing tiMSCs produced no or limited bone formation, whereas similar injections of BMP6-overexpressing aiMSCs resulted in substantial bone formation. Upon orthotopic injection into radial defects, all three cell types regenerated bone and contributed to defect repair. In conclusion, MSCs can be derived from iPSCs and exhibit self-renewal without tumorigenic ability. Compared with BM-MSCs, aiMSCs acquire more of a stem cell phenotype, whereas tiMSCs acquire more of a differentiated osteoblast phenotype, which aids bone regeneration but does not allow the cells to induce ectopic bone formation (even when triggered by bone morphogenetic proteins), unless in an orthotopic site of bone fracture. Significance Mesenchymal stem cells (MSCs) are currently the most established cells for skeletal tissue engineering and regeneration of various skeletal conditions; however, availability of autologous MSCs is very limited. This study demonstrates a new method to differentiate human fibroblast-derived induced pluripotent stem cells (iPSCs) to cells with MSC properties, which we comprehensively characterized including differentiation potential and transcriptomic analysis. We showed that these iPS-derived MSCs are able to regenerate nonunion bone defects in mice more efficiently than bone marrow-derived human MSCs when overexpressing BMP6 using a nonviral transfection method.
Collapse
Affiliation(s)
- Dmitriy Sheyn
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shiran Ben-David
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Galina Shapiro
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sandra De Mel
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Maxim Bez
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Loren Ornelas
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- iPSC Core Facility, The David and Janet Polak Stem Cell Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Anais Sahabian
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- iPSC Core Facility, The David and Janet Polak Stem Cell Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dhruv Sareen
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, Jerusalem, Israel
- iPSC Core Facility, The David and Janet Polak Stem Cell Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Xiaoyu Da
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Gadi Pelled
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wafa Tawackoli
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zhenqiu Liu
- Biostatistics and Bioinformatics Core, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dan Gazit
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, Jerusalem, Israel
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zulma Gazit
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Skeletal Biotech Laboratory, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW In this review, we summarize the current status of clinical trials using therapeutic cells produced from human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). We also discuss combined cell and gene therapy via correction of defined mutations in human pluripotent stem cells and provide commentary on key obstacles facing widescale clinical adoption of pluripotent stem cell-based therapy. RECENT FINDINGS Initial data suggest that hESC/hiPSC-derived cell products used for retinal repair and spinal cord injury are safe for human use. Early-stage studies for treatment of cardiac injury and diabetes are also in progress. However, there remain key concerns regarding the safety and efficacy of these cells that need to be addressed in additional well designed clinical trials. Advances using the clustered regulatory interspaced short palindromic repeats (CRISPR)/Cas9 gene-editing system offer an improved tool for more rapid and on-target gene correction of genetic diseases. Combined gene and cell therapy using human pluripotent stem cells may provide an additional curative approach for disabling or lethal genetic and degenerative diseases wherein there are currently limited therapeutic opportunities. SUMMARY Human pluripotent stem cells are emerging as a promising tool to produce cells and tissues suitable for regenerative therapy for a variety of genetic and degenerative diseases.
Collapse
|
36
|
Emerging Implications for Extracellular Matrix-Based Technologies in Vascularized Composite Allotransplantation. Stem Cells Int 2016; 2016:1541823. [PMID: 26839554 PMCID: PMC4709778 DOI: 10.1155/2016/1541823] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/05/2015] [Indexed: 12/21/2022] Open
Abstract
Despite recent progress in vascularized composite allotransplantation (VCA), limitations including complex, high dose immunosuppression regimens, lifelong risk of toxicity from immunosuppressants, acute and most critically chronic graft rejection, and suboptimal nerve regeneration remain particularly challenging obstacles restricting clinical progress. When properly configured, customized, and implemented, biomaterials derived from the extracellular matrix (ECM) retain bioactive molecules and immunomodulatory properties that can promote stem cell migration, proliferation and differentiation, and constructive functional tissue remodeling. The present paper reviews the emerging implications of ECM-based technologies in VCA, including local immunomodulation, tissue repair, nerve regeneration, minimally invasive graft targeted drug delivery, stem cell transplantation, and other donor graft manipulation.
Collapse
|
37
|
Calderon D, Prot M, You S, Marquet C, Bellamy V, Bruneval P, Valette F, de Almeida P, Wu JC, Pucéat M, Menasché P, Chatenoud L. Control of Immune Response to Allogeneic Embryonic Stem Cells by CD3 Antibody-Mediated Operational Tolerance Induction. Am J Transplant 2016; 16:454-67. [PMID: 26492394 DOI: 10.1111/ajt.13477] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 07/02/2015] [Accepted: 07/07/2015] [Indexed: 01/25/2023]
Abstract
Implantation of embryonic stem cells (ESCs) and their differentiated derivatives into allogeneic hosts triggers an immune response that represents a hurdle to clinical application. We established in autoimmunity and in transplantation that CD3 antibody therapy induces a state of immune tolerance. Promising results have been obtained with CD3 antibodies in the clinic. In this study, we tested whether this strategy can prolong the survival of undifferentiated ESCs and their differentiated derivatives in histoincompatible hosts. Recipients of either mouse ESC-derived embryoid bodies (EBs) or cardiac progenitors received a single short tolerogenic regimen of CD3 antibody. In immunocompetent mice, allogeneic EBs and cardiac progenitors were rejected within 20-25 days. Recipients treated with CD3 antibody showed long-term survival of implanted cardiac progenitors or EBs. In due course, EBs became teratomas, the growth of which was self-limited. Regulatory CD4(+)FoxP3(+) T cells and signaling through the PD1/PDL1 pathway played key roles in the CD3 antibody therapeutic effect. Gene profiling emphasized the importance of TGF-β and the inhibitory T cell coreceptor Tim3 to the observed effect. These results demonstrate that CD3 antibody administered alone promotes prolonged survival of allogeneic ESC derivatives and thus could prove useful for enhancing cell engraftment in the absence of chronic immunosuppression.
Collapse
Affiliation(s)
- D Calderon
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,INSERM U1151, Hôpital Necker-Enfants Malades, Paris, France.,CNRS UMR 8253, Hôpital Necker-Enfants Malades, Paris, France
| | - M Prot
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,INSERM U1151, Hôpital Necker-Enfants Malades, Paris, France.,CNRS UMR 8253, Hôpital Necker-Enfants Malades, Paris, France
| | - S You
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,INSERM U1151, Hôpital Necker-Enfants Malades, Paris, France.,CNRS UMR 8253, Hôpital Necker-Enfants Malades, Paris, France
| | - C Marquet
- INSERM U1151, Hôpital Necker-Enfants Malades, Paris, France
| | - V Bellamy
- INSERM U970, Centre de Recherche Cardiovasculaire, Hôpital Européen Georges Pompidou, Paris, France
| | - P Bruneval
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,INSERM U970, Centre de Recherche Cardiovasculaire, Hôpital Européen Georges Pompidou, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Pathology, Paris, France
| | - F Valette
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,INSERM U1151, Hôpital Necker-Enfants Malades, Paris, France.,CNRS UMR 8253, Hôpital Necker-Enfants Malades, Paris, France
| | - P de Almeida
- Stanford Cardiovascular Institute and Departments of Medicine and Radiology, Stanford, CA
| | - J C Wu
- Stanford Cardiovascular Institute and Departments of Medicine and Radiology, Stanford, CA
| | - M Pucéat
- INSERM UMR-S910 Team Physiopathology of Cardiac Development, Aix-Marseille University, Medical School La Timone, Marseille, France
| | - P Menasché
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,INSERM U970, Centre de Recherche Cardiovasculaire, Hôpital Européen Georges Pompidou, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France
| | - L Chatenoud
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,INSERM U1151, Hôpital Necker-Enfants Malades, Paris, France.,CNRS UMR 8253, Hôpital Necker-Enfants Malades, Paris, France
| |
Collapse
|
38
|
Negroni E, Bigot A, Butler-Browne GS, Trollet C, Mouly V. Cellular Therapies for Muscular Dystrophies: Frustrations and Clinical Successes. Hum Gene Ther 2016; 27:117-26. [PMID: 26652770 DOI: 10.1089/hum.2015.139] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cell-based therapy for muscular dystrophies was initiated in humans after promising results obtained in murine models. Early trials failed to show substantial clinical benefit, sending researchers back to the bench, which led to the discovery of many hurdles as well as many new venues to optimize this therapeutic strategy. In this review we summarize progress in preclinical cell therapy approaches, with a special emphasis on human cells potentially attractive for human clinical trials. Future perspectives for cell therapy in skeletal muscle are discussed, including the perspective of combined therapeutic approaches.
Collapse
Affiliation(s)
- Elisa Negroni
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris, France
| | - Anne Bigot
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris, France
| | - Gillian S Butler-Browne
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris, France
| | - Capucine Trollet
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris, France
| | - Vincent Mouly
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, Paris, France
| |
Collapse
|
39
|
Hartman ME, Dai DF, Laflamme MA. Human pluripotent stem cells: Prospects and challenges as a source of cardiomyocytes for in vitro modeling and cell-based cardiac repair. Adv Drug Deliv Rev 2016; 96:3-17. [PMID: 25980938 DOI: 10.1016/j.addr.2015.05.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/27/2015] [Accepted: 05/07/2015] [Indexed: 12/13/2022]
Abstract
Human pluripotent stem cells (PSCs) represent an attractive source of cardiomyocytes with potential applications including disease modeling, drug discovery and safety screening, and novel cell-based cardiac therapies. Insights from embryology have contributed to the development of efficient, reliable methods capable of generating large quantities of human PSC-cardiomyocytes with cardiac purities ranging up to 90%. However, for human PSCs to meet their full potential, the field must identify methods to generate cardiomyocyte populations that are uniform in subtype (e.g. homogeneous ventricular cardiomyocytes) and have more mature structural and functional properties. For in vivo applications, cardiomyocyte production must be highly scalable and clinical grade, and we will need to overcome challenges including graft cell death, immune rejection, arrhythmogenesis, and tumorigenic potential. Here we discuss the types of human PSCs, commonly used methods to guide their differentiation into cardiomyocytes, the phenotype of the resultant cardiomyocytes, and the remaining obstacles to their successful translation.
Collapse
|
40
|
Pringproa K, Sathanawongs A, Khamphilai C, Sukkarinprom S, Oranratnachai A. Intravenous transplantation of mouse embryonic stem cells attenuates demyelination in an ICR outbred mouse model of demyelinating diseases. Neural Regen Res 2016; 11:1603-1609. [PMID: 27904491 PMCID: PMC5116839 DOI: 10.4103/1673-5374.193239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Induction of demyelination in the central nervous system (CNS) of experimental mice using cuprizone is widely used as an animal model for studying the pathogenesis and treatment of demyelination. However, different mouse strains used result in different pathological outcomes. Moreover, because current medicinal treatments are not always effective in multiple sclerosis patients, so the study of exogenous cell transplantation in an animal model is of great importance. The aims of the present study were to establish an alternative ICR outbred mouse model for studying demyelination and to evaluate the effects of intravenous cell transplantation in the present developed mouse model. Two sets of experiments were conducted. Firstly, ICR outbred and BALB/c inbred mice were fed with 0.2% cuprizone for 6 consecutive weeks; then demyelinating scores determined by luxol fast blue stain or immunolabeling with CNPase were evaluated. Secondly, attenuation of demyelination in ICR mice by intravenous injection of mES cells was studied. Scores for demyelination in the brains of ICR mice receiving cell injection (mES cells-injected group) and vehicle (sham-inoculated group) were assessed and compared. The results showed that cuprizone significantly induced demyelination in the cerebral cortex and corpus callosum of both ICR and BALB/c mice. Additionally, intravenous transplantation of mES cells potentially attenuated demyelination in ICR mice compared with sham-inoculated groups. The present study is among the earliest reports to describe the cuprizone-induced demyelination in ICR outbred mice. Although it remains unclear whether mES cells or trophic effects from mES cells are the cause of enhanced remyelination, the results of the present study may shed some light on exogenous cell therapy in central nervous system demyelinating diseases.
Collapse
Affiliation(s)
- Kidsadagon Pringproa
- Department of Veterinary Biosciences and Veterinary Public Heath, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anucha Sathanawongs
- Department of Veterinary Biosciences and Veterinary Public Heath, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chananthida Khamphilai
- Department of Veterinary Biosciences and Veterinary Public Heath, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sarocha Sukkarinprom
- Department of Veterinary Biosciences and Veterinary Public Heath, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Apichart Oranratnachai
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
41
|
Negroni E, Gidaro T, Bigot A, Butler-Browne GS, Mouly V, Trollet C. Invited review: Stem cells and muscle diseases: advances in cell therapy strategies. Neuropathol Appl Neurobiol 2015; 41:270-87. [PMID: 25405809 DOI: 10.1111/nan.12198] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 11/14/2014] [Indexed: 12/22/2022]
Abstract
Despite considerable progress to increase our understanding of muscle genetics, pathophysiology, molecular and cellular partners involved in muscular dystrophies and muscle ageing, there is still a crucial need for effective treatments to counteract muscle degeneration and muscle wasting in such conditions. This review focuses on cell-based therapy for muscle diseases. We give an overview of the different parameters that have to be taken into account in such a therapeutic strategy, including the influence of muscle ageing, cell proliferation and migration capacities, as well as the translation of preclinical results in rodent into human clinical approaches. We describe recent advances in different types of human myogenic stem cells, with a particular emphasis on myoblasts but also on other candidate cells described so far [CD133+ cells, aldehyde dehydrogenase-positive cells (ALDH+), muscle-derived stem cells (MuStem), embryonic stem cells (ES) and induced pluripotent stem cells (iPS)]. Finally, we provide an update of ongoing clinical trials using cell therapy strategies.
Collapse
Affiliation(s)
- Elisa Negroni
- Institut de Myologie, CNRS FRE3617, UPMC Univ Paris 06, UM76, INSERM U974, Sorbonne Universités, 47 bd de l'Hôpital, Paris, 75013, France
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
After decades of believing the heart loses the ability to regenerate soon after birth, numerous studies are now reporting that the adult heart may indeed be capable of regeneration, although the magnitude of new cardiac myocyte formation varies greatly. While this debate has energized the field of cardiac regeneration and led to a dramatic increase in our understanding of cardiac growth and repair, it has left much confusion in the field as to the prospects of regenerating the heart. Studies applying modern techniques of genetic lineage tracing and carbon-14 dating have begun to establish limits on the amount of endogenous regeneration after cardiac injury, but the underlying cellular mechanisms of this regeneration remained unclear. These same studies have also revealed an astonishing capacity for cardiac repair early in life that is largely lost with adult differentiation and maturation. Regardless, this renewed focus on cardiac regeneration as a therapeutic goal holds great promise as a novel strategy to address the leading cause of death in the developed world.
Collapse
Affiliation(s)
- Yiqiang Zhang
- Center for Cardiovascular Biology, Institute for Stem Cell Research and Division of Cardiology, Departments of Medicine and Pathology, University of Washington, Seattle, Washington
| | - John Mignone
- Center for Cardiovascular Biology, Institute for Stem Cell Research and Division of Cardiology, Departments of Medicine and Pathology, University of Washington, Seattle, Washington
| | - W Robb MacLellan
- Center for Cardiovascular Biology, Institute for Stem Cell Research and Division of Cardiology, Departments of Medicine and Pathology, University of Washington, Seattle, Washington
| |
Collapse
|
43
|
Tan Y, AlKhamees B, Jia D, Li L, Couture JF, Figeys D, Jinushi M, Wang L. MFG-E8 Is Critical for Embryonic Stem Cell-Mediated T Cell Immunomodulation. Stem Cell Reports 2015; 5:741-752. [PMID: 26455415 PMCID: PMC4649138 DOI: 10.1016/j.stemcr.2015.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 02/03/2023] Open
Abstract
The molecules and mechanisms pertinent to the low immunogenicity of undifferentiated embryonic stem cells (ESCs) remain poorly understood. Here, we provide evidence that milk fat globule epidermal growth factor 8 (MFG-E8) is a vital mediator in this phenomenon and directly suppresses T cell immune responses. MFG-E8 is enriched in undifferentiated ESCs but diminished in differentiated ESCs. Upregulation of MFG-E8 in ESCs increases the successful engraftment of both undifferentiated and differentiated ESCs across major histocompatibility complex barriers. MFG-E8 suppresses T cell activation/proliferation and inhibits Th1, Th2, and Th17 subpopulations while increasing regulatory T cell subsets. Neutralizing MFG-E8 substantially abrogates these effects, whereas addition of recombinant MFG-E8 to differentiated ESCs restores immunosuppression. Furthermore, we provide the evidence that MFG-E8 suppresses T cell activation and regulates T cell polarization by inhibiting PKCθ phosphorylation through the α3/5βV integrin receptor. Our findings offer an approach to facilitate transplantation acceptance. MFG-E8 is enriched in undifferentiated but diminished in differentiated ESCs MFG-E8 promotes allogeneic engraftment of ESC-derived tissues across the MHC barrier ESC-produced MFG-E8 inhibits Th1/Th2/Th17 while promoting regulatory T cells MFG-E8 modulates T cell polarization via inhibiting PKCθ phosphorylation
Collapse
Affiliation(s)
- Yuan Tan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Bodour AlKhamees
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Deyong Jia
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Li Li
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Jean-François Couture
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Daniel Figeys
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Masahisa Jinushi
- Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjiku-ku, Tokyo 160-8582, Japan.
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.
| |
Collapse
|
44
|
Stace ET, Dakin SG, Mouthuy PA, Carr AJ. Translating Regenerative Biomaterials Into Clinical Practice. J Cell Physiol 2015; 231:36-49. [DOI: 10.1002/jcp.25071] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 06/05/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Edward T. Stace
- National Institute of Health Research Musculoskeletal Biomedical Research Unit; Oxford United Kingdom
- Botnar Institute of Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences; University of Oxford; Oxford United Kingdom
| | - Stephanie G. Dakin
- National Institute of Health Research Musculoskeletal Biomedical Research Unit; Oxford United Kingdom
- Botnar Institute of Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences; University of Oxford; Oxford United Kingdom
| | - Pierre-Alexis Mouthuy
- National Institute of Health Research Musculoskeletal Biomedical Research Unit; Oxford United Kingdom
- Botnar Institute of Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences; University of Oxford; Oxford United Kingdom
| | - Andrew J. Carr
- National Institute of Health Research Musculoskeletal Biomedical Research Unit; Oxford United Kingdom
- Botnar Institute of Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences; University of Oxford; Oxford United Kingdom
| |
Collapse
|
45
|
Stem Cells and Regenerative Medicine: Myth or Reality of the 21th Century. Stem Cells Int 2015; 2015:734731. [PMID: 26300923 PMCID: PMC4537770 DOI: 10.1155/2015/734731] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 04/22/2015] [Accepted: 05/24/2015] [Indexed: 02/07/2023] Open
Abstract
Since the 1960s and the therapeutic use of hematopoietic stem cells of bone marrow origin, there has been an increasing interest in the study of undifferentiated progenitors that have the ability to proliferate and differentiate into various tissues. Stem cells (SC) with different potency can be isolated and characterised. Despite the promise of embryonic stem cells, in many cases, adult or even fetal stem cells provide a more interesting approach for clinical applications. It is undeniable that mesenchymal stem cells (MSC) from bone marrow, adipose tissue, or Wharton's Jelly are of potential interest for clinical applications in regenerative medicine because they are easily available without ethical problems for their uses. During the last 10 years, these multipotent cells have generated considerable interest and have particularly been shown to escape to allogeneic immune response and be capable of immunomodulatory activity. These properties may be of a great interest for regenerative medicine. Different clinical applications are under study (cardiac insufficiency, atherosclerosis, stroke, bone and cartilage deterioration, diabetes, urology, liver, ophthalmology, and organ's reconstruction). This review focuses mainly on tissue and organ regeneration using SC and in particular MSC.
Collapse
|
46
|
Guo YL, Carmichael GG, Wang R, Hong X, Acharya D, Huang F, Bai F. Attenuated Innate Immunity in Embryonic Stem Cells and Its Implications in Developmental Biology and Regenerative Medicine. Stem Cells 2015; 33:3165-73. [PMID: 26086534 DOI: 10.1002/stem.2079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/16/2015] [Indexed: 12/14/2022]
Abstract
Embryonic stem cells (ESCs) represent a promising cell source for regenerative medicine. Intensive research over the past 2 decades has led to the feasibility of using ESC-differentiated cells (ESC-DCs) in regenerative medicine. However, increasing evidence indicates that ESC-DCs generated by current differentiation methods may not have equivalent cellular functions to their in vivo counterparts. Recent studies have revealed that both human and mouse ESCs as well as some types of ESC-DCs lack or have attenuated innate immune responses to a wide range of infectious agents. These findings raise important concerns for their therapeutic applications since ESC-DCs, when implanted to a wound site of a patient, where they would likely be exposed to pathogens and inflammatory cytokines. Understanding whether an attenuated immune response is beneficial or harmful to the interaction between host and grafted cells becomes an important issue for ESC-based therapy. A substantial amount of recent evidence has demonstrated that the lack of innate antiviral responses is a common feature to ESCs and other types of pluripotent cells. This has led to the hypothesis that mammals may have adapted different antiviral mechanisms at different stages of organismal development. The underdeveloped innate immunity represents a unique and uncharacterized property of ESCs that may have important implications in developmental biology, immunology, and in regenerative medicine.
Collapse
Affiliation(s)
- Yan-Lin Guo
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Gordon G Carmichael
- The Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Ruoxing Wang
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Xiaoxiao Hong
- The Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Dhiraj Acharya
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Faqing Huang
- The Department of Chemistry and Biochemistry, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Fengwei Bai
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Ongoing research is constantly looking for means to modulate the immune system for long-lasting engraftment of pluripotent stem cells (PSC) during stem cell-based therapies. This study reviews data on in-vitro and in-vivo immunogenicity of embryonic and induced-PSC and describes how their immunological properties can be harnessed for tolerance induction in organ transplantation. RECENT FINDINGS Although PSC display immunomodulatory properties in vitro, they are capable of eliciting an immune response that leads to cell rejection when transplanted into immune-competent recipients. Nevertheless, long-term acceptance of PSC-derived cells/tissues in an allogeneic environment can be achieved using minimal host conditioning. Protocols for differentiating PSC towards haematopoietic stem cells, thymic epithelial precursors, dendritic cells, regulatory T cells and myeloid-derived suppressor cells are being developed, suggesting the possibility to use PSC-derived immunomodulatory cells to induce tolerance to a solid organ transplant. SUMMARY PSC and/or their derivatives possess unique immunological properties that allow for acceptance of PSC-derived tissue with minimal host conditioning. Investigators involved either in regenerative or in transplant medicine must join their efforts with the ultimate aim of using PSC as a source of donor-specific cells that would create a protolerogenic environment to achieve tolerance in solid organ transplantation.
Collapse
|
48
|
Kruse V, Hamann C, Monecke S, Cyganek L, Elsner L, Hübscher D, Walter L, Streckfuss-Bömeke K, Guan K, Dressel R. Human Induced Pluripotent Stem Cells Are Targets for Allogeneic and Autologous Natural Killer (NK) Cells and Killing Is Partly Mediated by the Activating NK Receptor DNAM-1. PLoS One 2015; 10:e0125544. [PMID: 25950680 PMCID: PMC4423859 DOI: 10.1371/journal.pone.0125544] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/25/2015] [Indexed: 02/07/2023] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) could be used to generate autologous cells for therapeutic purposes, which are expected to be tolerated by the recipient. However, iPSC-derived grafts are at risk of giving rise to teratomas in the host, if residuals of tumorigenic cells are not rejected by the recipient. We have analyzed the susceptibility of hiPSC lines to allogeneic and autologous natural killer (NK) cells. IL-2-activated, in contrast to resting NK cells killed hiPSC lines efficiently (P = 1.69 x 10(-39)). Notably, the specific lysis of the individual hiPSC lines by IL-2-activated NK cells was significantly different (P = 1.72 x 10(-6)) and ranged between 46 % and 64 % in 51Cr-release assays when compared to K562 cells. The hiPSC lines were killed by both allogeneic and autologous NK cells although autologous NK cells were less efficient (P=8.63 x 10(-6)). Killing was partly dependent on the activating NK receptor DNAM-1 (P = 8.22 x 10(-7)). The DNAM-1 ligands CD112 and CD155 as well as the NKG2D ligands MICA and MICB were expressed on the hiPSC lines. Low amounts of human leukocyte antigen (HLA) class I proteins, which serve as ligands for inhibitory and activating NK receptors were also detected. Thus, the susceptibility to NK cell killing appears to constitute a common feature of hiPSCs. Therefore, NK cells might reduce the risk of teratoma formation even after autologous transplantations of pluripotent stem cell-derived grafts that contain traces of pluripotent cells.
Collapse
Affiliation(s)
- Vanessa Kruse
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Carina Hamann
- Department of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
| | - Sebastian Monecke
- Department of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
| | - Lukas Cyganek
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
| | - Leslie Elsner
- Department of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
| | - Daniela Hübscher
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
| | - Lutz Walter
- Primate Genetics Laboratory, German Primate Center, Göttingen, Germany
| | - Katrin Streckfuss-Bömeke
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
| | - Kaomei Guan
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
- * E-mail: (RD); (KG)
| | - Ralf Dressel
- Department of Cellular and Molecular Immunology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner site Göttingen, Germany
- * E-mail: (RD); (KG)
| |
Collapse
|
49
|
Noh MJ, Copeland O, O’Mara M, Lee KH. Cell mediated gene therapy: A guide for doctors in the clinic. World J Med Genet 2015; 5:1-13. [DOI: 10.5496/wjmg.v5.i1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/16/2014] [Accepted: 01/19/2015] [Indexed: 02/06/2023] Open
Abstract
The recent approval of gene therapy products in Europe and Asia and the upsurge of gene therapy products in clinical trials signal the rebound of this technology not only for many orphan diseases but also for non-life threatening diseases. Following the success of induced pluripotent stem (iPS) cells in research, other modified ex vivo gene therapies are also knocking on the door of the clinic. Historically, gene therapy has experienced many ups and downs and still faces many challenges. During the past 10 years, many new ideas have been tried, and the goal of making this technology a more effective treatment modality through greater safety and control is coming within reach. The first clinical trial of iPS cells has begun, and cell mediated gene therapy products have reached phase III in some countries. The potential for tumorigenicity and immunogenicity are still concerns with these products, so physicians should understand the biological aspects of engineered cells in the clinic. In this review article, we attempted to provide a summary update of the current state of knowledge regarding this technology: that is, we reviewed products that have finished clinical trials, are still in clinical trials and/or are at the research stage. We also focused on the challenges, future directions, and strategies for making this technology available in the clinic. In addition, the available measures for making gene therapy products safer are within the scope of this article. It is also important to understand the manufacturing process for gene therapy products, because cell characteristics can change during the cell expansion process. When physicians use gene therapy products in the clinic, they should be aware of the viability, temperature sensitivity and stability of these cells because biologic products are different from chemical products. Although we may not be able to answer all possible questions and concerns, we believe that this is the right time for physicians to increase their interest in and understanding of this evolving technology.
Collapse
|
50
|
Buikema JW, Van Der Meer P, Sluijter JPG, Domian IJ. Concise review: Engineering myocardial tissue: the convergence of stem cells biology and tissue engineering technology. Stem Cells 2015; 31:2587-98. [PMID: 23843322 DOI: 10.1002/stem.1467] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 05/09/2013] [Accepted: 05/14/2013] [Indexed: 12/11/2022]
Abstract
Advanced heart failure represents a leading public health problem in the developed world. The clinical syndrome results from the loss of viable and/or fully functional myocardial tissue. Designing new approaches to augment the number of functioning human cardiac muscle cells in the failing heart serve as the foundation of modern regenerative cardiovascular medicine. A number of clinical trials have been performed in an attempt to increase the number of functional myocardial cells by the transplantation of a diverse group of stem or progenitor cells. Although there are some encouraging suggestions of a small early therapeutic benefit, to date, no evidence for robust cell or tissue engraftment has been shown, emphasizing the need for new approaches. Clinically meaningful cardiac regeneration requires the identification of the optimum cardiogenic cell types and their assembly into mature myocardial tissue that is functionally and electrically coupled to the native myocardium. We here review recent advances in stem cell biology and tissue engineering and describe how the convergence of these two fields may yield novel approaches for cardiac regeneration.
Collapse
Affiliation(s)
- Jan Willem Buikema
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | | | | |
Collapse
|