51
|
Bárcena C, López-Otín C. A fruitful liaison of ZSCAN10 and ROS on the road to rejuvenation. Nat Cell Biol 2017; 19:1012-1013. [PMID: 28855729 DOI: 10.1038/ncb3602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Induced pluripotent stem cells derived from aged donors (A-iPSCs) usually show genomic instability that affects their utility and raises concerns about their safety. Now, a study highlights the importance of ZSCAN10-dependent recovery of glutathione-ROS homeostasis in counteracting the genomic defects in A-iPSCs.
Collapse
Affiliation(s)
- Clea Bárcena
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
52
|
EMT/MET at the Crossroad of Stemness, Regeneration and Oncogenesis: The Ying-Yang Equilibrium Recapitulated in Cell Spheroids. Cancers (Basel) 2017; 9:cancers9080098. [PMID: 28758926 PMCID: PMC5575601 DOI: 10.3390/cancers9080098] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 07/20/2017] [Accepted: 07/26/2017] [Indexed: 12/21/2022] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) is an essential trans-differentiation process, which plays a critical role in embryonic development, wound healing, tissue regeneration, organ fibrosis, and cancer progression. It is the fundamental mechanism by which epithelial cells lose many of their characteristics while acquiring features typical of mesenchymal cells, such as migratory capacity and invasiveness. Depending on the contest, EMT is complemented and balanced by the reverse process, the mesenchymal-to-epithelial transition (MET). In the saving economy of the living organisms, the same (Ying-Yang) tool is integrated as a physiological strategy in embryonic development, as well as in the course of reparative or disease processes, prominently fibrosis, tumor invasion and metastasis. These mechanisms and their related signaling (e.g., TGF-β and BMPs) have been effectively studied in vitro by tissue-derived cell spheroids models. These three-dimensional (3D) cell culture systems, whose phenotype has been shown to be strongly dependent on TGF-β-regulated EMT/MET processes, present the advantage of recapitulating in vitro the hypoxic in vivo micro-environment of tissue stem cell niches and their formation. These spheroids, therefore, nicely reproduce the finely regulated Ying-Yang equilibrium, which, together with other mechanisms, can be determinant in cell fate decisions in many pathophysiological scenarios, such as differentiation, fibrosis, regeneration, and oncogenesis. In this review, current progress in the knowledge of signaling pathways affecting EMT/MET and stemness regulation will be outlined by comparing data obtained from cellular spheroids systems, as ex vivo niches of stem cells derived from normal and tumoral tissues. The mechanistic correspondence in vivo and the possible pharmacological perspective will be also explored, focusing especially on the TGF-β-related networks, as well as others, such as SNAI1, PTEN, and EGR1. This latter, in particular, for its ability to convey multiple types of stimuli into relevant changes of the cell transcriptional program, can be regarded as a heterogeneous "stress-sensor" for EMT-related inducers (growth factor, hypoxia, mechano-stress), and thus as a therapeutic target.
Collapse
|
53
|
An autocrine inflammatory forward-feedback loop after chemotherapy withdrawal facilitates the repopulation of drug-resistant breast cancer cells. Cell Death Dis 2017; 8:e2932. [PMID: 28703802 PMCID: PMC5550865 DOI: 10.1038/cddis.2017.319] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 05/03/2017] [Accepted: 06/08/2017] [Indexed: 12/17/2022]
Abstract
Stromal cells, infiltrating immune cells, paracrine factors and extracellular matrix have been extensively studied in cancers. However, autocrine factors produced by tumor cells and communications between autocrine factors and intracellular signaling pathways in the development of drug resistance, cancer stem-like cells (CSCs) and tumorigenesis have not been well investigated, and the precise mechanism and tangible approaches remain elusive. Here we reveal a new mechanism by which cytokines produced by breast cancer cells after chemotherapy withdrawal activate both Wnt/β-catenin and NF-κB pathways, which in turn further promote breast cancer cells to produce and secrete cytokines, forming an autocrine inflammatory forward-feedback loop to facilitate the enrichment of drug-resistant breast cancer cells and/or CSCs. Such an unexpected autocrine forward-feedback loop and CSC enrichment can be effectively blocked by inhibition of Wnt/β-catenin and NF-κB signaling. It can also be diminished by IL8-neutralizing antibody or blockade of IL8 receptors CXCR1/2 with reparixin. Administration of reparixin after chemotherapy withdrawal effectively attenuates tumor masses in a human xenograft model and abolishes paclitaxel-enriched CSCs in the secondary transplantation. These results are partially supported by the latest clinical data set. Breast cancer patients treated with chemotherapeutic drugs exhibited poor survival rate (66.7 vs 282.8 months, P=0.00071) and shorter disease-free survival time if their tumor samples expressed high level of IL8, CXCR1, CXCR2 genes and Wnt target genes. Taken together, this study provides new insights into the communication between autocrine niches and signaling pathways in the development of chemotherapy resistance and CSCs; it also offers a tangible approach in breast cancer treatment.
Collapse
|
54
|
Abstract
For >4 decades, the holy grail in the treatment of acute myocardial infarction has been the mitigation of lethal injury. Despite promising initial results and decades of investigation by the cardiology research community, the only treatment with proven efficacy is early reperfusion of the occluded coronary artery. The remarkable record of failure has led us and others to wonder if cardioprotection is dead. The path to translation, like the ascent to Everest, is certainly littered with corpses. We do, however, highlight a therapeutic principle that provides a glimmer of hope: cellular postconditioning. Administration of cardiosphere-derived cells after reperfusion limits infarct size measured acutely, while providing long-term structural and functional benefits. The recognition that cell therapy may be cardioprotective, and not just regenerative, merits further exploration before we abandon the pursuit entirely.
Collapse
Affiliation(s)
- David J Lefer
- From Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans (D.J.L.); and Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.).
| | - Eduardo Marbán
- From Cardiovascular Center of Excellence and Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans (D.J.L.); and Cedars-Sinai Heart Institute, Los Angeles, CA (E.M.)
| |
Collapse
|
55
|
Dinh PUC, Cores J, Hensley MT, Vandergriff AC, Tang J, Allen TA, Caranasos TG, Adler KB, Lobo LJ, Cheng K. Derivation of therapeutic lung spheroid cells from minimally invasive transbronchial pulmonary biopsies. Respir Res 2017; 18:132. [PMID: 28666430 PMCID: PMC5493087 DOI: 10.1186/s12931-017-0611-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/12/2017] [Indexed: 12/21/2022] Open
Abstract
Background Resident stem and progenitor cells have been identified in the lung over the last decade, but isolation and culture of these cells remains a challenge. Thus, although these lung stem and progenitor cells provide an ideal source for stem-cell based therapy, mesenchymal stem cells (MSCs) remain the most popular cell therapy product for the treatment of lung diseases. Surgical lung biopsies can be the tissue source but such procedures carry a high risk of mortality. Methods In this study we demonstrate that therapeutic lung cells, termed “lung spheroid cells” (LSCs) can be generated from minimally invasive transbronchial lung biopsies using a three-dimensional culture technique. The cells were then characterized by flow cytometry and immunohistochemistry. Angiogenic potential was tested by in-vitro HUVEC tube formation assay. In-vivo bio- distribution of LSCs was examined in athymic nude mice after intravenous delivery. Results From one lung biopsy, we are able to derive >50 million LSC cells at Passage 2. These cells were characterized by flow cytometry and immunohistochemistry and were shown to represent a mixture of lung stem cells and supporting cells. When introduced systemically into nude mice, LSCs were retained primarily in the lungs for up to 21 days. Conclusion Here, for the first time, we demonstrated that direct culture and expansion of human lung progenitor cells from pulmonary tissues, acquired through a minimally invasive biopsy, is possible and straightforward with a three-dimensional culture technique. These cells could be utilized in long-term expansion of lung progenitor cells and as part of the development of cell-based therapies for the treatment of lung diseases such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0611-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Phuong-Uyen C Dinh
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Jhon Cores
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, USA
| | - M Taylor Hensley
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Adam C Vandergriff
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, USA
| | - Junnan Tang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA.,Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tyler A Allen
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Thomas G Caranasos
- Divisions of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kenneth B Adler
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Leonard J Lobo
- Pulmonary Diseases and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA. .,Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, USA.
| |
Collapse
|
56
|
Fuchi N, Miura K, Doi H, Li TS, Masuzaki H. Feasibility of placenta-derived mesenchymal stem cells as a tool for studying pregnancy-related disorders. Sci Rep 2017; 7:46220. [PMID: 28401946 PMCID: PMC5388876 DOI: 10.1038/srep46220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 03/10/2017] [Indexed: 12/25/2022] Open
Abstract
The cellular and molecular mechanisms responsible for pregnancy-related disorders remain unclear. We investigated the feasibility of using placenta-derived mesenchymal stem cells (MSCs) as a tool to study such pregnancy-related disorders. We isolated and expanded adequate numbers of cells with characteristic features of MSCs from the chorionic plate (CP-MSCs), chorionic villi (CV-MSCs), and decidua basalis (DB-MSCs) of human term placental tissues. All placenta-derived MSCs expressed pregnancy-associated C14MC microRNA (miRNA) (miR-323-3p). Interestingly, the placenta-specific C19MC miRNAs (miR-518b and miR517a) were clearly expressed in CP-MSCs and CV-MSCs of foetal origin, but were barely expressed in DB-MSCs of maternal origin. Furthermore, expression levels of placenta-specific C19MC miRNAs in CV-MSCs remained stable during the ex vivo expansion process and across different pregnancy phases (first trimester versus third trimester). High-efficiency siRNA transfection was confirmed in twice-passaged CV-MSCs with little toxicity, and microarray analysis was used to screen for miR-518b target genes. Placenta-derived MSCs, especially CV-MSCs, are a potential tool for investigating the role of placental miRNAs in pregnancy-related disorders.
Collapse
Affiliation(s)
- Naoki Fuchi
- Department of Obstetrics and Gynaecology, Nagasaki University Graduate School of Medicine, Nagasaki, Japan.,Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Kiyonori Miura
- Department of Obstetrics and Gynaecology, Nagasaki University Graduate School of Medicine, Nagasaki, Japan
| | - Hanako Doi
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Hideaki Masuzaki
- Department of Obstetrics and Gynaecology, Nagasaki University Graduate School of Medicine, Nagasaki, Japan
| |
Collapse
|
57
|
Sub-physiological oxygen levels optimal for growth and survival of human atrial cardiac stem cells. Mol Cell Biochem 2017; 432:109-122. [PMID: 28386845 DOI: 10.1007/s11010-017-3002-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/04/2017] [Indexed: 12/21/2022]
Abstract
Cardiac stem cells reside in niches where the oxygen levels are close to 3%. For cytotherapy, cells are conventionally expanded in ambient oxygen (21% O2) which represents hyperoxia compared to the oxygen tension of niches. Cardiosphere-derived cells (CDCs) are then transplanted to host tissue with lower-O2 levels. The high-O2 gradient can reduce the efficacy of cultured cells. Based on the assumption that minimizing injury due to O2 gradients will enhance the yield of functionally efficient cells, CDCs were cultured in 3% O2 and compared with cells maintained in ambient O2. CDCs were isolated from human right atrial explants and expanded in parallel in 21 and 3% oxygen and compared with regard to survival, proliferation, and retention of stemness. Increased cell viability even in the tenth passage and enhanced cardiosphere formation was observed in cells expanded in 3% O2. The cell yield from seven passages was fourfold higher for cells cultured in 3% O2. Preservation of stemness in hypoxic environment was evident from the proportion of c-kit-positive cells and reduced myogenic differentiation. Hypoxia promoted angiogenesis and reduced the tendency to differentiate to noncardiac lineages (adipocytes and osteocytes). Mimicking the microenvironment at transplantation, when shifted to 5% O2, viability and proliferation rate were significantly higher for CDCs expanded in 3% O2. Expansion of CDCs, from atria in sub-physiological oxygen, helps in obtaining a higher yield of healthy cells with better preservation of stem cell characteristics. The cells so cultured are expected to improve engraftment and facilitate myocardial regeneration.
Collapse
|
58
|
Guo CY, Yan C, Luo L, Goto S, Urata Y, Xu JJ, Wen XM, Kuang YK, Tou FF, Li TS. Enhanced expression of PKM2 associates with the biological properties of cancer stem cells from A549 human lung cancer cells. Oncol Rep 2017; 37:2161-2166. [PMID: 28259998 DOI: 10.3892/or.2017.5438] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/25/2017] [Indexed: 11/05/2022] Open
Abstract
Cancer cells express the M2 isoform of glycolytic enzyme pyruvate kinase (PKM2) for favoring the survival under a hypoxic condition. Considering the relative low oxygen microenvironment in stem cell niche, we hypothesized that an enhanced PKM2 expression associates with the biological properties of cancer stem cells. We used A549 human lung cancer cell line and surgical resected lung cancer tissue samples from patients for experiments. We confirmed the co-localization of PKM2 and CD44, a popular marker for cancer stem cells in lung cancer tissue samples from patients. The expression of PKM2 was clearly observed in approximately 80% of the A549 human lung cancer cells. Remarkably, enhanced expression of PKM2 was specially observed in these cells that also positively expressed CD44. Downregulation of PKM2 in CD44+ cancer stem cells by siRNA significantly impaired the potency for spheroid formation, decreased the cell survival under fetal bovine serum deprivation and hypoxic conditions, but increased their sensitivity to anti-cancer drug of cisplatin and γ-ray. The enhanced expression of PKM2 seems to associate with the biological properties of cancer stem cells from A549 human lung cancer cells. Selective targeting of PKM2 may provide a new strategy for cancer therapy, especially for patients with therapeutic resistance.
Collapse
Affiliation(s)
- Chang-Ying Guo
- Department of Thoracic Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chen Yan
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Lan Luo
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Shinji Goto
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Yoshishige Urata
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Jian-Jun Xu
- Department of Thoracic Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiao-Ming Wen
- Department of Thoracic Surgery, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi 330029, P.R. China
| | - Yu-Kang Kuang
- Department of Thoracic Surgery, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi 330029, P.R. China
| | - Fang-Fang Tou
- Department of Thoracic Surgery, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi 330029, P.R. China
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| |
Collapse
|
59
|
Chen C, Termglinchan V, Karakikes I. Concise Review: Mending a Broken Heart: The Evolution of Biological Therapeutics. Stem Cells 2017; 35:1131-1140. [PMID: 28233392 DOI: 10.1002/stem.2602] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 12/19/2016] [Accepted: 01/07/2017] [Indexed: 12/21/2022]
Abstract
Heart failure (HF), a common sequela of cardiovascular diseases, remains a staggering clinical problem, associated with high rates of morbidity and mortality worldwide. Advances in pharmacological, interventional, and operative management have improved patient care, but these interventions are insufficient to halt the progression of HF, particularly the end-stage irreversible loss of functional cardiomyocytes. Innovative therapies that could prevent HF progression and improve the function of the failing heart are urgently needed. Following successful preclinical studies, two main strategies have emerged as potential solutions: cardiac gene therapy and cardiac regeneration through stem and precursor cell transplantation. Many potential gene- and cell-based therapies have entered into clinical studies, intending to ameliorate cardiac dysfunction in patients with advanced HF. In this review, we focus on the recent advances in cell- and gene-based therapies in the context of cardiovascular disease, emphasizing the most advanced therapies. The principles and mechanisms of action of gene and cell therapies for HF are discussed along with the limitations of current approaches. Finally, we highlight the emerging technologies that hold promise to revolutionize the biological therapies for cardiovascular diseases. Stem Cells 2017;35:1131-1140.
Collapse
Affiliation(s)
- Caressa Chen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Vittavat Termglinchan
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Ioannis Karakikes
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA.,Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
60
|
Hensley MT, Tang J, Woodruff K, Defrancesco T, Tou S, Williams CM, Breen M, Meurs K, Keene B, Cheng K. Intracoronary allogeneic cardiosphere-derived stem cells are safe for use in dogs with dilated cardiomyopathy. J Cell Mol Med 2017; 21:1503-1512. [PMID: 28296006 PMCID: PMC5543505 DOI: 10.1111/jcmm.13077] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/21/2016] [Indexed: 12/18/2022] Open
Abstract
Cardiosphere‐derived cells (CDCs) have been shown to reduce scar size and increase viable myocardium in human patients with mild/moderate myocardial infarction. Studies in rodent models suggest that CDC therapy may confer therapeutic benefits in patients with non‐ischaemic dilated cardiomyopathy (DCM). We sought to determine the safety and efficacy of allogeneic CDC in a large animal (canine) model of spontaneous DCM. Canine CDCs (cCDCs) were grown from a donor dog heart. Similar to human CDCs, cCDCs express CD105 and are slightly positive for c‐kit and CD90. Thirty million of allogeneic cCDCs was infused into the coronary vessels of Doberman pinscher dogs with spontaneous DCM. Adverse events were closely monitored, and cardiac functions were measured by echocardiography. No adverse events occurred during and after cell infusion. Histology on dog hearts (after natural death) revealed no sign of immune rejection from the transplanted cells.
Collapse
Affiliation(s)
- Michael Taylor Hensley
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Junnan Tang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA.,Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kathleen Woodruff
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Teresa Defrancesco
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Sandra Tou
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Christina M Williams
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Mathew Breen
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA.,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Kathryn Meurs
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Bruce Keene
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA.,Joint Department of Biomedical Engineering, University of North Carolina Chapel Hill and North Carolina State University, Raleigh and Chapel Hill, NC, USA.,Eshelman School of Pharmacy, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
61
|
Chimenti I, Massai D, Morbiducci U, Beltrami AP, Pesce M, Messina E. Stem Cell Spheroids and Ex Vivo Niche Modeling: Rationalization and Scaling-Up. J Cardiovasc Transl Res 2017; 10:150-166. [PMID: 28289983 DOI: 10.1007/s12265-017-9741-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/27/2017] [Indexed: 02/08/2023]
Abstract
Improved protocols/devices for in vitro culture of 3D cell spheroids may provide essential cues for proper growth and differentiation of stem/progenitor cells (S/PCs) in their niche, allowing preservation of specific features, such as multi-lineage potential and paracrine activity. Several platforms have been employed to replicate these conditions and to generate S/PC spheroids for therapeutic applications. However, they incompletely reproduce the niche environment, with partial loss of its highly regulated network, with additional hurdles in the field of cardiac biology, due to debated resident S/PCs therapeutic potential and clinical translation. In this contribution, the essential niche conditions (metabolic, geometric, mechanical) that allow S/PCs maintenance/commitment will be discussed. In particular, we will focus on both existing bioreactor-based platforms for the culture of S/PC as spheroids, and on possible criteria for the scaling-up of niche-like spheroids, which could be envisaged as promising tools for personalized cardiac regenerative medicine, as well as for high-throughput drug screening.
Collapse
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Rome, Italy
| | - Diana Massai
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic-, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Umberto Morbiducci
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | | | - Maurizio Pesce
- Tissue Engineering Research Unit, "Centro Cardiologico Monzino", IRCCS, Milan, Italy
| | - Elisa Messina
- Department of Pediatrics and Infant Neuropsychiatry, "Umberto I" Hospital, "La Sapienza" University, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
62
|
Abstract
The hearts of lower vertebrates such as fish and salamanders display scarless regeneration following injury, although this feature is lost in adult mammals. The remarkable capacity of the neonatal mammalian heart to regenerate suggests that the underlying machinery required for the regenerative process is evolutionarily retained. Recent studies highlight the epicardial covering of the heart as an important source of the signalling factors required for the repair process. The developing epicardium is also a major source of cardiac fibroblasts, smooth muscle, endothelial cells and stem cells. Here, we examine animal models that are capable of scarless regeneration, the role of the epicardium as a source of cells, signalling mechanisms implicated in the regenerative process and how these mechanisms influence cardiomyocyte proliferation. We also discuss recent advances in cardiac stem cell research and potential therapeutic targets arising from these studies.
Collapse
Affiliation(s)
| | - Nadia Rosenthal
- National Heart and Lung Institute, Imperial College London, London, UK Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia The Jackson Laboratory, Bar Harbor, ME, USA
| |
Collapse
|
63
|
Pagano F, Angelini F, Siciliano C, Tasciotti J, Mangino G, De Falco E, Carnevale R, Sciarretta S, Frati G, Chimenti I. Beta2-adrenergic signaling affects the phenotype of human cardiac progenitor cells through EMT modulation. Pharmacol Res 2017; 127:41-48. [PMID: 28099883 DOI: 10.1016/j.phrs.2017.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 12/12/2016] [Accepted: 01/13/2017] [Indexed: 01/08/2023]
Abstract
Human cardiac progenitor cells (CPCs) offer great promises to cardiac cell therapy for heart failure. Many in vivo studies have shown their therapeutic benefits, paving the way for clinical translation. The 3D model of cardiospheres (CSs) represents a unique niche-like in vitro microenvironment, which includes CPCs and supporting cells. CSs have been shown to form through a process mediated by epithelial-to-mesenchymal transition (EMT). β2-Adrenergic signaling significantly affects stem/progenitor cells activation and mobilization in multiple tissues, and crosstalk between β2-adrenergic signaling and EMT processes has been reported. In the present study, we aimed at investigating the biological response of CSs to β2-adrenergic stimuli, focusing on EMT modulation in the 3D culture system of CSs. We treated human CSs and CS-derived cells (CDCs) with the β2-blocker butoxamine (BUT), using either untreated or β2 agonist (clenbuterol) treated CDCs as control. BUT-treated CS-forming cells displayed increased migration capacity and a significant increase in their CS-forming ability, consistently associated with increased expression of EMT-related genes, such as Snai1. Moreover, long-term BUT-treated CDCs contained a lower percentage of CD90+ cells, and this feature has been previously correlated with higher cardiogenic and therapeutic potential of the CDCs population. In addition, long-term BUT-treated CDCs had an increased ratio of collagen-III/collagen-I gene expression levels, and showed decreased release of inflammatory cytokines, overall supporting a less fibrosis-prone phenotype. In conclusion, β2 adrenergic receptor block positively affected the stemness vs commitment balance within CSs through the modulation of type1-EMT (so called "developmental"). These results further highlight type-1 EMT to be a key process affecting the features of resident cardiac progenitor cells, and mediating their response to the microenvironment.
Collapse
Affiliation(s)
- Francesca Pagano
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Francesco Angelini
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Camilla Siciliano
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Julia Tasciotti
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Giorgio Mangino
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Elena De Falco
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Roberto Carnevale
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Sebastiano Sciarretta
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy; Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy
| | - Giacomo Frati
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy; Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy
| | - Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy.
| |
Collapse
|
64
|
Pesce M, Messina E, Chimenti I, Beltrami AP. Cardiac Mechanoperception: A Life-Long Story from Early Beats to Aging and Failure. Stem Cells Dev 2016; 26:77-90. [PMID: 27736363 DOI: 10.1089/scd.2016.0206] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The life-long story of the heart starts concomitantly with primary differentiation events occurring in multipotent progenitors located in the so-called heart tube. This initially tubular structure starts a looping process, which leads to formation of the final four-chambered heart with a primary contribution of geometric and position-associated cell sensing. While this establishes the correct patterning of the final cardiac structure, it also provides feedbacks to fundamental cellular machineries controlling proliferation and differentiation, thus ensuring a coordinated restriction of cell growth and a myocyte terminal differentiation. Novel evidences provided by embryological and cell engineering studies have clarified the relevance of mechanics-supported position sensing for the correct recognition of cell fate inside developing embryos and multicellular aggregates. One of the main components of this pathway, the Hippo-dependent signal transduction machinery, is responsible for cell mechanics intracellular transduction with important consequences for gene transcription and cell growth control. Being the Hippo pathway also directly connected to stress responses and altered metabolism, it is tempting to speculate that permanent alterations of mechanosensing may account for modifying self-renewal control in tissue homeostasis. In the present contribution, we translate these concepts to the aging process and the failing of the human heart, two pathophysiologic conditions that are strongly affected by stress responses and altered metabolism.
Collapse
Affiliation(s)
- Maurizio Pesce
- 1 Tissue Engineering Research Unit, Centro Cardiologico Monzino, IRCCS , Milan, Italy
| | - Elisa Messina
- 2 Department of Pediatric Cardiology, "Sapienza" University , Rome, Italy
| | - Isotta Chimenti
- 3 Department of Medical Surgical Science and Biotechnology, "Sapienza" University , Rome, Italy
| | | |
Collapse
|
65
|
Β-blockers treatment of cardiac surgery patients enhances isolation and improves phenotype of cardiosphere-derived cells. Sci Rep 2016; 6:36774. [PMID: 27841293 PMCID: PMC5107949 DOI: 10.1038/srep36774] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 10/20/2016] [Indexed: 02/08/2023] Open
Abstract
Β-blockers (BB) are a primary treatment for chronic heart disease (CHD), resulting in prognostic and symptomatic benefits. Cardiac cell therapy represents a promising regenerative treatment and, for autologous cell therapy, the patients clinical history may correlate with the biology of resident progenitors and the quality of the final cell product. This study aimed at uncovering correlations between clinical records of biopsy-donor CHD patients undergoing cardiac surgery and the corresponding yield and phenotype of cardiospheres (CSs) and CS-derived cells (CDCs), which are a clinically relevant population for cell therapy, containing progenitors. We describe a statistically significant association between BB therapy and improved CSs yield and CDCs phenotype. We show that BB-CDCs have a reduced fibrotic-like CD90 + subpopulation, with reduced expression of collagen-I and increased expression of cardiac genes, compared to CDCs from non-BB donors. Moreover BB-CDCs had a distinctive microRNA expression profile, consistent with reduced fibrotic features (miR-21, miR-29a/b/c downregulation), and enhanced regenerative potential (miR-1, miR-133, miR-101 upregulation) compared to non-BB. In vitro adrenergic pharmacological treatments confirmed cytoprotective and anti-fibrotic effects of β1-blocker on CDCs. This study shows anti-fibrotic and pro-commitment effects of BB treatment on endogenous cardiac reparative cells, and suggests adjuvant roles of β-blockers in cell therapy applications.
Collapse
|
66
|
Hasan AS, Luo L, Yan C, Zhang TX, Urata Y, Goto S, Mangoura SA, Abdel-Raheem MH, Zhang S, Li TS. Cardiosphere-Derived Cells Facilitate Heart Repair by Modulating M1/M2 Macrophage Polarization and Neutrophil Recruitment. PLoS One 2016; 11:e0165255. [PMID: 27764217 PMCID: PMC5072626 DOI: 10.1371/journal.pone.0165255] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/07/2016] [Indexed: 12/23/2022] Open
Abstract
Cardiosphere-derived cells (CDCs), one of the promising stem cell sources for myocardial repair, have been tested in clinical trials and resulted in beneficial effects; however, the relevant mechanisms are not fully understood. In this study, we examined the hypothesis that CDCs favor heart repair by switching the macrophages from a pro-inflammatory phenotype (M1) into a regulatory anti-inflammatory phenotype (M2). Macrophages from mice were cultured with CDCs-conditioned medium or with fibroblasts-conditioned medium as a control. Immunostaining showed that CDCs-conditioned medium significantly enhanced the expression of CD206 (a marker for M2 macrophages), but decreased the expression of CD86 (a marker for M1 macrophages) 3 days after culture. For animal studies, we used an acute myocardial infarction model of mice. We injected CDCs, fibroblasts, or saline only into the border zone of infarction. Then we collected the heart tissues for histological analysis 5 and 14 days after treatment. Compared with control animals, CDCs treatment significantly decreased M1 macrophages and neutrophils but increased M2 macrophages in the infarcted heart. Furthermore, CDCs-treated mice had reduced infarct size and fewer apoptotic cells compared to the controls. Our data suggest that CDCs facilitate heart repair by modulating M1/M2 macrophage polarization and neutrophil recruitment, which may provide a new insight into the mechanisms of stem cell-based myocardial repair.
Collapse
Affiliation(s)
- Al Shaimaa Hasan
- Department of Stem Cell Biology, Atomic Bomb Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Medical Pharmacology, Qena Faculty of Medicine, South Valley University, Qena, Egypt
| | - Lan Luo
- Department of Stem Cell Biology, Atomic Bomb Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Chen Yan
- Department of Stem Cell Biology, Atomic Bomb Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tian-Xia Zhang
- Department of Stem Cell Biology, Atomic Bomb Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yoshishige Urata
- Department of Stem Cell Biology, Atomic Bomb Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shinji Goto
- Department of Stem Cell Biology, Atomic Bomb Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Safwat A. Mangoura
- Department of Medical Pharmacology, Qena Faculty of Medicine, South Valley University, Qena, Egypt
| | - Mahmoud H. Abdel-Raheem
- Department of Medical Pharmacology, Qena Faculty of Medicine, South Valley University, Qena, Egypt
| | - Shouhua Zhang
- Department of General Surgery, Jiangxi Provincial Children's Hospital, Nanchang, Jiangxi, China
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Institute, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
67
|
Santini MP, Forte E, Harvey RP, Kovacic JC. Developmental origin and lineage plasticity of endogenous cardiac stem cells. Development 2016; 143:1242-58. [PMID: 27095490 DOI: 10.1242/dev.111591] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Over the past two decades, several populations of cardiac stem cells have been described in the adult mammalian heart. For the most part, however, their lineage origins and in vivo functions remain largely unexplored. This Review summarizes what is known about different populations of embryonic and adult cardiac stem cells, including KIT(+), PDGFRα(+), ISL1(+)and SCA1(+)cells, side population cells, cardiospheres and epicardial cells. We discuss their developmental origins and defining characteristics, and consider their possible contribution to heart organogenesis and regeneration. We also summarize the origin and plasticity of cardiac fibroblasts and circulating endothelial progenitor cells, and consider what role these cells have in contributing to cardiac repair.
Collapse
Affiliation(s)
- Maria Paola Santini
- Cardiovascular Research Centre, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Elvira Forte
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst 2010, Australia St Vincent's Clinical School, University of New South Wales, Kensington 2052, Australia Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst 2010, Australia St Vincent's Clinical School, University of New South Wales, Kensington 2052, Australia Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington 2052, Australia
| | - Jason C Kovacic
- Cardiovascular Research Centre, Icahn School of Medicine at Mount Sinai, New York City, NY, USA Stem Cells Australia, Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
68
|
Mayfield AE, Fitzpatrick ME, Latham N, Tilokee EL, Villanueva M, Mount S, Lam BK, Ruel M, Stewart DJ, Davis DR. The impact of patient co-morbidities on the regenerative capacity of cardiac explant-derived stem cells. Stem Cell Res Ther 2016; 7:60. [PMID: 27225482 PMCID: PMC4880978 DOI: 10.1186/s13287-016-0321-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/05/2016] [Accepted: 04/11/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Although patient-sourced cardiac stem cells repair damaged myocardium, the extent to which medical co-morbidities influence cardiac-derived cell products is uncertain. Therefore, we investigated the influence of atherosclerotic risk factors on the regenerative performance of human cardiac explant-derived cells (EDCs). METHODS In this study, the Long Term Stratification for survivors of acute coronary syndromes model was used to quantify the burden of cardiovascular risk factors within a group of patients with established atherosclerosis. EDCs were cultured from human atrial appendages and injected into immunodeficient mice 7 days post-left coronary ligation. Cytokine arrays and enzyme linked immunoassays were used to determine the release of cytokines by EDCs in vitro, and echocardiography was used to determine regenerative capabilities in vivo. RESULTS EDCs sourced from patients with more cardiovascular risk factors demonstrated a negative correlation with production of pro-healing cytokines (such as stromal cell derived factor 1α) and exosomes which had negative effects on the promotion of angiogenesis and chemotaxis. Reductions in exosomes and pro-healing cytokines with accumulating medical co-morbidities were associated with increases in production of the pro-inflammatory cytokine interleukin-6 (IL-6) by EDCs. Increased patient co-morbidities were also correlated with significant attenuation in improvements of left ventricular ejection fraction. CONCLUSIONS The regenerative performance of the earliest precursor cell population cultured from human explant tissue declines with accumulating medical co-morbidities. This effect is associated with diminished production of pro-cardiogenic cytokines and exosomes while IL-6 is markedly increased. Predictors of cardiac events demonstrated a lower capacity to support angiogenesis and repair injured myocardium in a mouse model of myocardial infarction.
Collapse
Affiliation(s)
- Audrey E Mayfield
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | - Megan E Fitzpatrick
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, K1H8L6, Canada
| | - Nicholas Latham
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | - Everad L Tilokee
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | - Melanie Villanueva
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | - Seth Mount
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | - Bu-Khanh Lam
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | - Marc Ruel
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada
| | - Duncan J Stewart
- Ottawa Hospital Research Institute, Division of Regenerative Medicine, Department of Medicine, University of Ottawa, Ottawa, K1H8L6, Canada
| | - Darryl R Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, University of Ottawa, Ottawa, K1Y4W7, Canada.
| |
Collapse
|
69
|
Luo L, Urata Y, Yan C, Hasan AS, Goto S, Guo CY, Tou FF, Xie Y, Li TS. Radiation Exposure Decreases the Quantity and Quality of Cardiac Stem Cells in Mice. PLoS One 2016; 11:e0152179. [PMID: 27195709 PMCID: PMC4873219 DOI: 10.1371/journal.pone.0152179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 03/09/2016] [Indexed: 01/03/2023] Open
Abstract
Radiation exposure may increase cardiovascular disease risks; however, the precise molecular/cellular mechanisms remain unclear. In the present study, we examined the hypothesis that radiation impairs cardiac stem cells (CSCs), thereby contributing to future cardiovascular disease risks. Adult C57BL/6 mice were exposed to 3 Gy γ-rays, and heart tissues were collected 24 hours later for further experiments. Although c-kit-positive cells were rarely found, radiation exposure significantly induced apoptosis and DNA damage in the cells of the heart. The ex vivo expansion of CSCs from freshly harvested atrial tissues showed a significantly lower production of CSCs in irradiated mice compared with healthy mice. The proliferative activity of CSCs evaluated by Ki-67 expression was not significantly different between the groups. However, compared to the healthy control, CSCs expanded from irradiated mice showed significantly lower telomerase activity, more 53BP1 foci in the nuclei, lower expression of c-kit and higher expression of CD90. Furthermore, CSCs expanded from irradiated mice had significantly poorer potency in the production of insulin-like growth factor-1. Our data suggest that radiation exposure significantly decreases the quantity and quality of CSCs, which may serve as sensitive bio-parameters for predicting future cardiovascular disease risks.
Collapse
Affiliation(s)
- Lan Luo
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Yoshishige Urata
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Chen Yan
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Al Shaimaa Hasan
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Shinji Goto
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Chang-Ying Guo
- Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, PR China
| | - Fang-Fang Tou
- Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, PR China
| | - Yucai Xie
- Department of Cardiology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
- * E-mail:
| |
Collapse
|
70
|
Lee JH, Han YS, Lee SH. Long-Duration Three-Dimensional Spheroid Culture Promotes Angiogenic Activities of Adipose-Derived Mesenchymal Stem Cells. Biomol Ther (Seoul) 2016; 24:260-7. [PMID: 26869524 PMCID: PMC4859789 DOI: 10.4062/biomolther.2015.146] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/05/2015] [Accepted: 11/11/2015] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) offer significant therapeutic promise for various regenerative therapies. However, MSC-based therapy for injury exhibits low efficacy due to the pathological environment in target tissues and the differences between in vitro and in vivo conditions. To address this issue, we developed adipose-derived MSC spheroids as a novel delivery method to preserve the stem cell microenvironment. MSC spheroids were generated by suspension culture for 3 days, and their sizes increased in a time-dependent manner. After re-attachment of MSC spheroids to the plastic dish, their adhesion capacity and morphology were not altered. MSC spheroids showed enhanced production of hypoxia-induced angiogenic cytokines such as vascular endothelial growth factor (VEGF), stromal cell derived factor (SDF), and hepatocyte growth factor (HGF). In addition, spheroid culture promoted the preservation of extracellular matrix (ECM) components, such as laminin and fibronectin, in a culture time- and spheroid size-dependent manner. Furthermore, phosphorylation of AKT, a cell survival signal, was significantly higher and the expression of pro-apoptotic molecules, poly (ADP ribose) polymerase-1 (PARP-1) and cleaved caspase-3, was markedly lower in the spheroids than in MSCs in monolayers. In the murine hindlimb ischemia model, transplanted MSC spheroids showed better proliferation than MSCs in monolayer. These findings suggest that MSC spheroids promote MSC bioactivities via secretion of angiogenic cytokines, preservation of ECM components, and regulation of apoptotic signals. Therefore, MSC spheroid-based cell therapy may serve as a simple and effective strategy for regenerative medicine.
Collapse
Affiliation(s)
- Jun Hee Lee
- Laboratory for Vascular Medicine & Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea.,Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea.,Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| |
Collapse
|
71
|
PIWI-interacting RNA (piRNA) signatures in human cardiac progenitor cells. Int J Biochem Cell Biol 2016; 76:1-11. [PMID: 27131603 DOI: 10.1016/j.biocel.2016.04.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 04/01/2016] [Accepted: 04/25/2016] [Indexed: 12/22/2022]
Abstract
Cardiac progenitors, such as cardiospheres and cardiosphere-derived cells, represent an attractive cell source for cardiac regeneration. The PIWI-interacting RNAs, piRNAs, are an intriguing class of small non-coding RNAs, implicated in the regulation of epigenetic state, maintenance of genomic integrity and stem cell functions. Although non-coding RNAs are an exploiting field in cardiovascular research, the piRNA signatures of cardiac progenitors has not been evaluated yet.We profiled, through microarrays, 15,311 piRNAs expressed in cardiospheres, cardiosphere-derived cells and cardiac fibroblasts. Results showed a set of differentially expressed piRNAs (fold change ≥2, p<0.01): 641 piRNAs were upregulated and 1,301 downregulated in the cardiospheres compared to cardiosphere-derived cells, while 255 and 708 piRNAs resulted up- and down-regulated, respectively, if compared to cardiac fibroblasts. We also identified 181 piRNAs that are overexpressed and 129 are downregulated in cardiosphere-derived cells respect to cardiac fibroblasts.Bioinformatics analysis showed that the deregulated piRNAs were mainly distributed on few chromosomes, suggesting that piRNAs are organized in discrete genomic clusters.Furthermore, the bioinformatics search showed that the most upregulated piRNAs target transposons, especially belonged to LINE-1 class, as validated by qRT-PCR. This reduction is also associated to an activation of AKT signaling, which is beneficial for cardiac regeneration.The present study is the first to show a highly consistent piRNA expression pattern for human cardiac progenitors, likely responsible of their different regenerative power. Moreover, this piRNome analysis may provide new methods for characterize cardiac progenitors and may shed new light on the understanding the complex molecular mechanisms of cardiac regeneration.
Collapse
|
72
|
Nakashima Y, Omasa T. What Kind of Signaling Maintains Pluripotency and Viability in Human-Induced Pluripotent Stem Cells Cultured on Laminin-511 with Serum-Free Medium? Biores Open Access 2016; 5:84-93. [PMID: 27096107 PMCID: PMC4834485 DOI: 10.1089/biores.2016.0001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Xeno-free medium contains no animal-derived components, but is composed of minimal growth factors and is serum free; the medium may be supplemented with insulin, transferrin, and selenium (ITS medium). Serum-free and xeno-free culture of human-induced pluripotent stem cells (hiPSCs) uses a variety of components based on ITS medium and Dulbecco's modified Eagle's medium/Ham's nutrient mixture F12 (DMEM/F12) that contain high levels of iron salt and glucose. Culture of hiPSCs also requires scaffolding materials, such as extracellular matrix, collagen, fibronectin, laminin, proteoglycan, and vitronectin. The scaffolding component laminin-511, which is composed of α5, β1, and γ1 chains, binds to α3β1, α6β1, and α6β4 integrins on the cell membrane to induce activation of the PI3K/AKT- and Ras/MAPK-dependent signaling pathways. In hiPSCs, the interaction of laminin-511/α6β1 integrin with the cell–cell adhesion molecule E-cadherin confers protection against apoptosis through the Ras homolog gene family member A (RhoA)/Rho kinase (ROCK) signaling pathway (the major pathways for cell death) and the proto-oncogene tyrosine-protein kinase Fyn (Fyn)-RhoA-ROCK signaling pathway. The expression levels of α6β1 integrin and E-cadherin on cell membranes are controlled through the activation of insulin receptor/insulin, FGF receptor/FGF2, or activin-like kinase 5 (ALK5)-dependent TGF-β signaling. A combination of growth factors, medium constituents, cell membrane-located E-cadherin, and α6β1 integrin-induced signaling is required for pluripotent cell proliferation and for optimal cell survival on a laminin-511 scaffold. In this review, we discuss and explore the influence of growth factors on the cadherin and integrin signaling pathways in serum-free and xeno-free cultures of hiPSCs during the preparation of products for regenerative medicinal therapies. In addition, we suggest the optimum serum-free medium components for use with laminin-511, a new scaffold for hiPSC culture.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Department of Material and Life Science, Graduate School of Engineering, Osaka University , Osaka, Japan
| | - Takeshi Omasa
- Department of Material and Life Science, Graduate School of Engineering, Osaka University , Osaka, Japan
| |
Collapse
|
73
|
Angelini F, Ionta V, Rossi F, Miraldi F, Messina E, Giacomello A. Foetal bovine serum-derived exosomes affect yield and phenotype of human cardiac progenitor cell culture. ACTA ACUST UNITED AC 2016; 6:15-24. [PMID: 27340620 PMCID: PMC4916547 DOI: 10.15171/bi.2016.03] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/29/2016] [Accepted: 03/05/2016] [Indexed: 12/19/2022]
Abstract
![]()
Introduction: Cardiac progenitor cells (CPCs) represent a powerful tool in cardiac regenerative medicine. Pre-clinical studies suggest that most of the beneficial effects promoted by the injected cells are due to their paracrine activity exerted on endogenous cells and tissue. Exosomes are candidate mediators of this paracrine effects. According to their potential, many researchers have focused on characterizing exosomes derived from specific cell types, but, up until now, only few studies have analyzed the possible in vitro effects of bovine serum-derived exosomes on cell proliferation or differentiation.
Methods: The aim of this study was to analyse, from a qualitative and quantitative point of view, the in vitro effects of bovine serum exosomes on human CPCs cultured either as cardiospheres or as monolayers of cardiosphere-forming cells.
Results: Effects on proliferation, yield and molecular patterning were detected. We show, for the first time, that exogenous bovine exosomes support the proliferation and migration of human cardiosphere-forming cells, and that their depletion affects cardiospheres formation, in terms of size, yield and extra-cellular matrix production.
Conclusion: These results stress the importance of considering differential biological effects of exogenous cell culture supplements on the final phenotype of primary human cell cultures.
Collapse
Affiliation(s)
- Francesco Angelini
- Pasteur Institute - Cenci Bolognetti Foundation, "Sapienza" University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Vittoria Ionta
- Department of Molecular Medicine, "Sapienza" University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Fabrizio Rossi
- Department of Molecular Medicine, "Sapienza" University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Fabio Miraldi
- Department of Cardiocirculatory Pathophysiology, Anesthesiology and General Surgery, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Elisa Messina
- Department of Pediatric Cardiology, "Sapienza" University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Alessandro Giacomello
- Department of Molecular Medicine, "Sapienza" University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
74
|
Microtissues in Cardiovascular Medicine: Regenerative Potential Based on a 3D Microenvironment. Stem Cells Int 2016; 2016:9098523. [PMID: 27073399 PMCID: PMC4814701 DOI: 10.1155/2016/9098523] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/01/2016] [Accepted: 02/21/2016] [Indexed: 02/06/2023] Open
Abstract
More people die annually from cardiovascular diseases than from any other cause. In particular, patients who suffer from myocardial infarction may be affected by ongoing adverse remodeling processes of the heart that may ultimately lead to heart failure. The introduction of stem and progenitor cell-based applications has raised substantial hope for reversing these processes and inducing cardiac regeneration. However, current stem cell therapies using single-cell suspensions have failed to demonstrate long-lasting efficacy due to the overall low retention rate after cell delivery to the myocardium. To overcome this obstacle, the concept of 3D cell culture techniques has been proposed to enhance therapeutic efficacy and cell engraftment based on the simulation of an in vivo-like microenvironment. Of great interest is the use of so-called microtissues or spheroids, which have evolved from their traditional role as in vitro models to their novel role as therapeutic agents. This review will provide an overview of the therapeutic potential of microtissues by addressing primarily cardiovascular regeneration. It will accentuate their advantages compared to other regenerative approaches and summarize the methods for generating clinically applicable microtissues. In addition, this review will illustrate the unique properties of the microenvironment within microtissues that makes them a promising next-generation therapeutic approach.
Collapse
|
75
|
Nakamura T, Hosoyama T, Kawamura D, Takeuchi Y, Tanaka Y, Samura M, Ueno K, Nishimoto A, Kurazumi H, Suzuki R, Ito H, Sakata K, Mikamo A, Li TS, Hamano K. Influence of aging on the quantity and quality of human cardiac stem cells. Sci Rep 2016; 6:22781. [PMID: 26947751 PMCID: PMC4780032 DOI: 10.1038/srep22781] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/19/2016] [Indexed: 01/01/2023] Open
Abstract
Advanced age affects various tissue-specific stem cells and decreases their regenerative ability. We therefore examined whether aging affected the quantity and quality of cardiac stem cells using cells obtained from 26 patients of various ages (from 2 to 83 years old). We collected fresh right atria and cultured cardiosphere-derived cells (CDCs), which are a type of cardiac stem cell. Then we investigated growth rate, senescence, DNA damage, and the growth factor production of CDCs. All samples yielded a sufficient number of CDCs for experiments and the cellular growth rate was not obviously associated with age. The expression of senescence-associated b-galactosidase and the DNA damage marker, gH2AX, showed a slightly higher trend in CDCs from older patients (≥65 years). The expression of VEGF, HGF, IGF-1, SDF-1, and TGF-b varied among samples, and the expression of these beneficial factors did not decrease with age. An in vitro angiogenesis assay also showed that the angiogenic potency of CDCs was not impaired, even in those from older patients. Our data suggest that the impact of age on the quantity and quality of CDCs is quite limited. These findings have important clinical implications for autologous stem cell transplantation in elderly patients.
Collapse
Affiliation(s)
- Tamami Nakamura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Tohru Hosoyama
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan.,Center for Regenerative Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Daichi Kawamura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Yuriko Takeuchi
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Yuya Tanaka
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Makoto Samura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Koji Ueno
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan.,Center for Regenerative Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Arata Nishimoto
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Hiroshi Kurazumi
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Ryo Suzuki
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Hiroshi Ito
- Department of Cardiovascular Surgery, Saiseikai Shimonoseki General Hospital, 8-5-1 Yasuoka, Shimonoseki, Yamaguchi 759-6603, Japan
| | - Kensuke Sakata
- Department of Cardiovascular Surgery, Saiseikai Shimonoseki General Hospital, 8-5-1 Yasuoka, Shimonoseki, Yamaguchi 759-6603, Japan
| | - Akihito Mikamo
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-kogushi, Yamaguchi, Ube 755-8505, Japan
| |
Collapse
|
76
|
Gallet R, Tseliou E, Dawkins J, Middleton R, Valle J, Angert D, Reich H, Luthringer D, Kreke M, Smith R, Marbán L, Marbán E. Intracoronary delivery of self-assembling heart-derived microtissues (cardiospheres) for prevention of adverse remodeling in a pig model of convalescent myocardial infarction. Circ Cardiovasc Interv 2016; 8:CIRCINTERVENTIONS.115.002391. [PMID: 25953823 DOI: 10.1161/circinterventions.115.002391] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Preclinical studies in rodents and pigs indicate that the self-assembling microtissues known as cardiospheres may be more effective than dispersed cardiosphere-derived cells. However, the more desirable intracoronary route has been assumed to be unsafe for cardiosphere delivery: Cardiospheres are large (30-150 μm), raising concerns about likely microembolization. We questioned these negative assumptions by evaluating the safety and efficacy of optimized intracoronary delivery of cardiospheres in a porcine model of convalescent myocardial infarction. METHODS AND RESULTS First, we standardized the size of cardiospheres by modifying culture conditions. Then, dosage was determined by infusing escalating doses of cardiospheres in the left anterior descending artery of naive pigs, looking for acute adverse effects. Finally, in a randomized efficacy study, 14 minipigs received allogeneic cardiospheres (1.3 × 10(6)) or vehicle 1 month after myocardial infarction. Animals underwent magnetic resonance imaging before infusion and 1 month later to assess left ventricular ejection fraction, scar mass, and viable mass. In the dosing study, we did not observe any evidence of microembolization after cardiosphere infusion. In the post-myocardial infarction study, cardiospheres preserved LV function, reduced scar mass and increased viable mass, whereas placebo did not. Moreover, cardiosphere decreased collagen content, and increased vessel densities and myocardial perfusion. Importantly, intracoronary cardiospheres decreased left ventricular end-diastolic pressure and increased cardiac output. CONCLUSIONS Intracoronary delivery of cardiospheres is safe. Intracoronary cardiospheres are also remarkably effective in decreasing scar, halting adverse remodeling, increasing myocardial perfusion, and improving hemodynamic status after myocardial infarction in pigs. Thus, cardiospheres may be viable therapeutic candidates for intracoronary infusion in selected myocardial disorders.
Collapse
Affiliation(s)
- Romain Gallet
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.)
| | - Eleni Tseliou
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.)
| | - James Dawkins
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.)
| | - Ryan Middleton
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.)
| | - Jackelyn Valle
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.)
| | - David Angert
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.)
| | - Heidi Reich
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.)
| | - Daniel Luthringer
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.)
| | - Michelle Kreke
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.)
| | - Rachel Smith
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.)
| | - Linda Marbán
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.)
| | - Eduardo Marbán
- From the Cedars-Sinai Heart Institute, Los Angeles, CA (R.G., E.T., J.D., R.M., J.V., D.A., H.R., D.L., M.K., R.S., L.M.); and Capricor Inc, Los Angeles, CA (M.K., R.S., L.M.).
| |
Collapse
|
77
|
Vahdat S, Mousavi SA, Omrani G, Gholampour M, Sotoodehnejadnematalahi F, Ghazizadeh Z, Gharechahi J, Baharvand H, Salekdeh GH, Aghdami N. Cellular and molecular characterization of human cardiac stem cells reveals key features essential for their function and safety. Stem Cells Dev 2016; 24:1390-404. [PMID: 25867933 DOI: 10.1089/scd.2014.0222] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell therapy of heart diseases is emerging as one of the most promising known treatments in recent years. Transplantation of cardiac stem cells (CSCs) may be one of the best strategies to cure adult or pediatric heart diseases. As these patient-derived stem cells need to be isolated from small heart biopsies, it is important to select the best isolation method and CSC subpopulation with the best cardiogenic functionality. We employed three different protocols including c-KIT(+) cell sorting, clonogenic expansion, and explants culture to isolate c-KIT(+) cells, clonogenic expansion-derived cells (CEDCs), and cardiosphere-derived cells (CDCs), respectively. Evaluation of isolated CSC characteristics in vitro and after rat myocardial infarction (MI) model transplantation revealed that although c-KIT(+) and CDCs had higher MI regenerative potential, CEDCs had more commitment into cardiomyocytes and needed lower passages that were essential to reach a definite cell count. Furthermore, genome-wide expression analysis showed that subsequent passages caused changes in characteristics of cells, downregulation of cell cycle-related genes, and upregulation of differentiation and carcinogenic genes, which might lead to senescence, commitment, and possible tumorigenicity of the cells. Because of different properties of CSC subpopulations, we suggest that appropriate CSCs subpopulation should be chosen based on their experimental or clinical use.
Collapse
Affiliation(s)
- Sadaf Vahdat
- 1Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,2Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Seyed Ahmad Mousavi
- 3Department of Molecular Systems Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Gholamreza Omrani
- 4Department of Cardiac Surgery, Rajaei Cardiovascular Medical Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Maziar Gholampour
- 4Department of Cardiac Surgery, Rajaei Cardiovascular Medical Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Fattah Sotoodehnejadnematalahi
- 1Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zaniar Ghazizadeh
- 1Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Javad Gharechahi
- 3Department of Molecular Systems Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- 1Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,5Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Ghasem Hosseini Salekdeh
- 3Department of Molecular Systems Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,6Department of Systems Biology, Agricultural Biotechnology Research Institute of Iran, Karaj, Iran
| | - Nasser Aghdami
- 1Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,7Department of Regenerative Biomedicine at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
78
|
Mount S, Davis DR. Electrical effects of stem cell transplantation for ischaemic cardiomyopathy: friend or foe? J Physiol 2016; 594:2511-24. [PMID: 26584682 DOI: 10.1113/jp270540] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/18/2015] [Indexed: 01/07/2023] Open
Abstract
Despite advances in other realms of cardiac care, the mortality attributable to ischaemic cardiomyopathy has only marginally decreased over the last 10 years. These findings highlight the growing realization that current pharmacological and device therapies rarely reverse disease progression and rationalize a focus on novel means to reverse, repair and re-vascularize damaged hearts. As such, multiple candidate cell types have been used to regenerate damaged hearts either directly (through differentiation to form new tissue) or indirectly (via paracrine effects). Emerging literature suggests that robust engraftment of electrophysiolgically heterogeneous tissue from transplanted cells comes at the cost of a high incidence of ventricular arrhythmias. Similar electrophysiological studies of haematological stem cells raised early concerns that transplant of depolarized, inexcitable cells that also induce paracrine-mediated electrophysiological remodelling may be pro-arrhythmic. However, meta-analyses suggest that patients receiving haematological stem cells paradoxically may experience a decrease in ventricular arrhythmias, an observation potentially related to the extremely poor long-term survival of injected cells. Finally, early clinical and preclinical data from technologies capable of differentiating to a mature cardiomyocyte phenotype (such as cardiac-derived stem cells) suggests that these cells are not pro-arrhythmic although they too lack robust long-term engraftment. These results highlight the growing understanding that as next generation cell therapies are developed, emphasis should also be placed on understanding possible anti-arrhythmic contributions of transplanted cells while vigilance is needed to predict and treat the inadvertent effects of regenerative cell therapies on the electrophysiological stability of the ischaemic cardiomyopathic heart.
Collapse
Affiliation(s)
- Seth Mount
- University of Ottawa Heart Institute, Ottawa, Canada, K1Y 4W7
| | - Darryl R Davis
- University of Ottawa Heart Institute, Ottawa, Canada, K1Y 4W7
| |
Collapse
|
79
|
Widespread Myocardial Delivery of Heart-Derived Stem Cells by Nonocclusive Triple-Vessel Intracoronary Infusion in Porcine Ischemic Cardiomyopathy: Superior Attenuation of Adverse Remodeling Documented by Magnetic Resonance Imaging and Histology. PLoS One 2016; 11:e0144523. [PMID: 26784932 PMCID: PMC4718597 DOI: 10.1371/journal.pone.0144523] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/19/2015] [Indexed: 12/26/2022] Open
Abstract
Single-vessel, intracoronary infusion of stem cells under stop-flow conditions has proven safe but achieves only limited myocardial coverage. Continuous flow intracoronary delivery to one or more coronary vessels may achieve broader coverage for treating cardiomyopathy, but has not been investigated. Using nonocclusive coronary guiding catheters, we infused allogeneic cardiosphere-derived cells (CDCs) either in a single vessel or sequentially in all three coronary arteries in porcine ischemic cardiomyopathy and used magnetic resonance imaging (MRI) to assess structural and physiological outcomes. Vehicle-infused animals served as controls. Single-vessel stop-flow and continuous-flow intracoronary infusion revealed equivalent effects on scar size and function. Sequential infusion into each of the three major coronary vessels under stop-flow or continuous-flow conditions revealed equal efficacy, but less elevation of necrotic biomarkers with continuous-flow delivery. In addition, multi-vessel delivery resulted in enhanced global and regional tissue function compared to a triple-vessel placebo-treated group. The functional benefits after global cell infusion were accompanied histologically by minimal inflammatory cellular infiltration, attenuated regional fibrosis and enhanced vessel density in the heart. Sequential multi-vessel non-occlusive delivery of CDCs is safe and provides enhanced preservation of left ventricular function and structure. The current findings provide preclinical validation of the delivery method currently undergoing clinical testing in the Dilated cardiomYopathy iNtervention With Allogeneic MyocardIally-regenerative Cells (DYNAMIC) trial of CDCs in heart failure patients.
Collapse
|
80
|
Teichroeb JH, Kim J, Betts DH. The role of telomeres and telomerase reverse transcriptase isoforms in pluripotency induction and maintenance. RNA Biol 2016; 13:707-19. [PMID: 26786236 DOI: 10.1080/15476286.2015.1134413] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Telomeres are linear guanine-rich DNA structures at the ends of chromosomes. The length of telomeric DNA is actively regulated by a number of mechanisms in highly proliferative cells such as germ cells, cancer cells, and pluripotent stem cells. Telomeric DNA is synthesized by way of the ribonucleoprotein called telomerase containing a reverse transcriptase (TERT) subunit and RNA component (TERC). TERT is highly conserved across species and ubiquitously present in their respective pluripotent cells. Recent studies have uncovered intricate associations between telomeres and the self-renewal and differentiation properties of pluripotent stem cells. Interestingly, the past decade's work indicates that the TERT subunit also has the capacity to modulate mitochondrial function, to remodel chromatin structure, and to participate in key signaling pathways such as the Wnt/β-catenin pathway. Many of these non-canonical functions do not require TERT's catalytic activity, which hints at possible functions for the extensive number of alternatively spliced TERT isoforms that are highly expressed in pluripotent stem cells. In this review, some of the established and potential routes of pluripotency induction and maintenance are highlighted from the perspectives of telomere maintenance, known TERT isoform functions and their complex regulation.
Collapse
Affiliation(s)
- Jonathan H Teichroeb
- a Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry , The University of Western Ontario , London , Ontario , Canada
| | - Joohwan Kim
- a Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry , The University of Western Ontario , London , Ontario , Canada
| | - Dean H Betts
- a Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry , The University of Western Ontario , London , Ontario , Canada.,b Children's Health Research Institute, Lawson Health Research Institute , London , Ontario , Canada
| |
Collapse
|
81
|
Kapelios CJ, Nanas JN, Malliaras K. Allogeneic cardiosphere-derived cells for myocardial regeneration: current progress and recent results. Future Cardiol 2016; 12:87-100. [DOI: 10.2217/fca.15.72] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Early-phase clinical testing of autologous cardiosphere-derived cells (CDCs) has yielded intriguing results, consistent with therapeutic myocardial regeneration. However, autologous therapy is associated with significant technical, timing, economic and logistic constraints, prompting researchers to explore the potential of allogeneic CDC therapy. CDCs exhibit a favorable immunologic antigenic profile and are hypoimmunogenic in vitro. Preclinical studies in immunologically mismatched animals demonstrate that allogeneic CDC transplantation without immunosuppression is safe and produces sustained functional and structural benefits through stimulation of endogenous regenerative pathways. Currently, allogeneic human CDCs are being tested clinically in the ALLSTAR and DYNAMIC trials. Potential establishment of clinical safety and efficacy of allogeneic CDCs combined with generation of highly standardized, ‘off-the-shelf’ allogeneic cellular products would facilitate broad clinical adoption of cell therapy.
Collapse
Affiliation(s)
- Chris J Kapelios
- 3rd Department of Cardiology, University of Athens School of Medicine, 67 Mikras Asias Street, 11 527, Athens, Greece
| | - John N Nanas
- 3rd Department of Cardiology, University of Athens School of Medicine, 67 Mikras Asias Street, 11 527, Athens, Greece
| | - Konstantinos Malliaras
- 3rd Department of Cardiology, University of Athens School of Medicine, 67 Mikras Asias Street, 11 527, Athens, Greece
| |
Collapse
|
82
|
JUDD J, XUAN W, HUANG GN. Cellular and molecular basis of cardiac regeneration. Turk J Biol 2016. [DOI: 10.3906/biy-1504-43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
83
|
Hosoyama T, Samura M, Kudo T, Nishimoto A, Ueno K, Murata T, Ohama T, Sato K, Mikamo A, Yoshimura K, Li TS, Hamano K. Cardiosphere-derived cell sheet primed with hypoxia improves left ventricular function of chronically infarcted heart. Am J Transl Res 2015; 7:2738-2751. [PMID: 26885271 PMCID: PMC4731671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/27/2015] [Indexed: 06/05/2023]
Abstract
Cardiosphere-derived cells (CDCs) isolated from postnatal heart tissue are a convenient and efficientresource for the treatment of myocardial infarction. However, poor retention of CDCs in infarcted hearts often causes less than ideal therapeutic outcomes. Cell sheet technology has been developed as a means of permitting longer retention of graft cells, and this therapeutic strategy has opened new avenues of cell-based therapy for severe ischemic diseases. However, there is still scope for improvement before this treatment can be routinely applied in clinical settings. In this study, we investigated whether hypoxic preconditioning enhances the therapeutic efficacy of CDC monolayer sheets. To induce hypoxia priming, CDC monolayer sheets were placed in an incubator adjusted to 2% oxygen for 24 hours, and then preconditioned mouse CDC sheets were implanted into the infarcted heart of old myocardial infarction mouse models. Hypoxic preconditioning of CDC sheets remarkably increased the expression of vascular endothelial growth factor through the PI3-kinase/Akt signaling pathway. Implantation of preconditioned CDC sheets improved left ventricular function inchronically infarcted hearts and reduced fibrosis. The therapeutic efficacy of preconditioned CDC sheets was higher than the CDC sheets that were cultured under normaxia condition. These results suggest that hypoxic preconditioning augments the therapeutic angiogenic and anti-fibrotic activity of CDC sheets. A combination of cell sheets and hypoxic preconditioning offers an attractive therapeutic protocol for CDC transplantation into chronically infarcted hearts.
Collapse
Affiliation(s)
- Tohru Hosoyama
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of MedicineUbe, Japan
- Center for Regenerative Medicine, Yamaguchi University Graduate School of MedicineUbe, Japan
| | - Makoto Samura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of MedicineUbe, Japan
| | - Tomoaki Kudo
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of MedicineUbe, Japan
| | - Arata Nishimoto
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of MedicineUbe, Japan
| | - Koji Ueno
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of MedicineUbe, Japan
- Center for Regenerative Medicine, Yamaguchi University Graduate School of MedicineUbe, Japan
| | - Tomoaki Murata
- Institute of Laboratory Animals, Yamaguchi University Graduate School of MedicineUbe, Japan
| | - Takashi Ohama
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi UniversityYamaguchi, Japan
| | - Koichi Sato
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi UniversityYamaguchi, Japan
| | - Akihito Mikamo
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of MedicineUbe, Japan
| | - Koichi Yoshimura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of MedicineUbe, Japan
- Graduate School of Health and Welfare, Yamaguchi Prefectural UniversityYamaguchi, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki UniversityNagasaki, Japan
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of MedicineUbe, Japan
| |
Collapse
|
84
|
Xie Y, Ibrahim A, Cheng K, Wu Z, Liang W, Malliaras K, Sun B, Liu W, Shen D, Cheol Cho H, Li T, Lu L, Lu G, Marbán E. Importance of cell-cell contact in the therapeutic benefits of cardiosphere-derived cells. Stem Cells 2015; 32:2397-406. [PMID: 24802280 DOI: 10.1002/stem.1736] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 03/19/2014] [Accepted: 04/04/2014] [Indexed: 12/30/2022]
Abstract
Cardiosphere-derived cells (CDCs) effect therapeutic regeneration after myocardial infarction (MI) both in animal models and in humans. Here, we test the hypothesis that cell-cell contact plays a role in mediating the observed therapeutic benefits of CDCs, above and beyond conventional paracrine effects. Human CDCs or vehicle were injected into immunodeficient (SCID) mouse hearts during acute MI. CDC transplantation augmented the proportion of cycling (Ki67(+) ) cardiomyocytes and improved ventricular function. CDC-conditioned media only modestly augmented the percentage of Ki67(+) cardiomyocytes (>control but <CDCs), but did not improve pump function. When neonatal rat ventricular myocytes (NRVMs) were cocultured with human CDCs in vitro, the percentage of cycling NRVMs (Ki67(+) or BrdU(+) nuclei) increased relative to solitary NRVM culture. To further dissect the relative contributions of soluble factors versus contact-dependent mechanisms, we compared CDCs grown with NRVMs in a transwell contact-free system versus admixed coculture. The percentage of cycling NRVMs was higher in admixed coculture than in the contact-free system. Pretreatment with inhibitors of MEK and PI3K, or with β1 integrin neutralizing antibody, blocked the ability of CDCs to promote myocyte cycling. While conditioned media are not inert, direct apposition of CDCs to cardiomyocytes produces greater enhancement of cardiomyocyte proliferation in vitro and in vivo, and improves function post-MI. Intact cardiomyocyte β1 integrin signaling is necessary for the contact-dependent cardioproliferative effects of CDCs.
Collapse
Affiliation(s)
- Yucai Xie
- Cedars-Sinai Heart Institute, Los Angeles, California, USA; Department of Cardiology, Shanghai Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Zuppinger C. 3D culture for cardiac cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1873-81. [PMID: 26658163 DOI: 10.1016/j.bbamcr.2015.11.036] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/23/2015] [Accepted: 11/30/2015] [Indexed: 01/26/2023]
Abstract
This review discusses historical milestones, recent developments and challenges in the area of 3D culture models with cardiovascular cell types. Expectations in this area have been raised in recent years, but more relevant in vitro research, more accurate drug testing results, reliable disease models and insights leading to bioartificial organs are expected from the transition to 3D cell culture. However, the construction of organ-like cardiac 3D models currently remains a difficult challenge. The heart consists of highly differentiated cells in an intricate arrangement.Furthermore, electrical “wiring”, a vascular system and multiple cell types act in concert to respond to the rapidly changing demands of the body. Although cardiovascular 3D culture models have been predominantly developed for regenerative medicine in the past, their use in drug screening and for disease models has become more popular recently. Many sophisticated 3D culture models are currently being developed in this dynamic area of life science. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Christian Zuppinger
- Cardiology, Bern University Hospital, Department of Clinical Research, MEM G803b, Murtenstrasse 35, CH-3008, Bern, Switzerland.
| |
Collapse
|
86
|
Siciliano C, Chimenti I, Ibrahim M, Napoletano C, Mangino G, Scaletta G, Zoccai GB, Rendina EA, Calogero A, Frati G, De Falco E. Cardiosphere Conditioned Media Influence the Plasticity of Human Mediastinal Adipose Tissue-Derived Mesenchymal Stem Cells. Cell Transplant 2015; 24:2307-22. [PMID: 26531290 DOI: 10.3727/096368914x685771] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Nowadays, cardiac regenerative medicine is facing many limitations because of the complexity to find the most suitable stem cell source and to understand the regenerative mechanisms involved. Mesenchymal stem cells (MSCs) have shown great regenerative potential due to their intrinsic properties and ability to restore cardiac functionality, directly by transdifferentiation and indirectly by paracrine effects. Yet, how MSCs could respond to definite cardiac-committing microenvironments, such as that created by resident cardiac progenitor cells in the form of cardiospheres (CSs), has never been addressed. Recently, a putative MSC pool has been described in the mediastinal fat (hmADMSCs), but both its biology and function remain hitherto unexplored. Accordingly, we investigated the potential of hmADMSCs to be committed toward a cardiovascular lineage after preconditioning with CS-conditioned media (CCM). Results indicated that CCM affects cell proliferation. Gene expression levels of multiple cardiovascular and stemness markers (MHC, KDR, Nkx2.5, Thy-1, c-kit, SMA) are significantly modulated, and the percentage of hmADMSCs preconditioned with CCM and positive for Nkx2.5, MHC, and KDR is significantly higher relative to FBS and explant-derived cell conditioned media (EDCM, the unselected stage before CS formation). Growth factor-specific and survival signaling pathways (i.e., Erk1/2, Akt, p38, mTOR, p53) present in CCM are all equally regulated. Nonetheless, earlier BAD phosphorylation (Ser112) occurs associated with the CS microenvironment (and to a lesser extent to EDCM), whereas faster phosphorylation of PRAS40 in FBS, and of Akt (Ser473) in EDCM and 5-azacytidine occurs compared to CCM. For the first time, we demonstrated that the MSC pool held in the mediastinal fat is adequately plastic to partially differentiate in vitro toward a cardiac-like lineage. Besides, we have provided novel evidence of the potent inductive niche-like microenvironment that the CS structure can reproduce in vitro. hmADMSCs can represent an interesting tool in order to exploit their possible role in cardiovascular diseases and treatment.
Collapse
Affiliation(s)
- Camilla Siciliano
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Isotta Chimenti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Mohsen Ibrahim
- Division of Thoracic Surgery, Department of Medical-Surgical Science and Translational Medicine, Sapienza University of Rome, S. Andrea Hospital, Rome, Italy
| | - Chiara Napoletano
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giorgio Mangino
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Gaia Scaletta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Giuseppe Biondi Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Erino Angelo Rendina
- Division of Thoracic Surgery, Department of Medical-Surgical Science and Translational Medicine, Sapienza University of Rome, S. Andrea Hospital, Rome, Italy
| | - Antonella Calogero
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy
| | - Elena De Falco
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
87
|
Hasan A, Khattab A, Islam MA, Hweij KA, Zeitouny J, Waters R, Sayegh M, Hossain MM, Paul A. Injectable Hydrogels for Cardiac Tissue Repair after Myocardial Infarction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2015; 2:1500122. [PMID: 27668147 PMCID: PMC5033116 DOI: 10.1002/advs.201500122] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 06/09/2015] [Indexed: 05/17/2023]
Abstract
Cardiac tissue damage due to myocardial infarction (MI) is one of the leading causes of mortality worldwide. The available treatments of MI include pharmaceutical therapy, medical device implants, and organ transplants, all of which have severe limitations including high invasiveness, scarcity of donor organs, thrombosis or stenosis of devices, immune rejection, and prolonged hospitalization time. Injectable hydrogels have emerged as a promising solution for in situ cardiac tissue repair in infarcted hearts after MI. In this review, an overview of various natural and synthetic hydrogels for potential application as injectable hydrogels in cardiac tissue repair and regeneration is presented. The review starts with brief discussions about the pathology of MI, its current clinical treatments and their limitations, and the emergence of injectable hydrogels as a potential solution for post MI cardiac regeneration. It then summarizes various hydrogels, their compositions, structures and properties for potential application in post MI cardiac repair, and recent advancements in the application of injectable hydrogels in treatment of MI. Finally, the current challenges associated with the clinical application of injectable hydrogels to MI and their potential solutions are discussed to help guide the future research on injectable hydrogels for translational therapeutic applications in regeneration of cardiac tissue after MI.
Collapse
Affiliation(s)
- Anwarul Hasan
- Center for Biomedical Engineering Department of Medicine Brigham and Women's Hospital Harvard Medical School Cambridge MA 02139 USA; Harvard-MIT Division of Health Sciences and Technology Massachusetts Institute of Technology Cambridge MA 02139 USA; Biomedical Engineering and Department of Mechanical Engineering Faculty of Engineering and Architecture American University of Beirut Beirut 1107 2020 Lebanon
| | - Ahmad Khattab
- Department of Electrical and Computer Engineering Faculty of Engineering and Architecture American University of Beirut Beirut 1107 2020 Lebanon
| | - Mohammad Ariful Islam
- Laboratory of Nanomedicine and Biomaterials Department of Anesthesiology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA; Laboratory for Nanoengineering and Drug Delivery Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
| | - Khaled Abou Hweij
- Department of Mechanical Engineering Faculty of Engineering and Architecture American University of Beirut Beirut 1107 2020 Lebanon
| | - Joya Zeitouny
- Department of Electrical and Computer Engineering Faculty of Engineering and Architecture American University of Beirut Beirut 1107 2020 Lebanon
| | - Renae Waters
- BioIntel Research Laboratory Department of Chemical and Petroleum Engineering Bioengineering Graduate Program School of Engineering University of Kansas Lawrence KS 66045 USA
| | | | - Md Monowar Hossain
- Department of Medicine Lyell McEwin Hospital University of Adelaide South Australia 5112 Australia
| | - Arghya Paul
- BioIntel Research Laboratory Department of Chemical and Petroleum Engineering Bioengineering Graduate Program School of Engineering University of Kansas Lawrence KS 66045 USA
| |
Collapse
|
88
|
PEDF and 34-mer inhibit angiogenesis in the heart by inducing tip cells apoptosis via up-regulating PPAR-γ to increase surface FasL. Apoptosis 2015; 21:60-8. [DOI: 10.1007/s10495-015-1186-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
89
|
Valiente-Alandi I, Albo-Castellanos C, Herrero D, Arza E, Garcia-Gomez M, Segovia JC, Capecchi M, Bernad A. Cardiac Bmi1(+) cells contribute to myocardial renewal in the murine adult heart. Stem Cell Res Ther 2015; 6:205. [PMID: 26503423 PMCID: PMC4620653 DOI: 10.1186/s13287-015-0196-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/15/2015] [Accepted: 10/02/2015] [Indexed: 12/19/2022] Open
Abstract
Introduction The mammalian adult heart maintains a continuous, low cardiomyocyte turnover rate throughout life. Although many cardiac stem cell populations have been studied, the natural source for homeostatic repair has not yet been defined. The Polycomb protein BMI1 is the most representative marker of mouse adult stem cell systems. We have evaluated the relevance and role of cardiac Bmi1+ cells in cardiac physiological homeostasis. Methods Bmi1CreER/+;Rosa26YFP/+ (Bmi1-YFP) mice were used for lineage tracing strategy. After tamoxifen (TM) induction, yellow fluorescent protein (YFP) is expressed under the control of Rosa26 regulatory sequences in Bmi1+ cells. These cells and their progeny were tracked by FACS, immunofluorescence and RT-qPCR techniques from 5 days to 1 year. Results FACS analysis of non-cardiomyocyte compartment from TM-induced Bmi1-YFP mice showed a Bmi1+-expressing cardiac progenitor cell (Bmi1-CPC: B-CPC) population, SCA-1 antigen-positive (95.9 ± 0.4 %) that expresses some stemness-associated genes. B-CPC were also able to differentiate in vitro to the three main cardiac lineages. Pulse-chase analysis showed that B-CPC remained quite stable for extended periods (up to 1 year), which suggests that this Bmi1+ population contains cardiac progenitors with substantial self-maintenance potential. Specific immunostaining of Bmi1-YFP hearts serial sections 5 days post-TM induction indicated broad distribution of B-CPC, which were detected in variably sized clusters, although no YFP+ cardiomyocytes (CM) were detected at this time. Between 2 to 12 months after TM induction, YFP+ CM were clearly identified (3 ± 0.6 % to 6.7 ± 1.3 %) by immunohistochemistry of serial sections and by flow cytometry of total freshly isolated CM. B-CPC also contributed to endothelial and smooth muscle (SM) lineages in vivo. Conclusions High Bmi1 expression identifies a non-cardiomyocyte resident cardiac population (B-CPC) that contributes to the main lineages of the heart in vitro and in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0196-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Iñigo Valiente-Alandi
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Carmen Albo-Castellanos
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,Vivebiotech, San Sebastian, Spain.
| | - Diego Herrero
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.
| | - Elvira Arza
- Microscopy Unit, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.
| | - Maria Garcia-Gomez
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)- Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain.
| | - José C Segovia
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)- Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain.
| | - Mario Capecchi
- Howard Hughes Medical Institute University of Utah, Salt Lake City, UT, USA.
| | - Antonio Bernad
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.
| |
Collapse
|
90
|
The potential of GMP-compliant platelet lysate to induce a permissive state for cardiovascular transdifferentiation in human mediastinal adipose tissue-derived mesenchymal stem cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:162439. [PMID: 26495284 PMCID: PMC4606096 DOI: 10.1155/2015/162439] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/04/2015] [Accepted: 06/10/2015] [Indexed: 01/13/2023]
Abstract
Human adipose tissue-derived mesenchymal stem cells (ADMSCs) are considered eligible candidates for cardiovascular stem cell therapy applications due to their cardiac transdifferentiation potential and immunotolerance. Over the years, the in vitro culture of ADMSCs by platelet lysate (PL), a hemoderivate containing numerous growth factors and cytokines derived from platelet pools, has allowed achieving a safe and reproducible methodology to obtain high cell yield prior to clinical administration. Nevertheless, the biological properties of PL are still to be fully elucidated. In this brief report we show the potential ability of PL to induce a permissive state of cardiac-like transdifferentiation and to cause epigenetic modifications. RTPCR results indicate an upregulation of Cx43, SMA, c-kit, and Thy-1 confirmed by immunofluorescence staining, compared to standard cultures with foetal bovine serum. Moreover, PL-cultured ADMSCs exhibit a remarkable increase of both acetylated histones 3 and 4, with a patient-dependent time trend, and methylation at lysine 9 on histone 3 preceding the acetylation. Expression levels of p300 and SIRT-1, two major regulators of histone 3, are also upregulated after treatment with PL. In conclusion, PL could unravel novel biological properties beyond its routine employment in noncardiac applications, providing new insights into the plasticity of human ADMSCs.
Collapse
|
91
|
Oltolina F, Zamperone A, Colangelo D, Gregoletto L, Reano S, Pietronave S, Merlin S, Talmon M, Novelli E, Diena M, Nicoletti C, Musarò A, Filigheddu N, Follenzi A, Prat M. Human Cardiac Progenitor Spheroids Exhibit Enhanced Engraftment Potential. PLoS One 2015; 10:e0137999. [PMID: 26375957 PMCID: PMC4572703 DOI: 10.1371/journal.pone.0137999] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 08/24/2015] [Indexed: 01/08/2023] Open
Abstract
A major obstacle to an effective myocardium stem cell therapy has always been the delivery and survival of implanted stem cells in the heart. Better engraftment can be achieved if cells are administered as cell aggregates, which maintain their extra-cellular matrix (ECM). We have generated spheroid aggregates in less than 24 h by seeding human cardiac progenitor cells (hCPCs) onto methylcellulose hydrogel-coated microwells. Cells within spheroids maintained the expression of stemness/mesenchymal and ECM markers, growth factors and their cognate receptors, cardiac commitment factors, and metalloproteases, as detected by immunofluorescence, q-RT-PCR and immunoarray, and expressed a higher, but regulated, telomerase activity. Compared to cells in monolayers, 3D spheroids secreted also bFGF and showed MMP2 activity. When spheroids were seeded on culture plates, the cells quickly migrated, displaying an increased wound healing ability with or without pharmacological modulation, and reached confluence at a higher rate than cells from conventional monolayers. When spheroids were injected in the heart wall of healthy mice, some cells migrated from the spheroids, engrafted, and remained detectable for at least 1 week after transplantation, while, when the same amount of cells was injected as suspension, no cells were detectable three days after injection. Cells from spheroids displayed the same engraftment capability when they were injected in cardiotoxin-injured myocardium. Our study shows that spherical in vivo ready-to-implant scaffold-less aggregates of hCPCs able to engraft also in the hostile environment of an injured myocardium can be produced with an economic, easy and fast protocol.
Collapse
Affiliation(s)
- Francesca Oltolina
- Dept. Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Andrea Zamperone
- Dept. Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Donato Colangelo
- Dept. Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Luca Gregoletto
- Dept. Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Simone Reano
- Dept. Translational Medicine, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Stefano Pietronave
- Dept. Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Simone Merlin
- Dept. Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Maria Talmon
- Dept. Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Eugenio Novelli
- Dept. of Cardiac Surgery, Clinica S. Gaudenzio, Novara, Italy
| | - Marco Diena
- Dept. of Cardiac Surgery, Clinica S. Gaudenzio, Novara, Italy
| | - Carmine Nicoletti
- Institute Pasteur Cenci-Bolognetti, DAHFMO, Roma, Italy
- Unit of Histology and Medical Embryology, IIM, Sapienza University of Rome, Rome, Italy
| | - Antonio Musarò
- Institute Pasteur Cenci-Bolognetti, DAHFMO, Roma, Italy
- Unit of Histology and Medical Embryology, IIM, Sapienza University of Rome, Rome, Italy
| | - Nicoletta Filigheddu
- Dept. Translational Medicine, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Antonia Follenzi
- Dept. Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
- Centro di Biotecnologie per la Ricerca Medica Applicata (BRMA), Novara, Italy
| | - Maria Prat
- Dept. Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
- Centro di Biotecnologie per la Ricerca Medica Applicata (BRMA), Novara, Italy
- * E-mail:
| |
Collapse
|
92
|
Jackson R, Tilokee EL, Latham N, Mount S, Rafatian G, Strydhorst J, Ye B, Boodhwani M, Chan V, Ruel M, Ruddy TD, Suuronen EJ, Stewart DJ, Davis DR. Paracrine Engineering of Human Cardiac Stem Cells With Insulin-Like Growth Factor 1 Enhances Myocardial Repair. J Am Heart Assoc 2015; 4:e002104. [PMID: 26363004 PMCID: PMC4599498 DOI: 10.1161/jaha.115.002104] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Insulin-like growth factor 1 (IGF-1) activates prosurvival pathways and improves postischemic cardiac function, but this key cytokine is not robustly expressed by cultured human cardiac stem cells. We explored the influence of an enhanced IGF-1 paracrine signature on explant-derived cardiac stem cell-mediated cardiac repair. METHODS AND RESULTS Receptor profiling demonstrated that IGF-1 receptor expression was increased in the infarct border zones of experimentally infarcted mice by 1 week after myocardial infarction. Human explant-derived cells underwent somatic gene transfer to overexpress human IGF-1 or the green fluorescent protein reporter alone. After culture in hypoxic reduced-serum media, overexpression of IGF-1 enhanced proliferation and expression of prosurvival transcripts and prosurvival proteins and decreased expression of apoptotic markers in both explant-derived cells and cocultured neonatal rat ventricular cardiomyocytes. Transplant of explant-derived cells genetically engineered to overexpress IGF-1 into immunodeficient mice 1 week after infarction boosted IGF-1 content within infarcted tissue and long-term engraftment of transplanted cells while reducing apoptosis and long-term myocardial scarring. CONCLUSIONS Paracrine engineering of explant-derived cells to overexpress IGF-1 provided a targeted means of improving cardiac stem cell-mediated repair by enhancing the long-term survival of transplanted cells and surrounding myocardium.
Collapse
Affiliation(s)
| | | | | | - Seth Mount
- University of Ottawa Heart InstituteOttawa, Canada
| | | | | | - Bin Ye
- University of Ottawa Heart InstituteOttawa, Canada
| | | | - Vincent Chan
- University of Ottawa Heart InstituteOttawa, Canada
| | - Marc Ruel
- University of Ottawa Heart InstituteOttawa, Canada
| | | | | | | | - Darryl R Davis
- University of Ottawa Heart InstituteOttawa, Canada
- Correspondence to: Darryl R. Davis, MD, University of Ottawa Heart Institute, H3214 40 Ruskin Ave, Ottawa, Ontario, Canada K1Y4W7. E-mail:
| |
Collapse
|
93
|
Henry E, Cores J, Hensley MT, Anthony S, Vandergriff A, de Andrade JBM, Allen T, Caranasos TG, Lobo LJ, Cheng K. Adult Lung Spheroid Cells Contain Progenitor Cells and Mediate Regeneration in Rodents With Bleomycin-Induced Pulmonary Fibrosis. Stem Cells Transl Med 2015; 4:1265-74. [PMID: 26359426 DOI: 10.5966/sctm.2015-0062] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/29/2015] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Lung diseases are devastating conditions and ranked as one of the top five causes of mortality worldwide according to the World Health Organization. Stem cell therapy is a promising strategy for lung regeneration. Previous animal and clinical studies have focused on the use of mesenchymal stem cells (from other parts of the body) for lung regenerative therapies. We report a rapid and robust method to generate therapeutic resident lung progenitors from adult lung tissues. Outgrowth cells from healthy lung tissue explants are self-aggregated into three-dimensional lung spheroids in a suspension culture. Without antigenic sorting, the lung spheroids recapitulate the stem cell niche and contain a natural mixture of lung stem cells and supporting cells. In vitro, lung spheroid cells can be expanded to a large quantity and can form alveoli-like structures and acquire mature lung epithelial phenotypes. In severe combined immunodeficiency mice with bleomycin-induced pulmonary fibrosis, intravenous injection of human lung spheroid cells inhibited apoptosis, fibrosis, and infiltration but promoted angiogenesis. In a syngeneic rat model of pulmonary fibrosis, lung spheroid cells outperformed adipose-derived mesenchymal stem cells in reducing fibrotic thickening and infiltration. Previously, lung spheroid cells (the spheroid model) had only been used to study lung cancer cells. Our data suggest that lung spheroids and lung spheroid cells from healthy lung tissues are excellent sources of regenerative lung cells for therapeutic lung regeneration. SIGNIFICANCE The results from the present study will lead to future human clinical trials using lung stem cell therapies to treat various incurable lung diseases, including pulmonary fibrosis. The data presented here also provide fundamental knowledge regarding how injected stem cells mediate lung repair in pulmonary fibrosis.
Collapse
Affiliation(s)
- Eric Henry
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, USA
| | - Jhon Cores
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, USA
| | - M Taylor Hensley
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Shirena Anthony
- Department of Biology, North Carolina State University, Raleigh, North Carolina, USA
| | - Adam Vandergriff
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, USA
| | - James B M de Andrade
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Tyler Allen
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Thomas G Caranasos
- Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Leonard J Lobo
- Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
94
|
Keith MCL, Bolli R. "String theory" of c-kit(pos) cardiac cells: a new paradigm regarding the nature of these cells that may reconcile apparently discrepant results. Circ Res 2015; 116:1216-30. [PMID: 25814683 DOI: 10.1161/circresaha.116.305557] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Although numerous preclinical investigations have consistently demonstrated salubrious effects of c-kit(pos) cardiac cells administered after myocardial infarction, the mechanism of action remains highly controversial. We and others have found little or no evidence that these cells differentiate into mature functional cardiomyocytes, suggesting paracrine effects. In this review, we propose a new paradigm predicated on a comprehensive analysis of the literature, including studies of cardiac development; we have (facetiously) dubbed this conceptual construct "string theory" of c-kit(pos) cardiac cells because it reconciles multifarious and sometimes apparently discrepant results. There is strong evidence that, during development, the c-kit receptor is expressed in different pools of cardiac progenitors (some capable of robust cardiomyogenesis and others with little or no contribution to myocytes). Accordingly, c-kit positivity, in itself, does not define the embryonic origins, lineage capabilities, or differentiation capacities of specific cardiac progenitors. C-kit(pos) cells derived from the first heart field exhibit cardiomyogenic potential during development, but these cells are likely depleted shortly before or after birth. The residual c-kit(pos) cells found in the adult heart are probably of proepicardial origin, possess a mesenchymal phenotype (resembling bone marrow mesenchymal stem/stromal cells), and are capable of contributing significantly only to nonmyocytic lineages (fibroblasts, smooth muscle cells, and endothelial cells). If these 2 populations (first heart field and proepicardium) express different levels of c-kit, the cardiomyogenic potential of first heart field progenitors might be reconciled with recent results of c-kit(pos) cell lineage tracing studies. The concept that c-kit expression in the adult heart identifies epicardium-derived, noncardiomyogenic precursors with a mesenchymal phenotype helps to explain the beneficial effects of c-kit(pos) cell administration to ischemically damaged hearts despite the observed paucity of cardiomyogenic differentiation of these cells.
Collapse
Affiliation(s)
- Matthew C L Keith
- From the Division of Cardiovascular Medicine, Department of Cardiology, University of Louisville, KY
| | - Roberto Bolli
- From the Division of Cardiovascular Medicine, Department of Cardiology, University of Louisville, KY.
| |
Collapse
|
95
|
Leite CF, Almeida TR, Lopes CS, Dias da Silva VJ. Multipotent stem cells of the heart-do they have therapeutic promise? Front Physiol 2015; 6:123. [PMID: 26005421 PMCID: PMC4424849 DOI: 10.3389/fphys.2015.00123] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 04/06/2015] [Indexed: 01/26/2023] Open
Abstract
The last decade has brought a comprehensive change in our view of cardiac remodeling processes under both physiological and pathological conditions, and cardiac stem cells have become important new players in the general mainframe of cardiac homeostasis. Different types of cardiac stem cells show different capacities for differentiation into the three major cardiac lineages: myocytes, endothelial cells and smooth muscle cells. Physiologically, cardiac stem cells contribute to cardiac homeostasis through continual cellular turnover. Pathologically, these cells exhibit a high level of proliferative activity in an apparent attempt to repair acute cardiac injury, indicating that these cells possess (albeit limited) regenerative potential. In addition to cardiac stem cells, mesenchymal stem cells represent another multipotent cell population in the heart; these cells are located in regions near pericytes and exhibit regenerative, angiogenic, antiapoptotic, and immunosuppressive properties. The discovery of these resident cardiac stem cells was followed by a number of experimental studies in animal models of cardiomyopathies, in which cardiac stem cells were tested as a therapeutic option to overcome the limited transdifferentiating potential of hematopoietic or mesenchymal stem cells derived from bone marrow. The promising results of these studies prompted clinical studies of the role of these cells, which have demonstrated the safety and practicability of cellular therapies for the treatment of heart disease. However, questions remain regarding this new therapeutic approach. Thus, the aim of the present review was to discuss the multitude of different cardiac stem cells that have been identified, their possible functional roles in the cardiac regenerative process, and their potential therapeutic uses in treating cardiac diseases.
Collapse
Affiliation(s)
- Camila F Leite
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| | - Thalles R Almeida
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| | - Carolina S Lopes
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| | - Valdo J Dias da Silva
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| |
Collapse
|
96
|
Nigro P, Perrucci GL, Gowran A, Zanobini M, Capogrossi MC, Pompilio G. c-kit(+) cells: the tell-tale heart of cardiac regeneration? Cell Mol Life Sci 2015; 72:1725-40. [PMID: 25575564 PMCID: PMC11113938 DOI: 10.1007/s00018-014-1832-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/18/2014] [Accepted: 12/30/2014] [Indexed: 12/21/2022]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in the developed world. Although ongoing therapeutic strategies ameliorate symptoms and prolong life for patients with cardiovascular diseases, they do not solve the critical issue related to the loss of cardiac tissue. Accordingly, stem/progenitor cell therapy has emerged as a paramount approach for cardiac repair and regeneration. In this regard, c-kit(+) cells have animated much interest and controversy. These cells are self-renewing, clonogenic, and multipotent and display a noteworthy potential to differentiate into all cardiovascular lineages. However, their functional contribution to cardiomyocyte turnover is one of the centrally debated issues concerning their regenerative potential. Regardless, plentiful preclinical and clinical studies have been conducted which provide evidence for the capacity of c-kit(+) cells to improve cardiac function. The purpose of this review is to give a comprehensive, impartial, critical description and evaluation of the literature on c-kit(+) cells from bench to bedside in order to address their true potential, benefits and controversies.
Collapse
Affiliation(s)
- Patrizia Nigro
- Laboratory of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via Parea 4, 20138, Milan, Italy,
| | | | | | | | | | | |
Collapse
|
97
|
Hensley MT, de Andrade J, Keene B, Meurs K, Tang J, Wang Z, Caranasos TG, Piedrahita J, Li TS, Cheng K. Cardiac regenerative potential of cardiosphere-derived cells from adult dog hearts. J Cell Mol Med 2015; 19:1805-13. [PMID: 25854418 PMCID: PMC4549031 DOI: 10.1111/jcmm.12585] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/27/2015] [Indexed: 11/28/2022] Open
Abstract
The regenerative potential of cardiosphere-derived cells (CDCs) for ischaemic heart disease has been demonstrated in mice, rats, pigs and a recently completed clinical trial. The regenerative potential of CDCs from dog hearts has yet to be tested. Here, we show that canine CDCs can be produced from adult dog hearts. These cells display similar phenotypes in comparison to previously studied CDCs derived from rodents and human beings. Canine CDCs can differentiate into cardiomyocytes, smooth muscle cells and endothelial cells in vitro. In addition, conditioned media from canine CDCs promote angiogenesis but inhibit cardiomyocyte death. In a doxorubicin-induced mouse model of dilated cardiomyopathy (DCM), intravenous infusion of canine CDCs improves cardiac function and decreases cardiac fibrosis. Histology revealed that injected canine CDCs engraft in the mouse heart and increase capillary density. Out study demonstrates the regenerative potential of canine CDCs in a mouse model of DCM.
Collapse
Affiliation(s)
- Michael Taylor Hensley
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - James de Andrade
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Bruce Keene
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Kathryn Meurs
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.,Center for Comparative Medicine and Translational Researches, North Carolina State University, Raleigh, NC, USA
| | - Junnan Tang
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.,Department of Cardiology, First Affiliated Hospital, College of Medicine, Zhengzhou University, Zhengzhou, China
| | - Zegen Wang
- The Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Thomas G Caranasos
- Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jorge Piedrahita
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.,Center for Comparative Medicine and Translational Researches, North Carolina State University, Raleigh, NC, USA
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.,Center for Comparative Medicine and Translational Researches, North Carolina State University, Raleigh, NC, USA.,The Cyrus Tang Hematology Center, Soochow University, Suzhou, China.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
| |
Collapse
|
98
|
Human cardiosphere-derived cells from advanced heart failure patients exhibit augmented functional potency in myocardial repair. JACC-HEART FAILURE 2015; 2:49-61. [PMID: 24511463 DOI: 10.1016/j.jchf.2013.08.008] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES This study sought to compare the regenerative potency of cells derived from healthy and diseased human hearts. BACKGROUND Results from pre-clinical studies and the CADUCEUS (CArdiosphere-Derived aUtologous stem CElls to reverse ventricUlar dySfunction) trial support the notion that cardiosphere-derived cells (CDCs) from normal and recently infarcted hearts are capable of regenerating healthy heart tissue after myocardial infarction (MI). It is unknown whether CDCs derived from advanced heart failure (HF) patients retain the same regenerative potency. METHODS In a mouse model of acute MI, we compared the regenerative potential and functional benefits of CDCs derived from 3 groups: 1) non-failing (NF) donor: healthy donor hearts post-transplantation; 2) MI: patients who had an MI 9 to 35 days before biopsy; and 3) HF: advanced cardiomyopathy tissue explanted at cardiac transplantation. RESULTS Cell growth and phenotype were identical in all 3 groups. Injection of HF CDCs led to the greatest therapeutic benefit in mice, with the highest left ventricular ejection fraction, thickest infarct wall, most viable tissue, and least scar 3 weeks after treatment. In vitro assays revealed that HF CDCs secreted higher levels of stromal cell-derived factor (SDF)-1, which may contribute to the cells' augmented resistance to oxidative stress, enhanced angiogenesis, and improved myocyte survival. Histological analysis indicated that HF CDCs engrafted better, recruited more endogenous stem cells, and induced greater angiogenesis and cardiomyocyte cell-cycle re-entry. CDC-secreted SDF-1 levels correlated with decreases in scar mass over time in CADUCEUS patients treated with autologous CDCs. CONCLUSIONS CDCs from advanced HF patients exhibit augmented potency in ameliorating ventricular dysfunction post-MI, possibly through SDF-1–mediated mechanisms.
Collapse
|
99
|
Sensitivity and dose dependency of radiation-induced injury in hematopoietic stem/progenitor cells in mice. Sci Rep 2015; 5:8055. [PMID: 25623887 PMCID: PMC4306913 DOI: 10.1038/srep08055] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 01/02/2015] [Indexed: 01/07/2023] Open
Abstract
We evaluated the sensitivity and dose dependency of radiation-induced injury in hematopoietic stem/progenitor cells. Adult C57BL/6 mice were daily exposed to 0, 2, 10, 50, and 250 mGy γ-ray for 1 month in succession, respectively. The damage of hematopoietic stem/progenitor cells in bone marrow were investigated within 2 hours (acute phase) or at 3 months (chronic phase) after the last exposure. Daily exposure to over 10 mGy γ-ray significantly decreased the number and colony-forming capacity of hematopoietic stem/progenitor cells at acute phase, and did not completely recover at chronic phase with 250 mGy exposure. Interestingly, the daily exposure to 10 or 50 mGy γ-ray decreased the formation of mixed types of colonies at chronic phase, but the total number of colonies was comparable to control. Immunostaining analysis showed that the formation of 53BP1 foci in c-kit+ stem/progenitor cells was significantly increased with daily exposure to 50 and 250 mGy at acute phase, and 250 mGy at chronic phase. Many genes involved in toxicity responses were up- or down-regulated with the exposures to all doses. Our data have clearly shown the sensitivity and dose dependency of radiation-induced injury in hematopoietic stem/progenitor cells of mice with daily exposures to 2 ~ 250 mGy γ-ray.
Collapse
|
100
|
Ge Z, Lal S, Le TYL, Dos Remedios C, Chong JJH. Cardiac stem cells: translation to human studies. Biophys Rev 2014; 7:127-139. [PMID: 28509972 DOI: 10.1007/s12551-014-0148-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/13/2014] [Indexed: 02/08/2023] Open
Abstract
The discovery of multiple classes of cardiac progenitor cells in the adult mammalian heart has generated hope for their use as a therapeutic in heart failure. However, successful results from animal models have not always yielded similar findings in human studies. Recent Phase I/II trials of c-Kit (SCIPIO) and cardiosphere-based (CADUCEUS) cardiac progenitor cells have demonstrated safety and some therapeutic efficacy. Gaps remain in our understanding of the origins, function and relationships between the different progenitor cell families, many of which are heterogeneous populations with overlapping definitions. Another challenge lies in the limitations of small animal models in replicating the human heart. Cryopreserved human cardiac tissue provides a readily available source of cardiac progenitor cells and may help address these questions. We review important findings and relative unknowns of the main classes of cardiac progenitor cells, highlighting differences between animal and human studies.
Collapse
Affiliation(s)
- Zijun Ge
- Bosch Institute, The University of Sydney, Sydney, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Sean Lal
- Bosch Institute, The University of Sydney, Sydney, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Thi Y L Le
- Department of Cardiology Westmead Hospital, Sydney, NSW, Australia.,Centre for Heart Research, Westmead Millennium Institute for Medical Research, 176 Hawkesbury Road, Westmead, Sydney, NSW, Australia, 2145
| | | | - James J H Chong
- Department of Cardiology Westmead Hospital, Sydney, NSW, Australia. .,Sydney Medical School, University of Sydney, Sydney, NSW, Australia. .,Centre for Heart Research, Westmead Millennium Institute for Medical Research, 176 Hawkesbury Road, Westmead, Sydney, NSW, Australia, 2145.
| |
Collapse
|