1
|
Yahyazadeh R, Baradaran Rahimi V, Askari VR. Stem cell and exosome therapies for regenerating damaged myocardium in heart failure. Life Sci 2024; 351:122858. [PMID: 38909681 DOI: 10.1016/j.lfs.2024.122858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Finding novel treatments for cardiovascular diseases (CVDs) is a hot topic in medicine; cell-based therapies have reported promising news for controlling dangerous complications of heart disease such as myocardial infarction (MI) and heart failure (HF). Various progenitor/stem cells were tested in various in-vivo, in-vitro, and clinical studies for regeneration or repairing the injured tissue in the myocardial to accelerate the healing. Fetal, adult, embryonic, and induced pluripotent stem cells (iPSC) have revealed the proper potency for cardiac tissue repair. As an essential communicator among cells, exosomes with specific contacts (proteins, lncRNAs, and miRNAs) greatly promote cardiac rehabilitation. Interestingly, stem cell-derived exosomes have more efficiency than stem cell transplantation. Therefore, stem cells induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), cardiac stem cells (CDC), and skeletal myoblasts) and their-derived exosomes will probably be considered an alternative therapy for CVDs remedy. In addition, stem cell-derived exosomes have been used in the diagnosis/prognosis of heart diseases. In this review, we explained the advances of stem cells/exosome-based treatment, their beneficial effects, and underlying mechanisms, which will present new insights in the clinical field in the future.
Collapse
Affiliation(s)
- Roghayeh Yahyazadeh
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Margiana R, Pilehvar Y, Amalia FL, Lestari SW, Supardi S, I'tishom R. Mesenchymal stem cell secretome: A promising therapeutic strategy for erectile dysfunction? Asian J Urol 2024; 11:391-405. [PMID: 39139521 PMCID: PMC11318444 DOI: 10.1016/j.ajur.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 12/06/2023] [Indexed: 08/15/2024] Open
Abstract
Objective The secretome, comprising bioactive chemicals released by mesenchymal stem cells (MSCs), holds therapeutic promise in regenerative medicine. This review aimed to explore the therapeutic potential of the MSC secretome in regenerative urology, particularly for treating erectile dysfunction (ED), and to provide an overview of preclinical and clinical research on MSCs in ED treatment and subsequently to highlight the rationales, mechanisms, preclinical investigations, and therapeutic potential of the MSC secretome in this context. Methods The review incorporated an analysis of preclinical and clinical research involving MSCs in the treatment of ED. Subsequently, it delved into the existing knowledge regarding the MSC secretome, exploring its therapeutic potential. The methods included a comprehensive examination of relevant literature to discern the processes underlying the therapeutic efficacy of the MSC secretome. Results Preclinical research indicated the effectiveness of the MSC secretome in treating various models of ED. However, the precise mechanisms of its therapeutic efficacy remain unknown. The review provided insights into the anti-inflammatory, pro-angiogenic, and trophic properties of the MSC secretome. It also discussed potential advantages, such as avoiding issues related to cellular therapy, including immunogenicity, neoplastic transformation, and cost. Conclusion This review underscores the significant therapeutic potential of the MSC secretome in regenerative urology, particularly for ED treatment. While preclinical studies demonstrate promising outcomes, further research is essential to elucidate the specific mechanisms underlying the therapeutic efficacy before clinical application. The review concludes by discussing future perspectives and highlighting the challenges associated with the clinical translation of the MSC secretome in regenerative urology.
Collapse
Affiliation(s)
- Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Indonesia General Academic Hospital, Depok, Indonesia
- Ciptomangunkusumo General Academic Hospital, Jakarta, Indonesia
| | - Younes Pilehvar
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Science, Urmia, Iran
| | - Fatkhurrohmah L. Amalia
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Dr. Kariadi Hospital, Semarang, Indonesia
| | - Silvia W. Lestari
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Indonesia General Academic Hospital, Depok, Indonesia
- Ciptomangunkusumo General Academic Hospital, Jakarta, Indonesia
- Department of Medical Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Supardi Supardi
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Reny I'tishom
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Biomedical Science, Faculty of Medicine, Universitas Airlangga Surabaya, Indonesia
| |
Collapse
|
3
|
Amini H, Namjoo AR, Narmi MT, Mardi N, Narimani S, Naturi O, Khosrowshahi ND, Rahbarghazi R, Saghebasl S, Hashemzadeh S, Nouri M. Exosome-bearing hydrogels and cardiac tissue regeneration. Biomater Res 2023; 27:99. [PMID: 37803483 PMCID: PMC10559618 DOI: 10.1186/s40824-023-00433-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/18/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND In recent years, cardiovascular disease in particular myocardial infarction (MI) has become the predominant cause of human disability and mortality in the clinical setting. The restricted capacity of adult cardiomyocytes to proliferate and restore the function of infarcted sites is a challenging issue after the occurrence of MI. The application of stem cells and byproducts such as exosomes (Exos) has paved the way for the alleviation of cardiac tissue injury along with conventional medications in clinics. However, the short lifespan and activation of alloreactive immune cells in response to Exos and stem cells are the main issues in patients with MI. Therefore, there is an urgent demand to develop therapeutic approaches with minimum invasion for the restoration of cardiac function. MAIN BODY Here, we focused on recent data associated with the application of Exo-loaded hydrogels in ischemic cardiac tissue. Whether and how the advances in tissue engineering modalities have increased the efficiency of whole-based and byproducts (Exos) therapies under ischemic conditions. The integration of nanotechnology and nanobiology for designing novel smart biomaterials with therapeutic outcomes was highlighted. CONCLUSION Hydrogels can provide suitable platforms for the transfer of Exos, small molecules, drugs, and other bioactive factors for direct injection into the damaged myocardium. Future studies should focus on the improvement of physicochemical properties of Exo-bearing hydrogel to translate for the standard treatment options.
Collapse
Affiliation(s)
- Hassan Amini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran
| | - Atieh Rezaei Namjoo
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Taghavi Narmi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Mardi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Narimani
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ozra Naturi
- Department of Organic and Biochemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Nafiseh Didar Khosrowshahi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Solmaz Saghebasl
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Shahriar Hashemzadeh
- Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, 51548/53431, Iran.
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Zhang MX, Song Y, Xu WL, Zhang LX, Li C, Li YL. Natural Herbal Medicine as a Treatment Strategy for Myocardial Infarction through the Regulation of Angiogenesis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:8831750. [PMID: 35600953 PMCID: PMC9119779 DOI: 10.1155/2022/8831750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/25/2022] [Indexed: 11/18/2022]
Abstract
Methods We conducted a literature search on the bioactive components of medicinal plants and their effects on angiogenesis after MI. We searched for articles in Web of Science, MEDLINE, PubMed, Scopus, Google Scholar, and China National Knowledge Infrastructure databases before April 2021. Results In this article, we summarized the mechanisms by which copper ions, microRNA, Akt1, inflammation, oxidative stress, mitochondria, and pericytes are involved in angiogenesis after myocardial infarction. In addition, we reviewed the angiogenic effects of natural herbal medicines such as Salvia miltiorrhiza Bunge Bunge, Carthamus tinctorius L., Pueraria lobata, Astragalus, Panax ginseng C.A. Mey., Panax notoginseng (Burkill) F.H. Chen, Cinnamomum cassia (L.) J. Presl, Rehmannia glutinosa (Gaertn.) DC., Leonurus japonicus Houtt, Scutellaria baicalensis Georgi., and Geum macrophyllum Willd. Conclusions Some herbs have the effect of promoting angiogenesis. In the future, natural proangiogenic drugs may become candidates for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Mu-xin Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yu Song
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wan-li Xu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ling-xiao Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yun-lun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| |
Collapse
|
5
|
Perdomo S, Brugnini A, Trias N, Menyou A, Silveira G, Ranero S, Lens D, Díaz L, Grille S. Mobilized and apheresis-collected endothelial progenitor cells with plerixafor. J Clin Apher 2022; 37:245-252. [PMID: 35114004 DOI: 10.1002/jca.21967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) are immature cells able to proliferate and contribute to endothelial repair, vascular homeostasis, neovascularization, and angiogenesis. It therefore seems likely that circulating EPCs have therapeutic potential in ischemic and vascular diseases. In this study we evaluated the efficiency of EPC mobilization and collection by large volume leukapheresis in subjects with hematological diseases, treated with plerixafor in association with G-CSF. METHODS Twenty-two patients with lymphoid malignancies underwent rHuG-CSF and plerixafor treatment followed by leukapheresis. Blood samples before and after treatment and apheresis liquid sample were taken and analyzed by flow cytometry in order to quantified EPC. RESULTS The percentage of CD34+ cells and EPCs among circulating total nuclear cells (TNCs) increased significantly by approximately 2-fold and 3-fold, respectively, after plerixafor treatment. Consequently, the absolute number of CD34+ cells and EPCs were increased 4-fold after plerixafor treatment. The median PB concentration of EPCs before and after treatment were 0.77/μL (0.31-2.15) and 3.41/μL (1.78-4.54), respectively, P < .0001. The total EPCs collected per patient were 3.3×107 (0.8×107 -6.8×107 ). CONCLUSION We have shown that plerixafor in combination with G-CSF allows the mobilization and collection of large amounts of EPCs along with CD34+ cells in lymphoid neoplasm patients. The possibility to collect and to store these cells could represent a promising therapeutic tool for the treatment of ischemic complications without the need of in vitro expansion.
Collapse
Affiliation(s)
- Susana Perdomo
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay
| | - Andreina Brugnini
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Natalia Trias
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Alba Menyou
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay
| | - Gonzalo Silveira
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Sabrina Ranero
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Daniela Lens
- Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Lilián Díaz
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay
| | - Sofía Grille
- Servicio Médico Integral, Centro de Trasplante de Médula Ósea, Montevideo, Uruguay.,Laboratorio de Citometría y Biología Molecular, Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.,Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
6
|
Lee CS, Kim J, Cho HJ, Kim HS. Cardiovascular Regeneration via Stem Cells and Direct Reprogramming: A Review. Korean Circ J 2022; 52:341-353. [PMID: 35502566 PMCID: PMC9064703 DOI: 10.4070/kcj.2022.0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
Despite recent advancements in treatment strategies, cardiovascular disease such as heart failure remains a significant source of global mortality. Stem cell technology and cellular reprogramming are rapidly growing fields that will continue to prove useful in cardiac regenerative therapeutics. This review provides information on the role of human pluripotent stem cells (hPSCs) in cardiac regeneration and discusses the practical applications of hPSC-derived cardiomyocytes (CMCs). Moreover, we discuss the practical applications of hPSC-derived CMCs while outlining the relevance of directly-reprogrammed CMCs in regenerative medicine. This review critically summarizes the most recent advances in the field will help to guide future research in this developing area. Cardiovascular disease (CVD) is the leading causes of morbidity and death globally. In particular, a heart failure remains a major problem that contributes to global mortality. Considerable advancements have been made in conventional pharmacological therapies and coronary intervention surgery for cardiac disorder treatment. However, more than 15% of patients continuously progress to end-stage heart failure and eventually require heart transplantation. Over the past year, numerous numbers of protocols to generate cardiomyocytes (CMCs) from human pluripotent stem cells (hPSCs) have been developed and applied in clinical settings. Number of studies have described the therapeutic effects of hPSCs in animal models and revealed the underlying repair mechanisms of cardiac regeneration. In addition, biomedical engineering technologies have improved the therapeutic potential of hPSC-derived CMCs in vivo. Recently substantial progress has been made in driving the direct differentiation of somatic cells into mature CMCs, wherein an intermediate cellular reprogramming stage can be bypassed. This review provides information on the role of hPSCs in cardiac regeneration and discusses the practical applications of hPSC-derived CMCs; furthermore, it outlines the relevance of directly reprogrammed CMCs in regenerative medicine.
Collapse
Affiliation(s)
- Choon-Soo Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
| | - Joonoh Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jai Cho
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hyo-Soo Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Strategic Center of Cell & Bio Therapy, Seoul National University Hospital, Seoul, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
7
|
Carbone RG, Monselise A, Bottino G, Negrini S, Puppo F. Stem cells therapy in acute myocardial infarction: a new era? Clin Exp Med 2021; 21:231-237. [PMID: 33484381 PMCID: PMC8053645 DOI: 10.1007/s10238-021-00682-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Stem cells transplantation after acute myocardial infarction (AMI) has been claimed to restore cardiac function. However, this therapy is still restricted to experimental studies and clinical trials. Early un-blinded studies suggested a benefit from stem cell therapy following AMI. More recent blinded randomized trials have produced mixed results and, notably, the last largest pan-European clinical trial showed the inconclusive results. Furthermore, mechanisms of potential benefit remain uncertain. This review analytically evaluates 34 blinded and un-blinded clinical trials comprising 3142 patients and is aimed to: (1) identify the pros and cons of stem cell therapy up to a 6-month follow-up after AMI comparing benefit or no effectiveness reported in clinical trials; (2) provide useful information for planning future clinical programs of cardiac stem cell therapy.
Collapse
Affiliation(s)
- R G Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | | | - G Bottino
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - S Negrini
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - F Puppo
- Department of Internal Medicine, University of Genoa, Genoa, Italy.
| |
Collapse
|
8
|
Nappi F, Nenna A, Larobina D, Martuscelli G, Singh SSA, Chello M, Ambrosio L. The Use of Bioactive Polymers for Intervention and Tissue Engineering: The New Frontier for Cardiovascular Therapy. Polymers (Basel) 2021; 13:446. [PMID: 33573282 PMCID: PMC7866823 DOI: 10.3390/polym13030446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 12/28/2022] Open
Abstract
Coronary heart disease remains one of the leading causes of death in most countries. Healthcare improvements have seen a shift in the presentation of disease with a reducing number of ST-segment elevation myocardial infarctions (STEMIs), largely due to earlier reperfusion strategies such as percutaneous coronary intervention (PCI). Stents have revolutionized the care of these patients, but the long-term effects of these devices have been brought to the fore. The conceptual and technologic evolution of these devices from bare-metal stents led to the creation and wide application of drug-eluting stents; further research introduced the idea of polymer-based resorbable stents. We look at the evolution of stents and the multiple advantages and disadvantages offered by each of the different polymers used to make stents in order to identify what the stent of the future may consist of whilst highlighting properties that are beneficial to the patient alongside the role of the surgeon, the cardiologist, engineers, chemists, and biophysicists in creating the ideal stent.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord de Saint-Denis, 93200 Paris, France
| | - Antonio Nenna
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.N.); (M.C.)
| | - Domenico Larobina
- Institute for Polymers, Composites and Biomaterials, National Research Council of Italy, 06128 Rome, Italy; (D.L.); (L.A.)
| | - Giorgia Martuscelli
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania Luigi Vanvitelli, 81100 Naples, Italy;
| | | | - Massimo Chello
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (A.N.); (M.C.)
| | - Luigi Ambrosio
- Institute for Polymers, Composites and Biomaterials, National Research Council of Italy, 06128 Rome, Italy; (D.L.); (L.A.)
| |
Collapse
|
9
|
Abstract
Stem cell therapy offers a breakthrough opportunity for the improvement of ischemic heart diseases. Numerous clinical trials and meta-analyses appear to confirm its positive but variable effects on heart function. Whereas these trials widely differed in design, cell type, source, and doses reinjected, cell injection route and timing, and type of cardiac disease, crucial key factors that may favour the success of cell therapy emerge from the review of their data. Various types of cell have been delivered. Injection of myoblasts does not improve heart function and is often responsible for severe ventricular arrythmia occurrence. Using bone marrow mononuclear cells is a misconception, as they are not stem cells but mainly a mix of various cells of hematopoietic lineages and stromal cells, only containing very low numbers of cells that have stem cell-like features; this likely explain the neutral results or at best the modest improvement in heart function reported after their injection. The true existence of cardiac stem cells now appears to be highly discredited, at least in adults. Mesenchymal stem cells do not repair the damaged myocardial tissue but attenuate post-infarction remodelling and contribute to revascularization of the hibernated zone surrounding the scar. CD34+ stem cells - likely issued from pluripotent very small embryonic-like (VSEL) stem cells - emerge as the most convincing cell type, inducing structural and functional repair of the ischemic myocardial area, providing they can be delivered in large amounts via intra-myocardial rather than intra-coronary injection, and preferentially after myocardial infarct rather than chronic heart failure.
Collapse
Affiliation(s)
- Philippe Hénon
- CellProthera SAS and Institut de Recherche en Hématologie et Transplantation, CellProthera SAS 12 rue du Parc, 68100, Mulhouse, France.
| |
Collapse
|
10
|
Peterson KM, Franchi F, Olthoff M, Chen IY, Paulmurugan R, Rodriguez-Porcel M. Pathway-specific reporter genes to study stem cell biology. Stem Cells 2020; 38:808-814. [PMID: 32129537 PMCID: PMC11443457 DOI: 10.1002/stem.3167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/03/2020] [Accepted: 02/04/2020] [Indexed: 01/03/2023]
Abstract
Little is known on the phenotypic characteristics of stem cells (SCs) after they are transplanted to the myocardium, in part due to lack of noninvasive platforms to study SCs directly in the living subject. Reporter gene imaging has played a valuable role in the noninvasive assessment of cell fate in vivo. In this study, we validated a pathway-specific reporter gene that can be used to noninvasively image the phenotype of SCs transplanted to the myocardium. Rat mesenchymal SCs (MSCs) were studied for phenotypic evidence of myogenic characteristics under in vitro conditions. After markers of myogenic characteristics were identified, we constructed a reporter gene sensor, comprising the firefly luciferase (Fluc) reporter gene driven by the troponin T (TnT) promoter (cardio MSCs had threefold expression in polymerase chain reaction compared to control MSCs) using a two-step signal amplification strategy. MSCs transfected with TnT-Fluc were studied and validated under in vitro conditions, showing a strong signal after MSCs acquired myogenic characteristics. Lastly, we observed that cardio MSCs had higher expression of the reporter sensor compared to control cells (0.005 ± 0.0005 vs 0.0025 ± 0.0008 Tnt-Fluc/ubiquitin-Fluc, P < .05), and that this novel sensor can detect the change in the phenotype of MSCs directly in the living subject. Pathway-specific reporter gene imaging allows assessment of changes in the phenotype of MSCs after delivery to the ischemic myocardium, providing important information on the phenotype of these cells. Imaging sensors like the one described here are critical to better understanding of the changes that SCs undergo after transplantation.
Collapse
Affiliation(s)
- Karen M Peterson
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Federico Franchi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Michaela Olthoff
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ian Y Chen
- Cardiology Section, Medical Services, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Cardiovascular Institute, Stanford University, Stanford, California, USA
| | - Ramasamy Paulmurugan
- Cardiovascular Institute, Stanford University, Stanford, California, USA
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, California, USA
| | | |
Collapse
|
11
|
Audam TN, Nong Y, Tomlin A, Jurkovic A, Li H, Zhu X, Long BW, Zheng YW, Weirick T, Brittian KR, Riggs DW, Gumpert A, Uchida S, Guo Y, Wysoczynski M, Jones SP. Cardiac mesenchymal cells from failing and nonfailing hearts limit ventricular dilation when administered late after infarction. Am J Physiol Heart Circ Physiol 2020; 319:H109-H122. [PMID: 32442025 DOI: 10.1152/ajpheart.00114.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although cell therapy-mediated cardiac repair offers promise for treatment/management of heart failure, lack of fundamental understanding of how cell therapy works limits its translational potential. In particular, whether reparative cells from failing hearts differ from cells derived from nonfailing hearts remains unexplored. Here, we assessed differences between cardiac mesenchymal cells (CMC) derived from failing (HF) versus nonfailing (Sham) hearts and whether the source of donor cells (i.e., from HF vs. Sham) limits reparative capacity, particularly when administered late after infarction. To determine the impact of the donor source of CMCs, we characterized the transcriptional profile of CMCs isolated from sham (Sham-CMC) and failing (HF-CMC) hearts. RNA-seq analysis revealed unique transcriptional signatures in Sham-CMC and HF-CMC, suggesting that the donor source impacts CMC. To determine whether the donor source affects reparative potential, C57BL6/J female mice were subjected to 60 min of regional myocardial ischemia and then reperfused for 35 days. In a randomized, controlled, and blinded fashion, vehicle, HF-CMC, or Sham-CMC were injected into the lumen of the left ventricle at 35 days post-MI. An additional 5 weeks later, cardiac function was assessed by echocardiography, which indicated that delayed administration of Sham-CMC and HF-CMC attenuated ventricular dilation. We also determined whether Sham-CMC and HF-CMC treatments affected ventricular histopathology. Our data indicate that the donor source (nonfailing vs. failing hearts) affects certain aspects of CMC, and these insights may have implications for future studies. Our data indicate that delayed administration of CMC limits ventricular dilation and that the source of CMC may influence their reparative actions.NEW & NOTEWORTHY Most preclinical studies have used only cells from healthy, nonfailing hearts. Whether donor condition (i.e., heart failure) impacts cells used for cell therapy is not known. We directly tested whether donor condition impacted the reparative effects of cardiac mesenchymal cells in a chronic model of myocardial infarction. Although cells from failing hearts differed in multiple aspects, they retained the potential to limit ventricular remodeling.
Collapse
Affiliation(s)
- Timothy N Audam
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Yibing Nong
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Alex Tomlin
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Andrea Jurkovic
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Hong Li
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Xiaoping Zhu
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Bethany W Long
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Yi Wei Zheng
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Tyler Weirick
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky.,Cardiovascular Innovation Institute, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Kenneth R Brittian
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Daniel W Riggs
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Anna Gumpert
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Shizuka Uchida
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky.,Cardiovascular Innovation Institute, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Yiru Guo
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Marcin Wysoczynski
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Steven P Jones
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
12
|
Li J, Zhou Z, Wen J, Jiang F, Xia Y. Human Amniotic Mesenchymal Stem Cells Promote Endogenous Bone Regeneration. Front Endocrinol (Lausanne) 2020; 11:543623. [PMID: 33133012 PMCID: PMC7562979 DOI: 10.3389/fendo.2020.543623] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Bone regeneration has become a research hotspot and therapeutic target in the field of bone and joint medicine. Stem cell-based therapy aims to promote endogenous regeneration and improves therapeutic effects and side-effects of traditional reconstruction of significant bone defects and disorders. Human amniotic mesenchymal stem cells (hAMSCs) are seed cells with superior paracrine functions on immune-regulation, anti-inflammation, and vascularized tissue regeneration. The present review summarized the source and characteristics of hAMSCs and analyzed their roles in tissue regeneration. Next, the therapeutic effects and mechanisms of hAMSCs in promoting bone regeneration of joint diseases and bone defects. Finally, the clinical application of hAMSCs from current clinical trials was analyzed. Although more studies are needed to confirm that hAMSC-based therapy to treat bone diseases, the clinical application prospect of the approach is worth investigating.
Collapse
Affiliation(s)
- Jin Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Zhixuan Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of General Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jin Wen
- Department of Prosthodontics, School of Medicine, College of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fei Jiang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of General Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- *Correspondence: Fei Jiang
| | - Yang Xia
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Yang Xia
| |
Collapse
|
13
|
Yang HM, Kim JY, Cho HJ, Lee JE, Jin S, Hur J, Kwon YW, Seong MW, Choi EK, Lee HY, Lee HS, Jeon M, Kim J, Yang J, Oh S, Suh KS, Yoon SS, Kim KB, Oh BH, Park YB, Kim HS. NFATc1+CD31+CD45- circulating multipotent stem cells derived from human endocardium and their therapeutic potential. Biomaterials 2019; 232:119674. [PMID: 31865194 DOI: 10.1016/j.biomaterials.2019.119674] [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: 10/07/2019] [Accepted: 12/05/2019] [Indexed: 10/25/2022]
Abstract
Many studies have shown the existence of cardiac stem cells in the myocardium and epicardial progenitor cells in the epicardium. However, the characteristics of stem cells in the endocardium has not been fully elucidated. In this study, we investigated the origin of newly identified cells in the blood and their therapeutic potential. The new population of cells, identified from human peripheral blood, was quite different from previously reported stem cells. These newly identified cells, which we named Circulating Multipotent Stem (CiMS) cells, were multipotent, and therefore differentiated into multiple lineages in vitro and in vivo. In order to determine the origin of these cells, we collected peripheral blood from a group of patients who underwent bone marrow, liver, heart, or kidney transplantation. We identified the endocardium as the origin of these cells because the Short Tandem Repeat profile of CiMS cells from the recipient had changed from the recipient's profile to the donor's profile after heart transplantation. CiMS cells significantly increased after stimuli to the endocardium, such as catheter ablation for arrhythmia or acute myocardial infarction. CiMS cells circulate in human peripheral blood and are easily obtainable, suggesting that these cells could be a promising tool for cell therapy.
Collapse
Affiliation(s)
- Han-Mo Yang
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Ju-Young Kim
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Hyun-Jai Cho
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Joo-Eun Lee
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Sooryeonhwa Jin
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Jin Hur
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Yoo-Wook Kwon
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Moon-Woo Seong
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Eue-Keun Choi
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Hae-Young Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Hyun-Seob Lee
- Genomics Core Facility, Department of Transdisciplinary Research and Collaboration, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Mika Jeon
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Joonoh Kim
- National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Jaeseok Yang
- Transplantation Center, Seoul National University Hospital, Seoul, South Korea
| | - Seil Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Kyung-Suk Suh
- Department of Surgery, Seoul National University Hospital, Seoul, South Korea
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Ki-Bong Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, South Korea
| | - Byung-Hee Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Young-Bae Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea
| | - Hyo-Soo Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea; National Research Laboratory for Stem Cell Niche, Seoul National University Hospital, Seoul, South Korea; Innovative Research Institute for Cell Therapy, Seoul National University Hospital, Seoul, South Korea; Molecular Medicine and Biopharmaceutical Sciences, Seoul National University, Seoul, South Korea.
| |
Collapse
|
14
|
Sabbah N, Tamari T, Elimelech R, Doppelt O, Rudich U, Zigdon-Giladi H. Predicting Angiogenesis by Endothelial Progenitor Cells Relying on In-Vitro Function Assays and VEGFR-2 Expression Levels. Biomolecules 2019; 9:biom9110717. [PMID: 31717420 PMCID: PMC6921061 DOI: 10.3390/biom9110717] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Clinical trials have demonstrated the safety and efficacy of autologous endothelial progenitor cell (EPC) therapy in various diseases. Since EPCs' functions are influenced by genetic, systemic and environmental factors, the therapeutic potential of each individual EPCs is unknown and may affect treatment outcome. Therefore, our aim was to compare EPCs function among healthy donors in order to predict blood vessel formation (angiogenesis) before autologous EPC transplantation. Human EPCs were isolated from the blood of ten volunteers. EPCs proliferation rate, chemoattractant ability, and CXCR4 mRNA levels were different among donors (p < 0.0001, p < 0.01, p < 0.001, respectively). A positive correlation was found between SDF-1, CXCR4, and EPCs proliferation (R = 0.736, p < 0.05 and R = 0.8, p < 0.01, respectively). In-vivo, blood vessels were counted ten days after EPCs transplantation in a subcutaneous mouse model. Mean vessel density was different among donors (p = 0.0001); nevertheless, donors with the lowest vessel densities were higher compared to control (p < 0.05). Finally, using a linear regression model, a mathematical equation was generated to predict blood vessel density relying on: (i) EPCs chemoattractivity, and (ii) VEGFR-2 mRNA levels. Results reveal differences in EPCs functions among healthy individuals, emphasizing the need for a potency assay to pave the way for standardized research and clinical use of human EPCs.
Collapse
Affiliation(s)
- Nadin Sabbah
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa 3109600, Israel; (N.S.); (T.T.); (R.E.); (O.D.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel;
| | - Tal Tamari
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa 3109600, Israel; (N.S.); (T.T.); (R.E.); (O.D.)
| | - Rina Elimelech
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa 3109600, Israel; (N.S.); (T.T.); (R.E.); (O.D.)
- Department of Periodontology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Ofri Doppelt
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa 3109600, Israel; (N.S.); (T.T.); (R.E.); (O.D.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel;
| | - Utai Rudich
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel;
| | - Hadar Zigdon-Giladi
- Laboratory for Bone Repair, Rambam Health Care Campus, Haifa 3109600, Israel; (N.S.); (T.T.); (R.E.); (O.D.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel;
- Department of Periodontology, Rambam Health Care Campus, Haifa 3109601, Israel
- Correspondence: ; Tel.: +972-4-854-3606
| |
Collapse
|
15
|
Abstract
Chemokines are small secreted proteins with chemoattractant properties that play a key role in inflammation. One such chemokine, Stromal cell-derived factor-1 (SDF-1) also known as CXCL12, and its receptor, CXCR4, are expressed and functional in cardiac myocytes. SDF-1 both stimulates and enhances the cellular signal which attracts potentially beneficial stem cells for tissue repair within the ischemic heart. Paradoxically however, this chemokine is known to act in concert with the inflammatory cytokines of the innate immune response which contributes to cellular injury through the recruitment of inflammatory cells during ischemia. In the present study, we have demonstrated that SDF-1 has dose dependent effects on freshly isolated cardiomyocytes. Using Tunnel and caspase 3-activation assays, we have demonstrated that the treatment of isolated adult rat cardiac myocyte with SDF-1 at higher concentrations (pathological concentrations) induced apoptosis. Furthermore, ELISA data demonstrated that the treatment of isolated adult rat cardiac myocyte with SDF-1 at higher concentrations upregulated TNF-α protein expression which directly correlated with subsequent apoptosis. There was a significant reduction in SDF-1 mediated apoptosis when TNF-α expression was neutralized which suggests that SDF-1 mediated apoptosis is TNF-α-dependent. The fact that certain stimuli are capable of driving cardiomyocytes into apoptosis indicates that these cells are susceptible to clinically relevant apoptotic triggers. Our findings suggest that the elevated SDF-1 levels seen in a variety of clinical conditions, including ischemic myocardial infarction, may either directly or indirectly contribute to cardiac cell death via a TNF-α mediated pathway. This highlights the importance of this receptor/ligand in regulating the cardiomyocyte response to stress conditions.
Collapse
|
16
|
Cho JW, Seo MS, Kang KK, Sung SE. Effect of human thymus adipose tissue-derived mesenchymal stem cells on myocardial infarction in rat model. Regen Ther 2019; 11:192-198. [PMID: 31453274 PMCID: PMC6700206 DOI: 10.1016/j.reth.2019.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 06/13/2019] [Accepted: 07/17/2019] [Indexed: 10/26/2022] Open
Abstract
Background and objective Stem cell (SC) therapy exhibits promising therapeutic efficiency against cardiovascular disease. The thymus adipose tissue (TAT) is familiar to cardiac surgeons with sternotomy; however, the application of TAT in SC therapy remains unknown. We assessed the effectiveness of TAT-derived mesenchymal SCs (TAT-MSCs) in the rat myocardial infarction (MI) model. Methods The human TATs were obtained from the patients who underwent coronary artery bypass graft surgery. In cell studies, we performed the cumulative population doubling level assessment, fluorescence-activated cell sorting analysis, and differentiation study. In animal studies, we segregated Sprague-Dawley rats (ischemia-reperfusion model) into three (sham, vehicle, and TAT-MSC) groups based on their corresponding treatment. Trans-thoracic echocardiogram (TTE) was obtained to assess the recovery of heart function in the 1st, 4th, 8th, and 12th week after surgical manipulations. After echocardiographic study, infarcted area of the heart was measured using triphenyl tetrazolium chloride (TTC) stain. Results The sham group exhibited significantly better systolic and diastolic function (SDF) than the other groups did. After one week of TAT-MSC or vehicle injection, the TAT-MSC group exhibited a significant improvement in the E/E' value (25.75 ± 1.09 vs. 24.20 ± 0.91, p < 0.001) compared to the vehicle group. Although statistically insignificant, the trend of improvement in SDF was better in the TAT-MSC group than in the vehicle group. The infarcted area measured by TTC staining was 22.81 ± 6.41% and 29.95 ± 9.09% in the TAT-MSC and vehicle groups, respectively (p = 0.04). Conclusion Although TTE results exhibited insignificant variations in SDF, a trend with improvement in the SDF of the heart was observed in the TAT-MSC group compared to the vehicle group. The infarcted area of heart indicated significant reduction in the TAT-MSC group compared to the vehicle group as confirmed by histopathological study.
Collapse
Affiliation(s)
- Jun Woo Cho
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Min Soo Seo
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Kyung Ku Kang
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Soo Eun Sung
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| |
Collapse
|
17
|
Yao Y, Li Y, Song Q, Hu C, Xie W, Xu C, Chen Q, Wang QK. Angiogenic Factor AGGF1-Primed Endothelial Progenitor Cells Repair Vascular Defect in Diabetic Mice. Diabetes 2019; 68:1635-1648. [PMID: 31092480 PMCID: PMC6905488 DOI: 10.2337/db18-1178] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/06/2019] [Indexed: 12/12/2022]
Abstract
Hyperglycemia-triggered vascular abnormalities are the most serious complications of diabetes mellitus (DM). The major cause of vascular dysfunction in DM is endothelial injury and dysfunction associated with the reduced number and dysfunction of endothelial progenitor cells (EPCs). A major challenge is to identify key regulators of EPCs to restore DM-associated vascular dysfunction. We show that EPCs from heterozygous knockout Aggf1+/- mice presented with impairment of proliferation, migration, angiogenesis, and transendothelial migration as in hyperglycemic mice fed a high-fat diet (HFD) or db/db mice. The number of EPCs from Aggf1+/- mice was significantly reduced. Ex vivo, AGGF1 protein can fully reverse all damaging effects of hyperglycemia on EPCs. In vivo, transplantation of AGGF1-primed EPCs successfully restores blood flow and blocks tissue necrosis and ambulatory impairment in HFD-induced hyperglycemic mice or db/db mice with diabetic hindlimb ischemia. Mechanistically, AGGF1 activates AKT, reduces nuclear localization of Fyn, which increases the nuclear level of Nrf2 and expression of antioxidative genes, and inhibits reactive oxygen species generation. These results suggest that Aggf1 is required for essential function of EPCs, AGGF1 fully reverses the damaging effects of hyperglycemia on EPCs, and AGGF1 priming of EPCs is a novel treatment modality for vascular complications in DM.
Collapse
Affiliation(s)
- Yufeng Yao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yong Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Qixue Song
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Changqin Hu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Wen Xie
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Qiuyun Chen
- Department of Cardiovascular and Metabolic Sciences, NB50, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| | - Qing K. Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Department of Cardiovascular and Metabolic Sciences, NB50, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH
- Corresponding author: Qing K. Wang, , or Qiuyun Chen,
| |
Collapse
|
18
|
The Human Coronary Collateral Circulation, Its Extracardiac Anastomoses and Their Therapeutic Promotion. Int J Mol Sci 2019; 20:ijms20153726. [PMID: 31366096 PMCID: PMC6696371 DOI: 10.3390/ijms20153726] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/04/2019] [Accepted: 07/12/2019] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular disease remains the leading global cause of death, and the number of patients with coronary artery disease (CAD) and exhausted therapeutic options (i.e., percutaneous coronary intervention (PCI), coronary artery bypass grafting (CABG) and medical treatment) is on the rise. Therefore, the evaluation of new therapeutic approaches to offer an alternative treatment strategy for these patients is necessary. A promising research field is the promotion of the coronary collateral circulation, an arterio-arterial network able to prevent or reduce myocardial ischemia in CAD. This review summarizes the basic principles of the human coronary collateral circulation, its extracardiac anastomoses as well as the different therapeutic approaches, especially that of stimulating the extracardiac collateral circulation via permanent occlusion of the internal mammary arteries.
Collapse
|
19
|
Duhme C, Lehwald N, Kehrel BE, Bauchrowitz E, Ngepi A, Schmelzle M, Kolokotronis T, Benhidjeb T, Krüger M, Jurk K, Knoefel WT, Robson SC, Schulte Am Esch J. CD133 + bone marrow stem cells (BMSC) control platelet activation - Role of ectoNTPDase-1 (CD39). Blood Cells Mol Dis 2019; 77:142-148. [PMID: 31075617 DOI: 10.1016/j.bcmd.2019.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/25/2019] [Accepted: 04/26/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND We previously demonstrated CD133+ bone marrow stem cells (BMSC) to promote hepatic proliferation for liver regeneration. Here, we evaluated the capacity of CD133+BMSC to utilize platelets for homing to vasculature and concomitant controlling their aggregability upon ADP stimulation. METHODS CD133+BMSC and platelets were co-cultured along micro endothelial cells under variable flow conditions and tested for homing levels along vasculature. Aggregometry and FACS analysis were utilized to evaluate platelet reactivity following co-incubation ± CD133+BMSC. RT-PCR and FACS analyses served to characterize ADP degrading ectonucleoside triphosphate diphosphohydrolase-1 (ectoNTPDase-1/CD39) expression on various cell types. RESULTS Platelets attracted human CD133+BMSC to autologous micro endothelium under shear stress unaffected by ADP stimulation. However, CD133+BMSC inhibited ADP-mediated platelet activation and aggregation. Latter was dependent on ectoNTPDase-1 expression levels. Platelet aggregatory control was increased with CD133+BMSC compared to CD133+PHSC. Different effects of those stem cell subtypes positively correlated with their FACS-detected expression levels of ectoNTPDase-1. CONCLUSION We provide evidence that CD133+BMSC are capable of controlling ADP-dependent platelet aggregation and activation by direct interaction dependent on cellular expression of ectoNTPDase-1. Whether different capacities of BMSC modulate platelet-depending thrombogenicity at sites of regeneration impact effectiveness and adverse event profiles of regenerative treatment requires further evaluation.
Collapse
Affiliation(s)
- Constanze Duhme
- Department of Surgery A, University Hospital Duesseldorf, Germany
| | - Nadja Lehwald
- Department of Surgery A, University Hospital Duesseldorf, Germany
| | - Beate E Kehrel
- Department of Anesthesiology Intensive Care and Pain Medicine, Experimental and Clinical Hemostasis, University of Muenster, Muenster, Germany
| | | | - Arlette Ngepi
- Department of Surgery A, University Hospital Duesseldorf, Germany
| | | | - Theodoros Kolokotronis
- Center of Visceral Medicine, Department of General and Visceral Surgery, Protestant Hospital of Bethel Foundation, Bielefeld, Germany
| | - Tahar Benhidjeb
- Center of Visceral Medicine, Department of General and Visceral Surgery, Protestant Hospital of Bethel Foundation, Bielefeld, Germany
| | - Martin Krüger
- Center of Visceral Medicine, Department of Gastroenterology and Internal Medicine, Protestant Hospital of Bethel Foundation, Bielefeld, Germany
| | - Kerstin Jurk
- Department of Anesthesiology Intensive Care and Pain Medicine, Experimental and Clinical Hemostasis, University of Muenster, Muenster, Germany; Center for Thrombosis and Hemostasis, Johannes Gutenberg-University, Mainz, Germany
| | | | - Simon C Robson
- The Transplant Institute and Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA, USA
| | - Jan Schulte Am Esch
- Center of Visceral Medicine, Department of General and Visceral Surgery, Protestant Hospital of Bethel Foundation, Bielefeld, Germany.
| |
Collapse
|
20
|
Yokohama A, Yokote K, Maruhashi T. Apheresis on aged patients/donors with complicated backgrounds like ischemic heart disease, arrhythmia, and others. Transfus Apher Sci 2018; 57:619-622. [PMID: 30262215 DOI: 10.1016/j.transci.2018.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Peripheral blood stem cells (PBSCs) are currently one of the most important stem cell sources for hematopoietic stem cell transplantation as well as cell therapy for ischemic heart disease or critical limb ischemia. Thus, it is sometimes necessary to collect autologous PBSCs from donors who have comorbidities. In terms yield, a sufficient number of PBSCs can be collected from donors with comorbidities for performing cell therapy if their age is < 60 years or up to a maximum of 70 years, although the number of PBSCs collected from older donors would probably be lower than that obtained from younger donors. On the other hand, granulocyte colony-stimulating factor (G-CSF) administration sometimes results in severe adverse events (AEs), such as ischemic heart disease and vascular thrombosis. Therefore, it is very important to perform strict medical check-ups according to the standards for donor operations in each country before apheresis. The apheresis procedure and G-CSF administration should be performed after administering the appropriate treatment. There is very less information available regarding AEs related to citrate administration during apheresis in aged donors with complicated medical histories. Medical staff should have knowledge of the electrocardiogram (ECG) QTc prolongation that occurs during apheresis owing to hypocalcemia caused by citrate administration, necessitating electrocardiographic monitoring of patients. Calcium should be administered during apheresis to prevent citrate related symptoms.
Collapse
Affiliation(s)
- Akihiko Yokohama
- Transfusion Service, Gunma University Hospital, Faculty of Medicine, Gunma University, Gunma, Japan.
| | - Keiko Yokote
- Transfusion Service, Gunma University Hospital, Faculty of Medicine, Gunma University, Gunma, Japan; Division of Nursing, Gunma University Hospital, Faculty of Medicine, Gunma University, Gunma, Japan
| | - Takayuki Maruhashi
- Transfusion Service, Gunma University Hospital, Faculty of Medicine, Gunma University, Gunma, Japan
| |
Collapse
|
21
|
Abou-Saleh H, Zouein FA, El-Yazbi A, Sanoudou D, Raynaud C, Rao C, Pintus G, Dehaini H, Eid AH. The march of pluripotent stem cells in cardiovascular regenerative medicine. Stem Cell Res Ther 2018; 9:201. [PMID: 30053890 PMCID: PMC6062943 DOI: 10.1186/s13287-018-0947-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular disease (CVD) continues to be the leading cause of global morbidity and mortality. Heart failure remains a major contributor to this mortality. Despite major therapeutic advances over the past decades, a better understanding of molecular and cellular mechanisms of CVD as well as improved therapeutic strategies for the management or treatment of heart failure are increasingly needed. Loss of myocardium is a major driver of heart failure. An attractive approach that appears to provide promising results in reducing cardiac degeneration is stem cell therapy (SCT). In this review, we describe different types of stem cells, including embryonic and adult stem cells, and we provide a detailed discussion of the properties of induced pluripotent stem cells (iPSCs). We also present and critically discuss the key methods used for converting somatic cells to pluripotent cells and iPSCs to cardiomyocytes (CMs), along with their advantages and limitations. Integrating and non-integrating reprogramming methods as well as characterization of iPSCs and iPSC-derived CMs are discussed. Furthermore, we critically present various methods of differentiating iPSCs to CMs. The value of iPSC-CMs in regenerative medicine as well as myocardial disease modeling and cardiac regeneration are emphasized.
Collapse
Affiliation(s)
- Haissam Abou-Saleh
- Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar
| | - Fouad A. Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ahmed El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, “Attikon” Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Christopher Rao
- Department of Surgery, Queen Elizabeth Hospital, Woolwich, London, UK
| | - Gianfranco Pintus
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Hassan Dehaini
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
22
|
Yang JX, Pan YY, Wang XX, Qiu YG, Mao W. Endothelial progenitor cells in age-related vascular remodeling. Cell Transplant 2018; 27:786-795. [PMID: 29882417 PMCID: PMC6047273 DOI: 10.1177/0963689718779345] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence has demonstrated that endothelial progenitor cells (EPCs) could facilitate the reendothelialization of injured arteries by replacing the dysfunctional endothelial cells, thereby suppressing the formation of neointima. Meanwhile, other findings suggest that EPCs may be involved in the pathogenesis of age-related vascular remodeling. This review is presented to summarize the characteristics of EPCs and age-related vascular remodeling. In addition, the role of EPCs in age-related vascular remodeling and possible solutions for improving the therapeutic effects of EPCs in the treatment of age-related diseases are discussed.
Collapse
Affiliation(s)
- Jin-Xiu Yang
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China.,2 Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Yan-Yun Pan
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Xing-Xiang Wang
- 2 Department of Cardiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Yuan-Gang Qiu
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| | - Wei Mao
- 1 Department of Cardiology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, P.R. China
| |
Collapse
|
23
|
Modulation of Inflammatory Reactions by Low-Dose Ionizing Radiation: Cytokine Release of Murine Endothelial Cells Is Dependent on Culture Conditions. J Immunol Res 2018; 2018:2856518. [PMID: 29967799 PMCID: PMC6008836 DOI: 10.1155/2018/2856518] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/14/2018] [Accepted: 04/05/2018] [Indexed: 01/04/2023] Open
Abstract
Background In many European countries, patients with a variety of chronical inflammatory diseases are treated with low-dose radiotherapy (LD-RT). In contrast to high-dose irradiation given to tumor patients, little is known about radiobiological mechanisms underlying this clinical successful LD-RT application. The objective of this study was to gain a better insight into the modulation of inflammatory reactions after LD-RT on the basis of endothelial cells (EC) as major participants and regulators of inflammation. Methods Three murine EC lines were cultivated under 2D and 3D culture conditions and irradiated with doses from 0.01 Gy to 2 Gy. To simulate an inflammatory situation, cells were activated with TNF-α. After LD-RT, a screening of numerous inflammatory markers was determined by multiplex assay, followed by detailed analyses of four cytokines (KC, MCP-1, RANTES, and G-CSF). Additionally, the monocyte binding to EC was analyzed. Results Cytokine concentrations were dependent on culture condition, IR dose, time point after IR, and EC origin. IR caused nonlinear dose-dependent effects on secretion of the proinflammatory cytokines KC, MCP-1, and RANTES. The monocyte adhesion was significantly enhanced after IR as well as activation. Conclusions The study shows that LD-RT, also using very low radiation doses, has a clear immunomodulatory effect on EC as major participants and regulators of inflammation.
Collapse
|
24
|
Han X, Wu A, Wang J, Chang H, Zhao Y, Zhang Y, Mao Y, Lou L, Gao Y, Zhang D, Li T, Yang T, Wang L, Feng C, Zhao M. Activation and Migration of Adventitial Fibroblasts Contributes to Vascular Remodeling. Anat Rec (Hoboken) 2018; 301:1216-1223. [PMID: 29406614 DOI: 10.1002/ar.23793] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/03/2017] [Accepted: 12/07/2017] [Indexed: 12/26/2022]
Abstract
The rat carotid artery balloon injury model was used to prove the activation and migration of adventitial fibroblasts. We found that at day 7 after injury, adventitial fibroblasts proliferated, transformed into myofibroblasts under transmission electron microscopy in the model group. Simultaneously, we proved that the adventitial cells migrated to the media and intima on seventh day after injury by directly labeled the adventitial cells by the in vivo gene transfer technique. Moreover, we captured the precise moment when the adventitial fibroblasts migrated from the adventitia to the media through the external elastic plate under transmission electron microscope. This study provides direct evidences that adventitial fibroblasts activate and migrate to the media and intima, then actively take part in revascularization. Anat Rec, 301:1216-1223, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiaowan Han
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Aiming Wu
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Jie Wang
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China.,Huayuanlu Community Health Service Center, Beijing 100088, China
| | - Hong Chang
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Yizhou Zhao
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Yan Zhang
- Department of Ultrapathology of the Neurosurgical Institute Affiliated Bayi Brain Hospital, Army General Hospital of PLA, Beijing 100700, China
| | - Yingqiu Mao
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lixia Lou
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Yonghong Gao
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China
| | - Dongmei Zhang
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Tong Li
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Tao Yang
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Lei Wang
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| | - Cuiling Feng
- Peking University People's Hospital, Beijing, China
| | - Mingjing Zhao
- Beijing University of Chinese Medicine, Dongzhimen Hospital, Key Laboratory of Chinese Internal Medicine, Ministry of Education & Beijing, Beijing 100700, China.,Beijing University of Chinese Medicine, Dongzhimen Hospital, Laboratory for Integrated Traditional Chinese and Western Medical Research of Qi-Blood, Beijing 100700, China
| |
Collapse
|
25
|
Van Pham P, Nguyen HT, Vu NB. Evolution of Stem Cell Products in Medicine: Future of Off-the-Shelf Products. STEM CELL DRUGS - A NEW GENERATION OF BIOPHARMACEUTICALS 2018. [DOI: 10.1007/978-3-319-99328-7_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
26
|
Dorobantu M, Popa-Fotea NM, Popa M, Rusu I, Micheu MM. Pursuing meaningful end-points for stem cell therapy assessment in ischemic cardiac disease. World J Stem Cells 2017; 9:203-218. [PMID: 29321822 PMCID: PMC5746641 DOI: 10.4252/wjsc.v9.i12.203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 11/08/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
Abstract
Despite optimal interventional and medical therapy, ischemic heart disease is still an important cause of morbidity and mortality worldwide. Although not included in standard of care rehabilitation, stem cell therapy (SCT) could be a solution for prompting cardiac regeneration. Multiple studies have been published from the beginning of SCT until now, but overall no unanimous conclusion could be drawn in part due to the lack of appropriate end-points. In order to appreciate the impact of SCT, multiple markers from different categories should be considered: Structural, biological, functional, physiological, but also major adverse cardiac events or quality of life. Imaging end-points are among the most used - especially left ventricle ejection fraction (LVEF) measured through different methods. Other imaging parameters are infarct size, myocardial viability and perfusion. The impact of SCT on all of the aforementioned end-points is controversial and debatable. 2D-echocardiography is widely exploited, but new approaches such as tissue Doppler, strain/strain rate or 3D-echocardiography are more accurate, especially since the latter one is comparable with the MRI gold standard estimation of LVEF. Apart from the objective parameters, there are also patient-centered evaluations to reveal the benefits of SCT, such as quality of life and performance status, the most valuable from the patient point of view. Emerging parameters investigating molecular pathways such as non-coding RNAs or inflammation cytokines have a high potential as prognostic factors. Due to the disadvantages of current techniques, new imaging methods with labelled cells tracked along their lifetime seem promising, but until now only pre-clinical trials have been conducted in humans. Overall, SCT is characterized by high heterogeneity not only in preparation, administration and type of cells, but also in quantification of therapy effects.
Collapse
Affiliation(s)
- Maria Dorobantu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Bucharest 014461, Romania
| | | | - Mihaela Popa
- Carol Davila, University of Medicine, "Carol Davila" University of Medicine and Pharmacy Bucharest, Bucharest 020022, Romania
| | - Iulia Rusu
- Carol Davila, University of Medicine, "Carol Davila" University of Medicine and Pharmacy Bucharest, Bucharest 020022, Romania
| | - Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Bucharest 014461, Romania.
| |
Collapse
|
27
|
Hacobian A, Hercher D. Pushing the Right Buttons: Improving Efficacy of Therapeutic DNA Vectors. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:226-239. [PMID: 29264951 DOI: 10.1089/ten.teb.2017.0353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Gene therapy represents a potent therapeutical application for regenerative medicine. So far, viral and nonviral approaches suffer from major drawbacks hindering efficient gene therapeutic applicability: the immunogenicity of viral systems on the one hand, and the low gene transfer efficiency of nonviral systems on the other hand. Therefore, there is a high demand for improvements of therapeutical systems at several levels. This review summarizes different DNA vector modifications to enhance biological efficacy and efficiency of therapeutical vectors, aiming for low toxicity, high specificity, and biological efficacy-the cornerstones for successful translation of gene therapy into the clinic. We aim to provide a step-by-step instruction to optimize their vectors to achieve the desired outcome of gene therapy. Our review provides the means to either construct a potent gene therapeutic vector de novo or to specifically address a bottleneck in the chain of events mandatory for therapeutic success. Although most of the introduced techniques can be translated into different areas, this review primarily addresses improvements for applications in transient gene therapy in the field of tissue engineering.
Collapse
Affiliation(s)
- Ara Hacobian
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Department of Molecular Biology, AUVA Research Center, The Austrian Cluster for Tissue Regeneration , Vienna, Austria
| | - David Hercher
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Department of Molecular Biology, AUVA Research Center, The Austrian Cluster for Tissue Regeneration , Vienna, Austria
| |
Collapse
|
28
|
Guo M, Shi JH, Wang PL, Shi DZ. Angiogenic Growth Factors for Coronary Artery Disease: Current Status and Prospects. J Cardiovasc Pharmacol Ther 2017; 23:130-141. [PMID: 29025278 DOI: 10.1177/1074248417735399] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Ming Guo
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun-He Shi
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA
| | - Pei-Li Wang
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Da-Zhuo Shi
- China Heart Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
29
|
Steinhoff G, Nesteruk J, Wolfien M, Große J, Ruch U, Vasudevan P, Müller P. Stem cells and heart disease - Brake or accelerator? Adv Drug Deliv Rev 2017; 120:2-24. [PMID: 29054357 DOI: 10.1016/j.addr.2017.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022]
Abstract
After two decades of intensive research and attempts of clinical translation, stem cell based therapies for cardiac diseases are not getting closer to clinical success. This review tries to unravel the obstacles and focuses on underlying mechanisms as the target for regenerative therapies. At present, the principal outcome in clinical therapy does not reflect experimental evidence. It seems that the scientific obstacle is a lack of integration of knowledge from tissue repair and disease mechanisms. Recent insights from clinical trials delineate mechanisms of stem cell dysfunction and gene defects in repair mechanisms as cause of atherosclerosis and heart disease. These findings require a redirection of current practice of stem cell therapy and a reset using more detailed analysis of stem cell function interfering with disease mechanisms. To accelerate scientific development the authors suggest intensifying unified computational data analysis and shared data knowledge by using open-access data platforms.
Collapse
Affiliation(s)
- Gustav Steinhoff
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Julia Nesteruk
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Markus Wolfien
- University Rostock, Institute of Computer Science, Department of Systems Biology and Bioinformatics, Ulmenstraße 69, 18057 Rostock, Germany.
| | - Jana Große
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Ulrike Ruch
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Praveen Vasudevan
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Paula Müller
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| |
Collapse
|
30
|
Rao KS, Spees JL. Harnessing Epicardial Progenitor Cells and Their Derivatives for Rescue and Repair of Cardiac Tissue After Myocardial Infarction. ACTA ACUST UNITED AC 2017; 3:149-158. [PMID: 29057207 DOI: 10.1007/s40610-017-0066-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Ischemic heart disease and stroke lead to the greatest number of deaths worldwide. Despite decreased time to intervention and improvements in the standard of care, 1 out of 5 patients that survive a myocardial infarction (MI) still face long-term chronic heart failure and a 5-year mortality rate of about 50%. Based on their multi-potency for differentiation and paracrine activity, epicardial cells and their derivatives have potential to rescue jeopardized tissue and/or promote cardiac regeneration. Here we review the diagnosis and treatment of MI, basic epicardial cell biology, and potential treatment strategies designed to harness the reparative properties of epicardial cells. RECENT FINDINGS During cardiac development, epicardial cells covering the surface of the heart generate migratory progenitor cells that contribute to the coronary vasculature and the interstitial fibroblasts. Epicardial cells also produce paracrine signals required for myocardial expansion and cardiac growth. In adults with myocardial infarction, epicardial cells and their derivatives provide paracrine factors that affect myocardial remodeling and repair. At present, the intrinsic mechanisms and extrinsic signals that regulate epicardial cell fate and paracrine activity in adults remain poorly understood. SUMMARY Human diseases that result in heart failure due to negative remodeling or extensive loss of viable cardiac tissue require new, effective treatments. Improved understanding of epicardial cell function(s) and epicardial-mediated secretion of growth factors, cytokines and hormones during cardiac growth, homeostasis and injury may lead to new ways to treat patients with myocardial infarction.
Collapse
Affiliation(s)
- Krithika S Rao
- Department of Medicine, Stem Cell Core, University of Vermont, Colchester, VT 05446
- Cardiovascular Research Institute, University of Vermont, Colchester, VT 05446
| | - Jeffrey L Spees
- Department of Medicine, Stem Cell Core, University of Vermont, Colchester, VT 05446
- Cardiovascular Research Institute, University of Vermont, Colchester, VT 05446
| |
Collapse
|
31
|
Abstract
Stroke remains a leading cause of death and disability worldwide. An increasing number of animal studies and preclinical trials have, however, provided evidence that regenerative cell-based therapies can lead to functional recovery in stroke patients. Stem cells can differentiate into neural lineages to replace lost neurons. Moreover, they provide trophic support to tissue at risk in the penumbra surrounding the infarct area, enhance vasculogenesis, and help promote survival, migration, and differentiation of the endogenous precursor cells after stroke. Stem cells are highly migratory and seem to be attracted to areas of brain pathology such as ischemic regions. The pathotropism may follow the paradigm of stem cell homing to bone marrow and leukocytes migrating to inflammatory tissue. The molecular signaling therefore may involve various chemokines, cytokines, and integrins. Among these, stromal cell-derived factor-1 (SDF-1)/CXC chemokine receptor-4 (CXCR4) signaling is required for the interaction of stem cells and ischemia-damaged host tissues. SDF-1 is secreted primarily by bone marrow fibroblasts and is required for BMSC homing to bone marrow. Overexpression of SDF-1 in ischemic tissues has been found to enhance stem cell recruitment from peripheral blood and to induce neoangiogenesis. Furthermore, SDF-1 expression in the lesioned area peaked within 7 days postischemia, in concordance with the time window of G-CSF therapy for stroke. Recent data have shown that SDF-1 expression is directly proportional to reduced tissue oxygen tension. SDF-1 gene expression is regulated by hypoxic-inducible factor-1 (HIF-1), a hypoxia-dependent stabilization transcription factor. Thus, ischemic tissue may recruit circulating progenitors regulated by hypoxia through differential expression of HIF-1α and SDF-1. In addition to SDF-1, β2-integrins also play a role in the homing of hematopoietic progenitor cells to sites of ischemia and are critical for their neovascularization capacity. In our recent report, increased expression of β1-integrins apparently contributed to the local neovasculization of the ischemic brain as well as its functional recovery. Identification of the molecular pathways involved in stem cell homing into the ischemic areas could pave the way for the development of new treatment regimens, perhaps using small molecules, designed to enhance endogeneous mobilization of stem cells in various disease states, including chronic stroke and other neurodegenerative diseases. For maximal functional recovery, however, regenerative therapy may need to follow combinatorial approaches, which may include cell replacement, trophic support, protection from oxidative stress, and the neutralization of the growth-inhibitory components for endogenous neuronal stem cells.
Collapse
Affiliation(s)
- Ying-Chao Chang
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Guang University College of Medicine, Kaohsiung, Taiwan
| | - Woei-Cherng Shyu
- Neuro-Medical Scientific Center, Tzu-Chi Buddhist General Hospital, Tzu-Chi University, Hualien, Taiwan
| | - Shinn-Zong Lin
- Neuro-Medical Scientific Center, Tzu-Chi Buddhist General Hospital, Tzu-Chi University, Hualien, Taiwan
| | - Hung Li
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
32
|
Stamm C, Liebold A, Steinhoff G, Strunk D. Stem Cell Therapy for Ischemic Heart Disease: Beginning or End of the Road? Cell Transplant 2017; 15 Suppl 1:S47-56. [PMID: 16826795 DOI: 10.3727/000000006783982313] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Despite improvements in emergency treatment, myocardial infarction is often the beginning of a downward spiral leading to congestive heart failure. Other than heart transplantation, current therapeutic means aim at enabling the organism to survive with a heart that is working at a fraction of its original capacity. It is therefore no surprise that cardiac stem cell therapy has raised many hopes. However, neither the ideal source and type of stem cell nor the critical cell number and mode of application have been defined so far. Early reports on myocardial repair by adult bone marrow stem cells from rodent models promoted an unparalleled boost of clinical and experimental cell therapy studies. The phenomenon of stem/progenitor cell-induced angiogenesis in ischemic myocardium has ever since been reproduced by numerous groups in a variety of small and large animal models. Myogenesis, however, is an altogether different matter. Many of the initial clinical studies were fueled by the suggestion that early hematopoietic stem cells have a plasticity high enough to enable cross-lineage differentiation into cells of cardiomyocyte phenotype, but the initial enthusiasm has largely faded. The myogenic potential of stroma cell-derived mesenchymal stem cells is much better documented in animal models, but transfer to the clinical setting faces a variety of obstacles. In clinical pilot trials, we and others have demonstrated the feasibility and safety of administering progenitor cells derived from autologous bone marrow to the myocardium of patients with ischemic heart disease. Clinical efficacy data are still rare, but the few controlled trials that have been completed uniformly show a tendency towards better heart function in cell-treated patients. This review is an attempt to describe the scientific basis for cardiac cell therapy from the point of view of the clinician, focusing on problems that arise with beginning translation into the clinical setting.
Collapse
Affiliation(s)
- Christof Stamm
- Department of Cardiac Surgery, University of Rostock, Germany.
| | | | | | | |
Collapse
|
33
|
Nigro P, Bassetti B, Cavallotti L, Catto V, Carbucicchio C, Pompilio G. Cell therapy for heart disease after 15 years: Unmet expectations. Pharmacol Res 2017; 127:77-91. [PMID: 28235633 DOI: 10.1016/j.phrs.2017.02.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/07/2017] [Accepted: 02/16/2017] [Indexed: 12/17/2022]
Abstract
Over the past two decades cardiac cell therapy (CCT) has emerged as a promising new strategy to cure heart diseases at high unmet need. Thousands of patients have entered clinical trials for acute or chronic heart conditions testing different cell types, including autologous or allogeneic bone marrow (BM)-derived mononuclear or selected cells, BM- or adipose tissue-derived mesenchymal cells, or cardiac resident progenitors based on their potential ability to regenerate scarred or dysfunctional myocardium. Nowadays, the original enthusiasm surrounding the regenerative medicine field has been cushioned by a cumulative body of evidence indicating an inefficient or modest efficacy of CCT in improving cardiac function, along with the continued lack of indisputable proof for long-term prognostic benefit. In this review, we have firstly comprehensively outlined the positive and negative results of cell therapy studies in patients with acute myocardial infarction, refractory angina and chronic heart failure. Next, we have discussed cell therapy- and patient-related variables (e.g. cell intrinsic and extrinsic characteristics as well as criteria of patient selection and proposed methodologies) that might have dampened the efficacy of past cell therapy trials. Finally, we have addressed critical factors to be considered before embarking on further clinical trials.
Collapse
Affiliation(s)
- Patrizia Nigro
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy
| | - Beatrice Bassetti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy
| | - Laura Cavallotti
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy
| | - Valentina Catto
- Cardiac Arrhythmia Research Centre, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy
| | - Corrado Carbucicchio
- Cardiac Arrhythmia Research Centre, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino-IRCCS, via Carlo Parea 4, 20138, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, via Festa del Perdono 7, 20122, Milan, Italy.
| |
Collapse
|
34
|
Implantation of a Poly-L-Lactide GCSF-Functionalized Scaffold in a Model of Chronic Myocardial Infarction. J Cardiovasc Transl Res 2017; 10:47-65. [PMID: 28116550 PMCID: PMC5323505 DOI: 10.1007/s12265-016-9718-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 11/03/2016] [Indexed: 12/17/2022]
Abstract
A previously developed poly-l-lactide scaffold releasing granulocyte colony-stimulating factor (PLLA/GCSF) was tested in a rabbit chronic model of myocardial infarction (MI) as a ventricular patch. Control groups were constituted by healthy, chronic MI and nonfunctionalized PLLA scaffold. PLLA-based electrospun scaffold efficiently integrated into a chronic infarcted myocardium. Functionalization of the biopolymer with GCSF led to increased fibroblast-like vimentin-positive cellular colonization and reduced inflammatory cell infiltration within the micrometric fiber mesh in comparison to nonfunctionalized scaffold; PLLA/GCSF polymer induced an angiogenetic process with a statistically significant increase in the number of neovessels compared to the nonfunctionalized scaffold; PLLA/GCSF implanted at the infarcted zone induced a reorganization of the ECM architecture leading to connective tissue deposition and scar remodeling. These findings were coupled with a reduction in end-systolic and end-diastolic volumes, indicating a preventive effect of the scaffold on ventricular dilation, and an improvement in cardiac performance.
Collapse
|
35
|
Abstract
Improvements in the care of patients with ischemic cardiovascular disease have led to improved survival but also a burgeoning population of patients with advanced ischemic heart disease. Cell therapies offer a novel approach toward cardiac "rejuvenation" via stimulation of new blood vessel growth, enhancing tissue perfusion, and via preservation or even regeneration of myocardial tissue, leading to improvements in cardiac performance after myocardial infarction and in patients with advanced heart failure. Here, we summarize and offer some thoughts on the state of the field of cell therapy for ischemic heart disease, targeting three separate conditions that have been the subject of significant clinical research: enhancing left ventricular recovery after MI, improving outcomes and symptoms in patients with congestive heart failure (CHF), and treatment of patients with refractory angina, despite maximal medical therapy.
Collapse
Affiliation(s)
- Thomas J Povsic
- Duke Clinical Research Institute and Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, 27708, USA.
| |
Collapse
|
36
|
Kang J, Kim TW, Hur J, Kim HS. Strategy to Prime the Host and Cells to Augment Therapeutic Efficacy of Progenitor Cells for Patients with Myocardial Infarction. Front Cardiovasc Med 2016; 3:46. [PMID: 27933299 PMCID: PMC5121226 DOI: 10.3389/fcvm.2016.00046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/08/2016] [Indexed: 11/23/2022] Open
Abstract
Cell therapy in myocardial infarction (MI) is an innovative strategy that is regarded as a rescue therapy to repair the damaged myocardium and to promote neovascularization for the ischemic border zone. Among several stem cell sources for this purpose, autologous progenitors from bone marrow or peripheral blood would be the most feasible and safest cell-source. Despite the theoretical benefit of cell therapy, this method is not widely adopted in the actual clinical practice due to its low therapeutic efficacy. Various methods have been used to augment the efficacy of cell therapy in MI, such as using different source of progenitors, genetic manipulation of cells, or priming of the cells or hosts (patients) with agents. Among these methods, the strategy to augment the therapeutic efficacy of the autologous peripheral blood mononuclear cells (PBMCs) by priming agents may be the most feasible and the safest method that can be applied directly to the clinic. In this review, we will discuss the current status and future directions of priming PBMCs or patients, as for cell therapy of MI.
Collapse
Affiliation(s)
- Jeehoon Kang
- Department of Medicine, Seoul National University Hospital, Seoul, South Korea; Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science & Technology, Seoul National University, Seoul, South Korea
| | - Tae-Won Kim
- Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science & Technology, Seoul National University, Seoul, South Korea; National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, South Korea
| | - Jin Hur
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital , Seoul , South Korea
| | - Hyo-Soo Kim
- Department of Medicine, Seoul National University Hospital, Seoul, South Korea; Molecular Medicine & Biopharmaceutical Science, Graduate School of Convergence Science & Technology, Seoul National University, Seoul, South Korea; National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
37
|
Feyen DA, Gaetani R, Doevendans PA, Sluijter JP. Stem cell-based therapy: Improving myocardial cell delivery. Adv Drug Deliv Rev 2016; 106:104-115. [PMID: 27133386 DOI: 10.1016/j.addr.2016.04.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/19/2016] [Accepted: 04/20/2016] [Indexed: 12/15/2022]
Abstract
Stem cell-based therapies form an exciting new class of medicine that attempt to provide the body with the building blocks required for the reconstruction of damaged organs. However, delivering cells to the correct location, while preserving their integrity and functional properties, is a complex undertaking. These challenges have led to the development of a highly dynamic interdisciplinary research field, wherein medical, biological, and chemical sciences have collaborated to develop strategies to overcome the physiological barriers imposed on the cellular therapeutics. In this respect, improving the acute retention and subsequent survival of stem cells is key to effectively increase the effect of the therapy, while proper tissue integration is imperative for stem cells to functionally replace lost cells in damaged organs. In this review, we will use the heart as an example to highlight the current knowledge of therapeutic stem cell utilization, the existing pitfalls and limitations, and the approaches that have been developed to overcome them.
Collapse
|
38
|
Lee SH, Hong JH, Cho KH, Noh JW, Cho HJ. Discrepancy between short-term and long-term effects of bone marrow-derived cell therapy in acute myocardial infarction: a systematic review and meta-analysis. Stem Cell Res Ther 2016; 7:153. [PMID: 27765070 PMCID: PMC5072331 DOI: 10.1186/s13287-016-0415-z] [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: 07/11/2016] [Revised: 09/18/2016] [Accepted: 09/27/2016] [Indexed: 12/11/2022] Open
Abstract
Background Bone marrow-derived cell therapy has been used to treat acute myocardial infarction. However, the therapeutic efficacy of this approach remains controversial. Here, we performed a systematic review and meta-analysis to evaluate short-term and long-term effectiveness of bone marrow-derived therapy. Methods We searched eight databases (Ovid-Medline, Ovid-EMBASE, Cochrane Library, KoreaMed, KMBASE, KISS, RISS, and KisTi) up to December 2014. Demographic characteristics, clinical outcomes, and adverse events were analyzed. We identified 5534 potentially relevant studies; 405 were subjected to a full-text review. Forty-three studies with 2635 patients were included in this review. Results No safety issues related to cell injection were reported during follow-up. At 6 months, cell-injected patients showed modest improvements in left ventricular ejection fraction (LVEF) compared with the control group. However, there were no differences between groups at other time points. In the cardiac MRI analysis, there were no significant differences in infarct size reduction between groups. Interestingly, mortality tended to be reduced at the 3-year follow-up, and at the 5-year follow-up, cell injection significantly decreased all-cause mortality. Conclusions This meta-analysis demonstrated discrepancies between short-term LV functional improvement and long-term all-cause mortality. Future clinical trials should include long-term follow-up outcomes to validate the therapeutic efficacy of cell therapy.
Collapse
Affiliation(s)
- Seon Heui Lee
- Department of Nursing Science, College of Nursing, Gachon University, Seoul, South Korea
| | - Jin Hyuk Hong
- Department of Biostatistics, Korea University College of Medicine, Seoul, South Korea
| | - Kyoung Hee Cho
- Department of Public Health, Graduate School, Yonsei University, Seoul, South Korea
| | - Jin-Won Noh
- Department of Healthcare Management, Eulji University, 212 Yangji-dong, Sujeong-gu, Seongnam-si, Gyeonggi, 461-713, South Korea.
| | - Hyun-Jai Cho
- Cardiovascular Center, Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| |
Collapse
|
39
|
Falavarjani KG, Habibi A, Pourhabibi A, Hosseini SB, Modarres M, Parvaresh MM, Shahriari-Ahmadi A. Retinal Toxicity of Intravitreal Granulocyte Colony-Stimulating Factor in Rabbit Eyes. Curr Eye Res 2016; 42:636-639. [DOI: 10.1080/02713683.2016.1227447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Khalil Ghasemi Falavarjani
- Department of Ophthalmology, Eye Research Center, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Abbas Habibi
- Department of Ophthalmology, Eye Research Center, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Arash Pourhabibi
- Department of Ophthalmology, Eye Research Center, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mehdi Modarres
- Department of Ophthalmology, Eye Research Center, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Parvaresh
- Department of Ophthalmology, Eye Research Center, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Shahriari-Ahmadi
- Department of Internal Medicine, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Prisco AR, Hoffmann BR, Kaczorowski CC, McDermott-Roe C, Stodola TJ, Exner EC, Greene AS. Tumor Necrosis Factor α Regulates Endothelial Progenitor Cell Migration via CADM1 and NF-kB. Stem Cells 2016; 34:1922-33. [PMID: 26867147 PMCID: PMC4931961 DOI: 10.1002/stem.2339] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 01/28/2016] [Indexed: 02/06/2023]
Abstract
Shortly after the discovery of endothelial progenitor cells (EPCs) in 1997, many clinical trials were conducted using EPCs as a cellular based therapy with the goal of restoring damaged organ function by inducing growth of new blood vessels (angiogenesis). Results were disappointing, largely because the cellular and molecular mechanisms of EPC-induced angiogenesis were not clearly understood. Following injection, EPCs must migrate to the target tissue and engraft prior to induction of angiogenesis. In this study EPC migration was investigated in response to tumor necrosis factor α (TNFα), a pro-inflammatory cytokine, to test the hypothesis that organ damage observed in ischemic diseases induces an inflammatory signal that is important for EPC homing. In this study, EPC migration and incorporation were modeled in vitro using a coculture assay where TNFα treated EPCs were tracked while migrating toward vessel-like structures. It was found that TNFα treatment of EPCs increased migration and incorporation into vessel-like structures. Using a combination of genomic and proteomic approaches, NF-kB mediated upregulation of CADM1 was identified as a mechanism of TNFα induced migration. Inhibition of NF-kB or CADM1 significantly decreased migration of EPCs in vitro suggesting a role for TNFα signaling in EPC homing during tissue repair. Stem Cells 2016;34:1922-1933.
Collapse
Affiliation(s)
- Anthony R. Prisco
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI
- Medical College of Wisconsin, Biotechnology and Bioengineering Center, Milwaukee, WI
| | - Brian R. Hoffmann
- Medical College of Wisconsin, Biotechnology and Bioengineering Center, Milwaukee, WI
- Medical College of Wisconsin, Department of Medicine, Division of Cardiology, Cardiovascular Center, Milwaukee, WI
| | - Catherine C. Kaczorowski
- University of Tennessee Health Science Center, Department of Anatomy and Neurobiology, Memphis, TN
| | - Chris McDermott-Roe
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI
- Medical College of Wisconsin, Human and Molecular Genetics Center, Milwaukee, WI
| | - Timothy J. Stodola
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI
- Medical College of Wisconsin, Biotechnology and Bioengineering Center, Milwaukee, WI
| | - Eric C. Exner
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI
- Medical College of Wisconsin, Biotechnology and Bioengineering Center, Milwaukee, WI
| | - Andrew S. Greene
- Medical College of Wisconsin, Department of Physiology, Milwaukee, WI
- Medical College of Wisconsin, Biotechnology and Bioengineering Center, Milwaukee, WI
| |
Collapse
|
41
|
Abstract
Despite that advances in medical treatment and interventional procedures have reduced the mortality rate in patients with coronary artery disease, the number of patients with refractory myocardial ischemia and congestive heart failure is rapidly increasing. Experimental studies have demonstrated that bone marrow (BM) contains adult stem cells that can induce neovascularization and improve heart function in ischemic myocardium. Recent insights into the understanding of the mechanisms involved in proliferation, recruitment, mobilization, and incorporation of BM-derived stem cells into the myocardium and blood vessels have prompted development of cellular transplantation therapy for heart diseases refractory to conventional therapy. Initial preliminary clinical studies indicated potential clinical benefit of BM therapy in patients with acute myocardial infarction and chronic myocardial ischemia. Nevertheless, many obstacles remain, such as long-term safety and optimal timing and treatment strategies for BM cell therapy, and these issues need to be addressed in rationally designed, randomized clinical trials.
Collapse
Affiliation(s)
- Hung-Fat Tse
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China. hftse@ hkucc.hku.hk
| | | |
Collapse
|
42
|
Vento A, Hämmäinen P, Pätilä T, Kankuri E, Harjula A. Somatic Stem Cell Transplantation for the Failing Heart. Scand J Surg 2016; 96:131-9. [PMID: 17679355 DOI: 10.1177/145749690709600208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- A Vento
- Cell Therapy Research Consortium, Helsinki University Central Hospital, 3rd Department of Surgery, Meilahti Hospital, Helsinki, Finland
| | | | | | | | | |
Collapse
|
43
|
Roan JN, Luo CY, Tsai MD, Wu IS, Chang SW, Huang CC, Tsai YS, Lam CF. Mobilization of Endothelial Progenitor Cells Following Creation of Arteriovenous Access in Patients with End-Stage Renal Disease. ACTA CARDIOLOGICA SINICA 2016; 31:24-32. [PMID: 27122843 DOI: 10.6515/acs20140310e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND A patent arteriovenous (AV) fistula induces activation of regional vascular endothelium and vascular shear force. Shear stress is an important physiological force in mobilizing endothelial progenitor cells (EPCs). This study aimed to explore the perioperative changes of circulating EPC levels for patients who require hemodialysis and underwent radiocephalic fistula operation. METHODS This prospective cohort study included patients who received a radiocephalic fistula surgery when they were between 25 and 65 years of age. The subjects were followed for 90 days postoperatively for any stenotic events or immaturity of the fistula. Blood samples were obtained on the day before surgery and at postoperation day (POD) 3 and 30. CD133+/KDR+ cells, defined as EPCs, were analyzed using flow cytometry. Blood flow of the fistula was followed on POD 3 and 30. RESULTS A total of 30 patients were enrolled in the study from July 2009 to December 2011. One patient dropped out of the study and seven patients developed a stenotic (or immature) AV fistula (7/29, 24.1%). There were positive linear relationships between EPC numbers and shear rate postoperatively, which were more significant on POD 30. In addition, postoperative mobilization of EPCs was significantly higher in patients who developed a stenotic fistula than those without. CONCLUSIONS The mobilization of circulating EPCs correlated with a compromised arteriovenous fistula. The biological significance of increased EPC numbers need to be determined in future studies. KEY WORDS Arteriovenous fistula; Endothelial progenitor cells.
Collapse
Affiliation(s)
- Jun-Neng Roan
- Institute of Clinical Medicine; ; Division of Cardiovascular Surgery, Department of Surgery, National Cheng Kung University College of Medicine and Hospital; ; Department of Surgery, Tainan Municipal Hospital
| | - Chwan-Yau Luo
- Division of Cardiovascular Surgery, Department of Surgery, National Cheng Kung University College of Medicine and Hospital
| | - Mang-Da Tsai
- Division of Cardiovascular Surgery, Department of Surgery, National Cheng Kung University College of Medicine and Hospital
| | - I-Shuan Wu
- Department of Surgery, Tainan Municipal Hospital
| | - Shih-Wei Chang
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan
| | - Chien-Chi Huang
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan
| | | | - Chen-Fuh Lam
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan; ; Department of Anesthesiology, Buddhist Tzu-Chi General Hospital and Tzu-Chi University School of Medicine, Hualien, Taiwan
| |
Collapse
|
44
|
Rurali E, Bassetti B, Perrucci GL, Zanobini M, Malafronte C, Achilli F, Gambini E. BM ageing: Implication for cell therapy with EPCs. Mech Ageing Dev 2016; 159:4-13. [PMID: 27045606 DOI: 10.1016/j.mad.2016.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/22/2016] [Accepted: 04/01/2016] [Indexed: 12/17/2022]
Abstract
The bone marrow (BM) is a well-recognized source of stem/progenitor cells for cell therapy in cardiovascular diseases (CVDs). Preclinical and clinical studies suggest that endothelial progenitor cells (EPCs) contribute to reparative process of vascular endothelium and participate in angiogenesis. As for all organs and cells across the lifespan, BM and EPCs are negatively impacted by ageing due to microenvironment modifications and EPC progressive dysfunctions. The encouraging results in terms of neovascularization observed in young animals after EPC administration were mitigated in aged patients treated for ischemic CVDs. The limited efficacy of EPC-based therapy in clinical setting might be ascribed at least partly to ageing. In this review, we comprehensively discussed the age-related changes of BM and EPCs and their implication for cardiovascular cell-therapies. Finally, we examined alternative approaches under investigation to enhance EPC potency.
Collapse
Affiliation(s)
- Erica Rurali
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Beatrice Bassetti
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Gianluca Lorenzo Perrucci
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Marco Zanobini
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | - Felice Achilli
- Cardiology Department, Azienda Ospedaliera San Gerardo, Monza, Italy
| | - Elisa Gambini
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino, IRCCS, Milan, Italy.
| |
Collapse
|
45
|
Massara M, Barillà D, De Caridi G, Serra R, Volpe A, Surace R, Foti G, Marcuccio D, Pucci G, Volpe P. Application of autologous platelet-rich plasma to enhance wound healing after lower limb revascularization: A case series and literature review. Semin Vasc Surg 2016; 28:195-200. [PMID: 27113287 DOI: 10.1053/j.semvascsurg.2016.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dermal tissue loss in patients affected by critical limb ischemia represents a serious wound-healing problem, with high morbidity, prolonged hospital stay, and high patient care costs. Treatment of ischemic foot lesions requires limb revascularization by endovascular or open surgical intervention and individualized patient-specific wound care, including antibiotic therapy; devitalized/infected wound debridement; and advanced wound dressing. In selected patients, spinal cord stimulation, vacuum-assisted closure therapy, and bioengineered tissue or skin substitutes and growth factors have been shown to improve wound healing. In this study, we present our preliminary results on topical application of autologous platelet-rich plasma to enhance the process of wound healing after revascularization of lower limbs in patients affected by critical limb ischemia.
Collapse
Affiliation(s)
- Mafalda Massara
- Unit of Vascular Surgery, Bianchi-Melacrino-Morelli Hospital, Reggio Calabria, Via Melacrino n.1, 89100, Italy; Cardiovascular and Thoracic Department, Policlinico G. Martino Hospital, University of Messina, Messina, Italy.
| | - David Barillà
- Unit of Vascular Surgery, Bianchi-Melacrino-Morelli Hospital, Reggio Calabria, Via Melacrino n.1, 89100, Italy
| | - Giovanni De Caridi
- Cardiovascular and Thoracic Department, Policlinico G. Martino Hospital, University of Messina, Messina, Italy
| | - Raffaele Serra
- Department of Surgical and Medical Science, University Magna Graecia of Catanzaro, Catanzaro, Italy; Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | | | - Rosangela Surace
- Calabria Cord Blood Bank, Service of Immunohaematology and Transfusion Medicine, Bianchi-Melacrino-Morelli Hospital, Reggio Calabria, Italy
| | - Giovanni Foti
- Calabria Cord Blood Bank, Service of Immunohaematology and Transfusion Medicine, Bianchi-Melacrino-Morelli Hospital, Reggio Calabria, Italy
| | - Daniela Marcuccio
- Calabria Cord Blood Bank, Service of Immunohaematology and Transfusion Medicine, Bianchi-Melacrino-Morelli Hospital, Reggio Calabria, Italy
| | - Giulia Pucci
- Calabria Cord Blood Bank, Service of Immunohaematology and Transfusion Medicine, Bianchi-Melacrino-Morelli Hospital, Reggio Calabria, Italy
| | - Pietro Volpe
- Unit of Vascular Surgery, Bianchi-Melacrino-Morelli Hospital, Reggio Calabria, Via Melacrino n.1, 89100, Italy
| |
Collapse
|
46
|
Impact of intracoronary injection of CD133+ bone marrow stem cells on coronary atherosclerotic progression in patients with STEMI. Coron Artery Dis 2016; 27:5-12. [DOI: 10.1097/mca.0000000000000302] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
47
|
Rinaldi B, Finicelli M, Donniacuo M, Di Bernardo G, Gritti G, Gaudio SD, Forte A, Peluso G, Cipollaro M, Rossi F, Galderisi U. G-CSF contributes at the healing of tunica media of arteriotomy-injured rat carotids by promoting differentiation of vascular smooth muscle cells. J Cell Physiol 2016; 231:215-223. [PMID: 26096962 DOI: 10.1002/jcp.25074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/08/2015] [Indexed: 12/24/2022]
Abstract
Restenosis is a complex pathophysiological disease whose causative mechanisms are not fully understood. Previous studies allowed us to demonstrate the efficacy of bone marrow mesenchymal stromal cells (MSCs) transplantation in limiting the pathophysiological remodeling in a model of arteriotomy-induced (re) stenosis. In the current research we studied the effectiveness of G-CSF treatment on male rate rats that were subjected carotid arteriotomy in order to evaluate a potentially effective non-invasive strategy that recapitulates the MSC-mediated recovery of injured vessels. WKY male rats were subjected carotid arteriotomy and given a nine day treatment (3 days pre- to 6 days post-arteriotomy) with G-CSF or saline. Carotids were harvested 7 and 30 days following arteriotomy (early- and late-phase, respectively). Although morphometrical analysis did not reveal differences in lumen narrowing between G-CSF- and PBS-carotids 30 days following arteriotomy, we detected a noticeable conservative effect of G-CSF treatment on vascular wall morphology. Histological and molecular analysis revealed an increase in cellularity within the tunica media with a concomitant increase of the VSMCs differentiation markers both at early- and late-phases of (re) stenotic response in G-CSF-treated carotids (Sm22-alpha, Myocd, and Smtn). These findings were accompanied by the downregulation of oxidative stress-related genes in G-CSF-injured rats. The effect exerted by G-CSF in our model of arteriotomy-induced (re) stenosis seemed support the recovery of the architecture of the tunica media of injured vessels by: (i) inducing VSMCs differentiation; and (ii) limiting the oxidative-stress response induced by arteriotomy.
Collapse
Affiliation(s)
- Barbara Rinaldi
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Centre of Excellence for Cardiovascular Diseases, Second University of Naples, Naples, Italy
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Second University of Naples, Naples, Italy
| | - Mauro Finicelli
- Institute of Biomedicine and Bioresources (IBBR); CNR, Naples, Italy
| | - Maria Donniacuo
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Second University of Naples, Naples, Italy
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Second University of Naples, Naples, Italy
| | - Giulia Gritti
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Second University of Naples, Naples, Italy
| | - Stefania Del Gaudio
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Second University of Naples, Naples, Italy
| | - Amalia Forte
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Second University of Naples, Naples, Italy
| | - Gianfranco Peluso
- Institute of Biomedicine and Bioresources (IBBR); CNR, Naples, Italy
| | - Marilena Cipollaro
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Second University of Naples, Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology, Second University of Naples, Naples, Italy
- Institute of Biomedicine and Bioresources (IBBR); CNR, Naples, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section, Second University of Naples, Naples, Italy
| |
Collapse
|
48
|
Abstract
"During the past decade, studies in animals and humans have suggested that cell therapy has positive effects for the treatment of heart failure. This clinical effect may be mediated by angiogenesis and reduction in fibrosis rather than by regeneration of myocytes. Increased microvasculature and decreased scar also likely lead to improved cardiac function in the failing heart. The effects of cell therapy are not limited to one type of cell or delivery technique. Well-designed, large-scale, randomized clinical trials with objective end points will help to fully realize the therapeutic potential of cell-based therapy for treating heart failure."
Collapse
Affiliation(s)
- Amit N Patel
- University of Utah School of Medicine, 30 North 1900 East 3c127 SOM, Salt Lake City, UT 84132, USA.
| | - Francisco Silva
- University of Utah School of Medicine, 30 North 1900 East 3c127 SOM, Salt Lake City, UT 84132, USA
| | - Amalia A Winters
- University of Utah School of Medicine, 30 North 1900 East 3c127 SOM, Salt Lake City, UT 84132, USA
| |
Collapse
|
49
|
Katsaros KM, Speidl WS, Demyanets S, Kastl SP, Krychtiuk KA, Wonnerth A, Zorn G, Tentzeris I, Farhan S, Maurer G, Wojta J, Huber K. G-CSF Predicts Cardiovascular Events in Patients with Stable Coronary Artery Disease. PLoS One 2015; 10:e0142532. [PMID: 26555480 PMCID: PMC4640870 DOI: 10.1371/journal.pone.0142532] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 08/30/2015] [Indexed: 12/19/2022] Open
Abstract
Granulocyte-colony-stimulating-factor (G-CSF) induces mobilization of progenitor cells but may also exert pro-inflammatory and pro-thrombotic effects. Treatment with recombinant G-CSF after acute myocardial infarction is currently under examination and has been associated with in-stent restenosis. However, it is not known whether plasma levels of endogenous G-CSF are also associated with an increased cardiovascular risk. Therefore we included 280 patients with angiographically proven stable coronary artery disease. G-CSF was measured by specific ELISA and patients were followed for a median of 30 months for the occurrence of major adverse cardiovascular events (MACE: death, myocardial infarction, re-hospitalization). Those with cardiac events during follow-up showed significant higher G-CSF levels (32.3 pg/mL IQR 21.4-40.5 pg/mL vs. 24.6 pg/mL IQR 16.4-34.9 pg/mL; p<0.05) at baseline. Patients with G-CSF plasma levels above the median had a 2-fold increased risk for MACE (p<0.05). This was independent from established cardiovascular risk factors. In addition, G-CSF above the median was a predictor of clinical in-stent restenosis after implantation of bare-metal stents (6.6% vs. 19.4%; p<0.05) but not of drug-eluting stents (7.7% vs. 7.6%; p = 0.98). This data suggests that endogenous plasma levels of G-CSF predict cardiovascular events independently from established cardiac risk factors and are associated with increased in-stent restenosis rates after implantation of bare metal stents.
Collapse
Affiliation(s)
- Katharina M. Katsaros
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - Walter S Speidl
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
| | - Svitlana Demyanets
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
| | - Stefan P. Kastl
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - Konstantin A. Krychtiuk
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - Anna Wonnerth
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
| | - Gerlinde Zorn
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
| | - Ioannis Tentzeris
- 3rd Medical Department, Cardiology and Intensive Care Medicine, Wilhelminenhospital, Vienna, Austria
| | - Serdar Farhan
- 3rd Medical Department, Cardiology and Intensive Care Medicine, Wilhelminenhospital, Vienna, Austria
| | - Gerald Maurer
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
- * E-mail:
| | - Kurt Huber
- 3rd Medical Department, Cardiology and Intensive Care Medicine, Wilhelminenhospital, Vienna, Austria
| |
Collapse
|
50
|
Nam TK, Park SW, Park YS, Kwon JT, Min BK, Hwang SN. Role of a Burr Hole and Calvarial Bone Marrow-Derived Stem Cells in the Ischemic Rat Brain: A Possible Mechanism for the Efficacy of Multiple Burr Hole Surgery in Moyamoya Disease. J Korean Neurosurg Soc 2015; 58:167-74. [PMID: 26539257 PMCID: PMC4630345 DOI: 10.3340/jkns.2015.58.3.167] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Revised: 03/18/2015] [Accepted: 06/11/2015] [Indexed: 01/06/2023] Open
Abstract
Objective This study investigates the role of a burr hole and calvarial bone marrow-derived stem cells (BMSCs) in a transient ischemic brain injury model in the rat and postulates a possible mechanism for the efficacy of multiple cranial burr hole (MCBH) surgery in moyamoya disease (MMD). Methods Twenty Sprague-Dawley rats (250 g, male) were divided into four groups : normal control group (n=5), burr hole group (n=5), ischemia group (n=5), and ischemia+burr hole group (n=5). Focal ischemia was induced by the transient middle cerebral artery occlusion (MCAO). At one week after the ischemic injury, a 2 mm-sized cranial burr hole with small cortical incision was made on the ipsilateral (left) parietal area. Bromodeoxyuridine (BrdU, 50 mg/kg) was injected intraperitoneally, 2 times a day for 6 days after the burr hole trephination. At one week after the burr hole trephination, brains were harvested. Immunohistochemical stainings for BrdU, CD34, VEGF, and Doublecortin and Nestin were done. Results In the ischemia+burr hole group, BrdU (+), CD34 (+), and Doublecortin (+) cells were found in the cortical incision site below the burr hole. A number of cells with Nestin (+) or VEGF (+) were found in the cerebral parenchyma around the cortical incision site. In the other groups, BrdU (+), CD34 (+), Doublecortin (+), and Nestin (+) cells were not detected in the corresponding area. These findings suggest that BrdU (+) and CD34 (+) cells are bone marrow-derived stem cells, which may be derived from the calvarial bone marrow through the burr hole. The existence of CD34 (+) and VEGF (+) cells indicates increased angiogenesis, while the existence of Doublecortin (+), Nestin (+) cells indicates increased neurogenesis. Conclusion Based on these findings, the BMSCs through burr holes seem to play an important role for the therapeutic effect of the MCBH surgery in MMD.
Collapse
Affiliation(s)
- Taek-Kyun Nam
- Department of Neurosurgery, Chung-Ang University College of Medicine, Seoul, Korea
| | - Seung-Won Park
- Department of Neurosurgery, Chung-Ang University College of Medicine, Seoul, Korea
| | - Yong-Sook Park
- Department of Neurosurgery, Chung-Ang University College of Medicine, Seoul, Korea
| | - Jeong-Taik Kwon
- Department of Neurosurgery, Chung-Ang University College of Medicine, Seoul, Korea
| | - Byung-Kook Min
- Department of Neurosurgery, Chung-Ang University College of Medicine, Seoul, Korea
| | - Sung-Nam Hwang
- Department of Neurosurgery, Chung-Ang University College of Medicine, Seoul, Korea
| |
Collapse
|