1
|
Gharib OA, Fahmy HA, Mohamed MA, Rashed LA. Potential impact of mesenchymal stem cells on nephrotoxicity induced by gamma irradiation and antiepileptic drugs cotherapy in rats. Cell Biochem Funct 2023; 41:1200-1208. [PMID: 37695119 DOI: 10.1002/cbf.3853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/12/2023]
Abstract
The goal of this study was to assess the influence of bone marrow-derived mesenchymal stem cells (BM-MSCs) on the nephrotoxicity induced by fractionated doses of gamma irradiation (Rad) and the cotherapy of levetiracetam and oxcarbazepine in male rats. Adult rats were randomly divided into four groups. Group I: Control, Group II: antiepileptic drugs (AEDs), Group III: AEDs +Rad and Group IV: AEDs + Rad + MSCs. Rats treated with AEDs and exposed to fractionated doses of γ-irradiation displayed a discernible increase in serum urea, creatinine, kidney injury marker, kidney malondialdehyde, transforming growth factor beta (TGF-β) and the relative expression of Smad3 along with a decrease in the relative expression of Smad7 and glutathione level. Alternatively, groups treated with BM-MSCs with AEDs and Rad showed a substantial modification in the majority of the evaluated parameters and looked to be successful in reducing the hazards of the combination therapy of AEDs and radiation. The reno-histopathological study supports the biochemical analysis. In conclusion, BM-MSCs exhibited therapeutic potential against nephrotoxicity induced by fractionated doses of γ-irradiation and AEDs. The outcome was brought about by the downregulation of the TGF-β/Smad pathway. BM-MSCs might be suggested as a valuable therapeutic strategy to overcome kidney injury induced by gamma irradiation during AEDs cotherapy.
Collapse
Affiliation(s)
- Ola A Gharib
- Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA), Nasr City, Cairo, Egypt
| | - Hanan A Fahmy
- Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA), Nasr City, Cairo, Egypt
| | - Marwa A Mohamed
- Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA), Nasr City, Cairo, Egypt
| | - Laila A Rashed
- Biochemistry and Molecular biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
2
|
Lopez-Lozano AP, Arevalo-Niño K, Gutierrez-Puente Y, Montiel-Hernandez JL, Urrutia-Baca VH, Del Angel-Mosqueda C, De la Garza-Ramos MA. SSEA-4 positive dental pulp stem cells from deciduous teeth and their induction to neural precursor cells. Head Face Med 2022; 18:9. [PMID: 35236383 PMCID: PMC8889676 DOI: 10.1186/s13005-022-00313-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 02/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Stage-specific embryonic antigen-4 (SSEA-4) is a marker for the identification of multipotent embryonic cells. It is also positive in neuroepithelial cells, precursor neural cells (NPC), and human dental pulp cells. The aim of this study was to evaluate the potential morphodifferentiation and histodifferentiation to NPC of SSEA-4 positive stem cells from human exfoliated deciduous teeth (SHED). METHODS A SHED population in culture, positive to SSEA-4, was obtained by magnetic cell separation. The cells were characterized by immunohistochemistry and flow cytometry. Subsequently, a neurosphere assay was performed in a medium supplemented with basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF); afterward, cells were neurodifferenciated with a neurobasal medium. Finally, indirect immunohistochemistry was performed to identify neuronal markers. RESULTS The morphological and histological changes in the SSEA-4 positive SHEDs were observed after induction with epidermal and fibroblast growth factors in neurobasal culture medium. At the end of induction, the markers Nestin, TuJ-1, and GFAP were identified. CONCLUSIONS The findings show that SSEA-4 positive SHEDs have a behavior similar to neuronal precursor cells. Our findings indicate that the dental pulp of deciduous teeth is a promising source for regeneration therapies associated with neurodegenerative diseases or peripheral nerve alterations.
Collapse
Affiliation(s)
- Ada Pricila Lopez-Lozano
- Facultad de Ciencias Biológicas, Instituto de Biotecnología, Universidad Autónoma de Nuevo León, Nuevo Leon, San Nicolas de los Garza, Mexico.,Unidad de Odontología Integral y Especialidades, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autonoma de Nuevo Leon, Nuevo Leon, Monterrey, Mexico
| | - Katiushka Arevalo-Niño
- Facultad de Ciencias Biológicas, Instituto de Biotecnología, Universidad Autónoma de Nuevo León, Nuevo Leon, San Nicolas de los Garza, Mexico
| | - Yolanda Gutierrez-Puente
- Facultad de Ciencias Biológicas, Instituto de Biotecnología, Universidad Autónoma de Nuevo León, Nuevo Leon, San Nicolas de los Garza, Mexico
| | - Jose Luis Montiel-Hernandez
- Facultad De Farmacia, Coordinacion De Posgrado, Universidad Autonoma del Estado de Morelos, Morelos, Cuernavaca, Mexico
| | - Victor Hugo Urrutia-Baca
- Unidad de Odontología Integral y Especialidades, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autonoma de Nuevo Leon, Nuevo Leon, Monterrey, Mexico
| | | | - Myriam Angelica De la Garza-Ramos
- Facultad de Ciencias Biológicas, Instituto de Biotecnología, Universidad Autónoma de Nuevo León, Nuevo Leon, San Nicolas de los Garza, Mexico. .,Unidad de Odontología Integral y Especialidades, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autonoma de Nuevo Leon, Nuevo Leon, Monterrey, Mexico. .,Facultad de Odontología, Universidad Autonoma de Nuevo Leon, Nuevo Leon, Monterrey, Mexico. .,Facultad de Odontología/CIDICS, Universidad Autonoma de Nuevo Leon Monterrey, San Nicolás de los Garza, Mexico.
| |
Collapse
|
3
|
Tang XL, Wysoczynski M, Gumpert AM, Li Y, Wu WJ, Li H, Stowers H, Bolli R. Effect of intravenous cell therapy in rats with old myocardial infarction. Mol Cell Biochem 2022; 477:431-444. [PMID: 34783963 PMCID: PMC8896398 DOI: 10.1007/s11010-021-04283-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 10/21/2021] [Indexed: 10/19/2022]
Abstract
Mounting evidence shows that cell therapy provides therapeutic benefits in experimental and clinical settings of chronic heart failure. However, direct cardiac delivery of cells via transendocardial injection is logistically complex, expensive, entails risks, and is not amenable to multiple dosing. Intravenous administration would be a more convenient and clinically applicable route for cell therapy. Thus, we determined whether intravenous infusion of three widely used cell types improves left ventricular (LV) function and structure and compared their efficacy. Rats with a 30-day-old myocardial infarction (MI) received intravenous infusion of vehicle (PBS) or 1 of 3 types of cells: bone marrow mesenchymal stromal cells (MSCs), cardiac mesenchymal cells (CMCs), and c-kit-positive cardiac cells (CPCs), at a dose of 12 × 106 cells. Rats were followed for 35 days after treatment to determine LV functional status by serial echocardiography and hemodynamic studies. Blood samples were collected for Hemavet analysis to determine inflammatory cell profile. LV ejection fraction (EF) dropped ≥ 20 points in all hearts at 30 days after MI and deteriorated further at 35-day follow-up in the vehicle-treated group. In contrast, deterioration of EF was halted in rats that received MSCs and attenuated in those that received CMCs or CPCs. None of the 3 types of cells significantly altered scar size, myocardial content of collagen or CD45-positive cells, or Hemavet profile. This study demonstrates that a single intravenous administration of 3 types of cells in rats with chronic ischemic cardiomyopathy is effective in attenuating the progressive deterioration in LV function. The extent of LV functional improvement was greatest with CPCs, intermediate with CMCs, and least with MSCs.
Collapse
Affiliation(s)
- Xian-Liang Tang
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Marcin Wysoczynski
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Anna M Gumpert
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Yan Li
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Wen-Jian Wu
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Hong Li
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Heather Stowers
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, 550 S Jackson Street, ACB Bldg, 3rd Floor, Louisville, KY, 40202, USA.
| |
Collapse
|
4
|
Costela Ruiz VJ, Melguizo Rodríguez L, Illescas Montes R, García Recio E, Arias Santiago S, Ruiz C, De Luna Bertos E. Human adipose tissue-derived mesenchymal stromal cells and their phagocytic capacity. J Cell Mol Med 2021; 26:178-185. [PMID: 34854223 PMCID: PMC8742185 DOI: 10.1111/jcmm.17070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/02/2021] [Accepted: 11/11/2021] [Indexed: 11/30/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have evidenced considerable therapeutic potential in numerous clinical fields, especially in tissue regeneration. The immunological characteristics of this cell population include the expression of Toll‐like receptors and mannose receptors, among others. The study objective was to determine whether MSCs have phagocytic capacity against different target particles. We isolated and characterized three human adipose tissue MSC (HAT‐MSC) lines from three patients and analysed their phagocytic capacity by flow cytometry, using fluorescent latex beads, and by transmission electron microscopy, using Escherichia coli, Staphylococcus aureus and Candida albicans as biological materials and latex beads as non‐biological material. The results demonstrate that HAT‐MSCs can phagocyte particles of different nature and size. The percentage of phagocytic cells ranged between 33.8% and 56.2% (mean of 44.37% ± 11.253) according to the cell line, and a high phagocytic index was observed. The high phagocytic capacity observed in MSCs, which have known regenerative potential, may offer an advance in the approach to certain local and systemic infections.
Collapse
Affiliation(s)
- Víctor J Costela Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Lucía Melguizo Rodríguez
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Rebeca Illescas Montes
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Enrique García Recio
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| | - Salvador Arias Santiago
- Biosanitary Research Institute, ibs Granada, Granada, Spain.,Surgical Medical Dermatology and Venereology Service, Department of Medicine, Virgen de las Nieves Hospital, Granada, Spain
| | - Concepción Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain.,Institute of Neuroscience, Centre for Medical Research (CIBM), Health Technology Park (PTS), University of Granada, Granada, Spain
| | - Elvira De Luna Bertos
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Biosanitary Research Institute, ibs Granada, Granada, Spain
| |
Collapse
|
5
|
Asker ME, Ali SI, Mohamed SH, Abdelaleem RMA, Younis NN. The efficacy of bone marrow-derived mesenchymal stem cells and/or erythropoietin in ameliorating kidney damage in gamma irradiated rats: Role of non-hematopoietic erythropoietin anti-apoptotic signaling. Life Sci 2021; 275:119388. [PMID: 33774028 DOI: 10.1016/j.lfs.2021.119388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 01/26/2021] [Accepted: 03/22/2021] [Indexed: 01/27/2023]
Abstract
Radiation-induced multiple organ injury, including γ-radiation nephropathy, is the most common. Even with dose fractionation strategy, residual late side effects are inevitable. Bone marrow-derived mesenchymal stem cells (BM-MSCs) transplantation and erythropoietin (EPO) have shown to be effective in treating chronic kidney disease and associated anemia. This study aimed to evaluate the effect of BM-MSCs and/or EPO in fractionated γ-irradiation induced kidney damage in rats. Adult male Wistar rats were randomized into 2 groups; normal and 8 Gy (fractionated dose of 2 Gy for 4 days) γ-irradiated rats. Animal from both groups were subdivided to receive the following treatments: BM-MSCs (1 × 106 cells/rat, i.v - once), EPO (100 IU/kg, i.p - every other day for 30 days) or their combined treatment (BM-MSCs and EPO). γ-Irradiated rats showed a noticeable elevation in serum urea and creatinine, kidney malondialdehyde (MDA) and caspase 3 activity. They also revealed significant drop in kidney glutathione (GSH) and Bcl2 protein contents. Conspicuously, they revealed down-regulation of renal EPO signaling (EPO, EPOR, pJAK2, pPI3K and pAkt). Conversely, groups treated with BM-MSCs and/or EPO revealed significant modulation in most tested parameters and appeared to be effective in minimizing the hazard effects of radiation. In conclusion, BM-MSCs and/or EPO exhibited therapeutic potentials against nephrotoxicity induced by fractionated dose of γ-irradiation. An effect mediated by antioxidant and non-hematopoietic EPO downstream anti-apoptotic signaling (PI3K/Akt) pathway. EPO potentiate the repair capabilities of BM-MSCs making this combined treatment a promising therapeutic strategy to overcome radiotherapy-induced kidney damage.
Collapse
Affiliation(s)
- Mervat E Asker
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Sousou I Ali
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Seham H Mohamed
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Atomic Energy Authority, PO Box 29, Nasr City, Cairo, Egypt
| | - Rasha M A Abdelaleem
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Atomic Energy Authority, PO Box 29, Nasr City, Cairo, Egypt
| | - Nahla N Younis
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt.
| |
Collapse
|
6
|
Hashemzadeh MR, Taghavizadeh Yazdi ME, Amiri MS, Mousavi SH. Stem cell therapy in the heart: Biomaterials as a key route. Tissue Cell 2021; 71:101504. [PMID: 33607524 DOI: 10.1016/j.tice.2021.101504] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases are one of the main concerns, nowadays causing a high rate of mortality in the world. The majority of conventional treatment protects the heart from failure progression. As a novel therapeutic way, Regenerative medicine in the heart includes cellular and noncellular approaches. Despite the irrefutable privileges of noncellular aspects such as administration of exosomes, utilizing of miRNAs, and growth factors, they cannot reverse necrotic or ischemic myocardium, hence recruiting of stem cells to help regenerative therapy in the heart seems indispensable. Stem cell lineages are varied and divided into two main groups namely pluripotent and adult stem cells. Not only has each of which own regenerative capacity, benefits, and drawbacks, but their turnover also close correlates with the target organ and/or tissue as well as the stage and level of failure. In addition to inefficient tissue integration due to the defects in delivering methods and poor retention of transplanted cells, the complexity of the heart and its movement also make more rigorous the repair process. Hence, utilizing biomaterials can make a key route to tackle such obstacles. In this review, we evaluate some natural products which can help stem cells in regenerative medicine of the cardiovascular system.
Collapse
Affiliation(s)
- Mohammad Reza Hashemzadeh
- Department of Stem Cells and Regenerative Medicine, Royesh Stem Cell Biotechnology Institute, Mashhad, Iran; Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | | | - Seyed Hadi Mousavi
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Vazir A, Fox K, Westaby J, Evans MJ, Westaby S. Can we remove scar and fibrosis from adult human myocardium? Eur Heart J 2020; 40:960-966. [PMID: 30203057 DOI: 10.1093/eurheartj/ehy503] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/09/2018] [Accepted: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
The pathological processes leading to heart failure are characterized by the formation of fibrosis and scar, yet the dynamics of scar production and removal are incompletely understood. Spontaneous disappearance of myocardial collagen is reported in infancy but doubted in adulthood where scar volume constitutes a better prognostic indicator than the conventional parameters of ventricular function. Whilst certain drugs are known to attenuate myocardial fibrosis evidence is emerging that stem cell therapy also has the potential to reduce scar size and improve myocardial viability. Both animal studies and clinical trials support the concept that, as in infancy, cellular processes can be triggered to remove collagen and regenerate injured myocardium. The molecular mechanisms likely involve anti-fibrotic cytokines growth factors and matrix-metalloproteinases. Autologous cardiac, bone-marrow and adipose tissue derived stem cells have each shown efficacy. Specific immune privileged mesenchymal stem cells and genetically modified immunomodulatory progenitor cells may in turn provide an allogenic source for the paracrine effects. Thus autologous and allogenic cells both have the potential through paracrine action to reduce scar volume, boost angiogenesis and improve ventricular morphology. The potential benefit of myocardial cell therapy for routine treatment of heart failure is an area that requires further study.
Collapse
Affiliation(s)
- Ali Vazir
- National Heart and Lung Institute, Imperial College London and ICMS, Royal Brompton Hospital, Dovehouse Street, London, UK
| | - Kim Fox
- National Heart and Lung Institute, Imperial College London and ICMS, Royal Brompton Hospital, Dovehouse Street, London, UK
| | - Joseph Westaby
- Department of Pathology, St George's University Hospital NHS Foundation Trust, Blackshaw Road, Tooting, London, UK
| | - Martin J Evans
- School of Biosciences, Cardiff University, The Sir Martin Evans Building, Museum Avenue, Cardiff, Wales, UK
| | - Stephen Westaby
- National Heart and Lung Institute, Imperial College London and ICMS, Royal Brompton Hospital, Dovehouse Street, London, UK.,Institute of Life Science, Swansea University, Singleton Park, Swansea, Wales, UK
| |
Collapse
|
8
|
Dao TTT, Bich Vu N, Phi LT, Le HTN, Phan NK, Ta VT, Van Pham P. Human adipose-derived mesenchymal stem cell could participate in angiogenesis in a mouse model of acute hindlimb ischemia. BIOMEDICAL RESEARCH AND THERAPY 2016. [DOI: 10.7603/s40730-016-0037-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
9
|
Faiella W, Atoui R. Therapeutic use of stem cells for cardiovascular disease. Clin Transl Med 2016; 5:34. [PMID: 27539581 PMCID: PMC4990528 DOI: 10.1186/s40169-016-0116-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 08/10/2016] [Indexed: 12/21/2022] Open
Abstract
Stem cell treatments are a desirable therapeutic option to regenerate myocardium and improve cardiac function after myocardial infarction. Several different types of cells have been explored, each with their own benefits and limitations. Induced pluripotent stem cells possess an embryonic-like state and therefore have a high proliferative capacity, but they also pose a risk of teratoma formation. Mesenchymal stem cells have been investigated from both bone marrow and adipose tissue. Their immunomodulatory characteristics may permit the use of allogeneic cells as universal donor cells in the future. Lastly, studies have consistently shown that cardiac stem cells are better able to express markers of cardiogenesis compared to other cell types, as well improve cardiac function. The ideal source of stem cells depends on multiple factors such as the ease of extraction/isolation, effectiveness of engraftment, ability to differentiate into cardiac lineages and effect on cardiac function. Although multiple studies highlight the benefits and limitations of each cell type and reinforce the successful potential use of these cells to regenerate damaged myocardium, more studies are needed to directly compare cells from various sources. It is interesting to note that research using stem cell therapies is also expanding to treat other cardiovascular diseases including non-ischemic cardiomyopathies.
Collapse
Affiliation(s)
- Whitney Faiella
- Division of Cardiac Surgery, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, P3E 5J1, Canada
| | - Rony Atoui
- Division of Cardiac Surgery, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, P3E 5J1, Canada.
| |
Collapse
|
10
|
Robinson AM, Miller S, Payne N, Boyd R, Sakkal S, Nurgali K. Neuroprotective Potential of Mesenchymal Stem Cell-Based Therapy in Acute Stages of TNBS-Induced Colitis in Guinea-Pigs. PLoS One 2015; 10:e0139023. [PMID: 26397368 PMCID: PMC4580595 DOI: 10.1371/journal.pone.0139023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/07/2015] [Indexed: 12/13/2022] Open
Abstract
Background & Aims The therapeutic benefits of mesenchymal stem cells (MSCs), such as homing ability, multipotent differentiation capacity and secretion of soluble bioactive factors which exert neuroprotective, anti-inflammatory and immunomodulatory properties, have been attributed to attenuation of autoimmune, inflammatory and neurodegenerative disorders. In this study, we aimed to determine the earliest time point at which locally administered MSC-based therapies avert enteric neuronal loss and damage associated with intestinal inflammation in the guinea-pig model of colitis. Methods At 3 hours after induction of colitis by 2,4,6-trinitrobenzene-sulfonate (TNBS), guinea-pigs received either human bone marrow-derived MSCs, conditioned medium (CM), or unconditioned medium by enema into the colon. Colon tissues were collected 6, 24 and 72 hours after administration of TNBS. Effects on body weight, gross morphological damage, immune cell infiltration and myenteric neurons were evaluated. RT-PCR, flow cytometry and antibody array kit were used to identify neurotrophic and neuroprotective factors released by MSCs. Results MSC and CM treatments prevented body weight loss, reduced infiltration of leukocytes into the colon wall and the myenteric plexus, facilitated repair of damaged tissue and nerve fibers, averted myenteric neuronal loss, as well as changes in neuronal subpopulations. The neuroprotective effects of MSC and CM treatments were observed as early as 24 hours after induction of inflammation even though the inflammatory reaction at the level of the myenteric ganglia had not completely subsided. Substantial number of neurotrophic and neuroprotective factors released by MSCs was identified in their secretome. Conclusion MSC-based therapies applied at the acute stages of TNBS-induced colitis start exerting their neuroprotective effects towards enteric neurons by 24 hours post treatment. The neuroprotective efficacy of MSC-based therapies can be exerted independently to their anti-inflammatory effects.
Collapse
Affiliation(s)
- Ainsley M. Robinson
- Centre for Chronic Diseases, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Sarah Miller
- Centre for Chronic Diseases, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Natalie Payne
- Department of Anatomy and Neuroscience, Monash University, Melbourne, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Richard Boyd
- Department of Anatomy and Neuroscience, Monash University, Melbourne, Australia
| | - Samy Sakkal
- Centre for Chronic Diseases, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Kulmira Nurgali
- Centre for Chronic Diseases, College of Health and Biomedicine, Victoria University, Melbourne, Australia
- * E-mail:
| |
Collapse
|
11
|
Lin G, Alwaal A, Zhang X, Wang J, Wang L, Li H, Wang G, Ning H, Lin CS, Xin Z, Lue TF. Presence of stem/progenitor cells in the rat penis. Stem Cells Dev 2015; 24:264-70. [PMID: 25162971 DOI: 10.1089/scd.2014.0360] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tissue resident stem cells are believed to exist in every organ, and their identification is commonly done using a combination of immunostaining for putative stem cell markers and label-retaining cell (LRC) strategy. In this study, we employed these approaches to identify potential stem cells in the penis. Newborn rats were intraperitoneally injected with thymidine analog, 5-ethynyl-2-deoxyuridine (EdU), and their penis was harvested at 7 h, 3 days, 1 week, and 4 weeks. It was processed for EdU stains and immunofluorescence staining for stem cell markers A2B5, PCNA, and c-kit. EdU-positive cells were counted for each time point and co-localized with each stem cell marker, then isolated and cultured in vitro followed by their characterization using flowcytometry and immunofluorescence. At 7 h post-EdU injection, 410 ± 105.3 penile corporal cells were labeled in each cross-section (∼28%). The number of EdU-positive cells at 3 days increased to 536 ± 115.6, while their percentage dropped to 25%. Progressively fewer EdU-positive cells were present in the sacrificed rat penis at longer time points (1 and 4 weeks). They were mainly distributed in the subtunic and perisinusoidal spaces, and defined as subtunic penile progenitor cells (STPCs) and perisinusoidal penile progenitor cells (PPCs). These cells expressed c-kit, A2B5, and PCNA. After culturing in vitro, only ∼0.324% corporal cells were EdU-labeled LRCs and expressed A2B5/PCNA. Therefore, labeling of penis cells by EdU occurred randomly, and label retaining was not associated with expression of c-kit, A2B5, or PCNA. The penile LRCs are mainly distributed within the subtunic and perisinusoidal space.
Collapse
Affiliation(s)
- Guiting Lin
- 1 Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California , San Francisco, San Francisco, California
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Behbahan IS, Keating A, Gale RP. Bone Marrow Therapies for Chronic Heart Disease. Stem Cells 2015; 33:3212-27. [PMID: 26086629 DOI: 10.1002/stem.2080] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 05/16/2015] [Indexed: 12/20/2022]
Abstract
Chronic heart failure is a leading cause of death. The demand for new therapies and the potential regenerative capacity of bone marrow-derived cells has led to numerous clinical trials. We critically discuss current knowledge of the biology and clinical application of bone marrow cells. It appears unlikely that bone marrow cells can develop into functional cardiomyocyte after infusion but may have favorable paracrine effects. Most, but not all, clinical trials report a modest short- but not long-term benefit of infusing bone marrow-derived cells. Effect size appears to correlate with stringency of study-design: the most stringent trials report the smallest effect-sizes. We conclude there may be short- but not substantial long-term benefit of infusing bone marrow-derived cells into persons with chronic heart failure and any benefit observed is unlikely to result from trans-differentiation of bone marrow-derived cells into functioning cardiomyocytes.
Collapse
Affiliation(s)
- Iman Saramipoor Behbahan
- Clinical Observer, Division of Hematology, Stanford MDS Center, Stanford University, Palo Alto, California, USA
| | - Armand Keating
- Division of Hematology, University of Toronto, Cell Therapy Program, Princess Margaret Hospital, Toronto, Canada
| | - Robert Peter Gale
- Section of Haematology, Division of Medicine, Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
13
|
Liang X, Ding Y, Zhang Y, Tse HF, Lian Q. Paracrine mechanisms of mesenchymal stem cell-based therapy: current status and perspectives. Cell Transplant 2015; 23:1045-59. [PMID: 23676629 DOI: 10.3727/096368913x667709] [Citation(s) in RCA: 670] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are one of a few stem cell types to be applied in clinical practice as therapeutic agents for immunomodulation and ischemic tissue repair. In addition to their multipotent differentiation potential, a strong paracrine capacity has been proposed as the principal mechanism that contributes to tissue repair. Apart from cytokine/chemokine secretion, MSCs also display a strong capacity for mitochondrial transfer and microvesicle (exosomes) secretion in response to injury with subsequent promotion of tissue regeneration. These unique properties of MSCs make them an invaluable cell type to repair damaged tissues/organs. Although MSCs offer great promise in the treatment of degenerative diseases and inflammatory disorders, there are still many challenges to overcome prior to their widespread clinical application. Particularly, their in-depth paracrine mechanisms remain a matter for debate and exploration. This review will highlight the discovery of the paracrine mechanism of MSCs, regulation of the paracrine biology of MSCs, important paracrine factors of MSCs in modulation of tissue repair, exosome and mitochondrial transfer for tissue repair, and the future perspective for MSC-based therapy.
Collapse
Affiliation(s)
- Xiaoting Liang
- Cardiology Division, Department of Medicine, University of Hong Kong, Hong Kong
| | | | | | | | | |
Collapse
|
14
|
Castells-Sala C, Martínez-Ramos C, Vallés-Lluch A, Monleón Pradas M, Semino C. in vitro development of bioimplants made up of elastomeric scaffolds with peptide gel filling seeded with human subcutaneous adipose tissue-derived progenitor cells. J Biomed Mater Res A 2015; 103:3419-30. [PMID: 25903327 DOI: 10.1002/jbm.a.35482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 03/31/2015] [Accepted: 04/13/2015] [Indexed: 12/17/2022]
Abstract
Myocardial tissue lacks the ability to regenerate itself significantly following a myocardial infarction. Thus, new strategies that could compensate this lack are of high interest. Cardiac tissue engineering (CTE) strategies are a relatively new approach that aims to compensate the tissue loss using combination of biomaterials, cells and bioactive molecules. The goal of the present study was to evaluate cell survival and growth, seeding capacity and cellular phenotype maintenance of subcutaneous adipose tissue-derived progenitor cells in a new synthetic biomaterial scaffold platform. Specifically, here we tested the effect of the RAD16-I peptide gel in microporous poly(ethyl acrylate) polymers using two-dimensional PEA films as controls. Results showed optimal cell adhesion efficiency and growth in the polymers coated with the self-assembling peptide RAD16-I. Importantly, subATDPCs seeded into microporous PEA scaffolds coated with RAD16-I maintained its phenotype and were able to migrate outwards the bioactive patch, hopefully toward the infarcted area once implanted. These data suggest that this bioimplant (scaffold/RAD16-I/cells) can be suitable for further in vivo implantation with the aim to improve the function of affected tissue after myocardial infarction.
Collapse
Affiliation(s)
- Cristina Castells-Sala
- Tissue Engineering Laboratory, Bioengineering Department, Institut Químic De Sarrià, Universitat Ramon Llull, Barcelona, Spain
| | - Cristina Martínez-Ramos
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica De Valencia, Cno. De Vera S/N, Valencia, 46022, Spain
| | - Ana Vallés-Lluch
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica De Valencia, Cno. De Vera S/N, Valencia, 46022, Spain
| | - Manuel Monleón Pradas
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica De Valencia, Cno. De Vera S/N, Valencia, 46022, Spain
| | - Carlos Semino
- Tissue Engineering Laboratory, Bioengineering Department, Institut Químic De Sarrià, Universitat Ramon Llull, Barcelona, Spain
| |
Collapse
|
15
|
Taghavi S, Sharp TE, Duran JM, Makarewich CA, Berretta RM, Starosta T, Kubo H, Barbe M, Houser SR. Autologous c-Kit+ Mesenchymal Stem Cell Injections Provide Superior Therapeutic Benefit as Compared to c-Kit+ Cardiac-Derived Stem Cells in a Feline Model of Isoproterenol-Induced Cardiomyopathy. Clin Transl Sci 2015; 8:425-31. [PMID: 25684108 DOI: 10.1111/cts.12251] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Cardiac- (CSC) and mesenchymal-derived (MSC) CD117+ isolated stem cells improve cardiac function after injury. However, no study has compared the therapeutic benefit of these cells when used autologously. METHODS MSCs and CSCs were isolated on day 0. Cardiomyopathy was induced (day 28) by infusion of L-isoproterenol (1,100 ug/kg/hour) from Alzet minipumps for 10 days. Bromodeoxyuridine (BrdU) was infused via minipumps (50 mg/mL) to identify proliferative cells during the injury phase. Following injury (day 38), autologous CSC (n = 7) and MSC (n = 4) were delivered by intracoronary injection. These animals were compared to those receiving sham injections by echocardiography, invasive hemodynamics, and immunohistochemistry. RESULTS Fractional shortening improved with CSC (26.9 ± 1.1% vs. 16.1 ± 0.2%, p = 0.01) and MSC (25.1 ± 0.2% vs. 12.1 ± 0.5%, p = 0.01) as compared to shams. MSC were superior to CSC in improving left ventricle end-diastolic (LVED) volume (37.7 ± 3.1% vs. 19.9 ± 9.4%, p = 0.03) and ejection fraction (27.7 ± 0.1% vs. 19.9 ± 0.4%, p = 0.02). LVED pressure was less in MSC (6.3 ± 1.3 mmHg) as compared to CSC (9.3 ± 0.7 mmHg) and sham (13.3 ± 0.7); p = 0.01. LV BrdU+ myocytes were higher in MSC (0.17 ± 0.03%) than CSC (0.09 ± 0.01%) and sham (0.06 ± 01%); p < 0.001. CONCLUSIONS Both CD117+ isolated CSC and MSC therapy improve cardiac function and attenuate pathological remodeling. However, MSC appear to confer additional benefit.
Collapse
Affiliation(s)
- Sharven Taghavi
- Temple University Hospital, Department of Surgery, Philadelphia, Pennsylvania, USA.,Temple University School of Medicine, Cardiovascular Research Center, Philadelphia, Pennsylvania, USA
| | - Thomas E Sharp
- Temple University School of Medicine, Cardiovascular Research Center, Philadelphia, Pennsylvania, USA
| | - Jason M Duran
- Temple University School of Medicine, Cardiovascular Research Center, Philadelphia, Pennsylvania, USA
| | - Catherine A Makarewich
- Temple University School of Medicine, Cardiovascular Research Center, Philadelphia, Pennsylvania, USA
| | - Remus M Berretta
- Temple University School of Medicine, Cardiovascular Research Center, Philadelphia, Pennsylvania, USA
| | - Tim Starosta
- Temple University School of Medicine, Cardiovascular Research Center, Philadelphia, Pennsylvania, USA
| | - Hajime Kubo
- Temple University School of Medicine, Cardiovascular Research Center, Philadelphia, Pennsylvania, USA
| | - Mary Barbe
- Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Steven R Houser
- Temple University School of Medicine, Cardiovascular Research Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
16
|
Porada CD, Rodman C, Ignacio G, Atala A, Almeida-Porada G. Hemophilia A: an ideal disease to correct in utero. Front Pharmacol 2014; 5:276. [PMID: 25566073 PMCID: PMC4263089 DOI: 10.3389/fphar.2014.00276] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/27/2014] [Indexed: 01/13/2023] Open
Abstract
Hemophilia A (HA) is the most frequent inheritable defect of the coagulation proteins. The current standard of care for patients with HA is prophylactic factor infusion, which is comprised of regular (2-3 times per week) intravenous infusions of recombinant or plasma-derived FVIII to maintain hemostasis. While this treatment has greatly increased the quality of life and lengthened the life expectancy for many HA patients, its high cost, the need for lifelong infusions, and the fact that it is unavailable to roughly 75% of the world's HA patients make this type of treatment far from ideal. In addition, this lifesaving therapy suffers from a high risk of treatment failure due to immune response to the infused FVIII. There is thus a need for novel treatments, such as those using stem cells and/or gene therapy, which have the potential to mediate long-term correction or permanent cure following a single intervention. In the present review, we discuss the clinical feasibility and unique advantages that an in utero approach to treating HA could offer, placing special emphasis on a new sheep model of HA we have developed and on the use of mesenchymal stromal cells (MSC) as cellular vehicles for delivering the FVIII gene.
Collapse
Affiliation(s)
| | | | | | | | - Graça Almeida-Porada
- Regenerative Medicine, Wake Forest Institute for Regenerative MedicineWinston-Salem, NC, USA
| |
Collapse
|
17
|
Zhao S, Wang Y, Gao C, Zhang J, Bao H, Wang Z, Gong P. Superparamagnetic iron oxide magnetic nanomaterial-labeled bone marrow mesenchymal stem cells for rat liver repair after hepatectomy. J Surg Res 2014; 191:290-301. [DOI: 10.1016/j.jss.2014.03.064] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 03/18/2014] [Accepted: 03/21/2014] [Indexed: 12/14/2022]
|
18
|
Saito Y, Shimada M, Utsunomiya T, Ikemoto T, Yamada S, Morine Y, Imura S, Mori H, Arakawa Y, Kanamoto M, Iwahashi S, Takasu C. Homing effect of adipose-derived stem cells to the injured liver: the shift of stromal cell-derived factor 1 expressions. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2014; 21:873-80. [PMID: 25131380 DOI: 10.1002/jhbp.147] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Whether systemically transplanted human adipose-derived stem cells (ADSCs) homed to the injured liver in nude mice under stress with subsequent hepatectomy (Hx) and ischemia-reperfusion (I/R) was investigated in the present study. The types of cells in the liver that were involved in the homing of ADSCs were clarified, with focus on the stromal-derived factor-1 (SDF-1)/C-X-C chemokine receptor type 4 (CXCR-4) axis. METHODS Adipose-derived stem cells were transplanted intravenously immediately after 70% Hx and I/R. ADSCs were traced by in vivo imaging for 24 h after transplantation and ADSCs were histologically detected in the liver. SDF-1 and CXCR-4 expressions in the liver were evaluated by real time RT-PCR. The immunohistochemical analysis of SDF-1 was also performed to identify SDF-1 expressing cells in the liver. RESULTS Adipose-derived stem cells were found in various organs immediately following transplantation and almost accumulated in remnant liver or spleen at 6 h after transplantation. ADSCs were also histologically revealed in the harvested liver. Hx and I/R injury significantly enhanced SDF-1 expressions regardless of ADSCs transplantation, and only ADSC transplantation increased CXCR-4 expressions. The predominant SDF-1 positive cells in the liver were equally identified in parenchymal and non-parenchymal cells at 6 h, but shifted to non-parenchymal cells at 24 h after transplantation. CONCLUSIONS Systemically transplanted ADSCs homed to the injured liver after transplantation, possibly based on the mechanisms of SDF-1/CXCR-4 axis. Therefore, systemic transplantation might be an effective and practical route for the transplantation of ADSCs.
Collapse
Affiliation(s)
- Yu Saito
- Department of Surgery, Institute of Health Biosciences, The University of Tokushima Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zheng SX, Weng YL, Zhou CQ, Wen ZZ, Huang H, Wu W, Wang JF, Wang T. Comparison of cardiac stem cells and mesenchymal stem cells transplantation on the cardiac electrophysiology in rats with myocardial infarction. Stem Cell Rev Rep 2014; 9:339-49. [PMID: 22544360 DOI: 10.1007/s12015-012-9367-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Whether transplanted cardiac stem cells (CSCs) and mesenchymal stem cells (MSCs) improved ventricular fibrillation threshold (VFT) similarly is still unclear. We sought to compare the effects of the CSC and MSC transplantation on the electrophysiological characteristics and VFT in rats with myocardial infarction (MI). METHODS MI was induced in 30 male Sprague-Dawley rats. Two weeks later, animals were randomized to receive 5 × 10(6) CSCs labeled with PKH26 in PBS or 5 × 10(6) MSCs labeled with PKH26 in phosphate buffer solution(PBS) or PBS alone injection into the infarcted anterior ventricular free wall. Six weeks after the injection, electrophysiological characteristics and VFT were measured. Labeled CSCs and MSCs were observed in 5 μm cryostat sections from each heart. RESULTS Malignant ventricular arrhythmias were significantly (P = 0.0055) less inducible in the CSC group than the MSC group. The VFTs were improved in the CSC group compared with the MSC group. Labeled CSCs and MSCs were identified in the infarct zone and infarct marginal zone. Labeled CSCs expressed Connexin-43, von Willebrand factor, α-smooth muscle actin and α-sarcomeric actin,while the Labeled MSCs expressed von Willebrand factor, α-smooth muscle actin and α-sarcomeric actin in vivo. CONCLUSIONS After 6 weeks of cell transplantation, CSCs are superior to MSCs in modulating the electrophysiological abnormality and improving the VFT in rats with MI. CSCs and MSCs express markers that suggest muscle, endothelium and vascular smooth muscle phenotypes in vivo, but MSCs rarely express Connexin-43.
Collapse
Affiliation(s)
- Shao-Xin Zheng
- Cardiovascular Medicine, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, 107 Yanjiang Xi Road, Guangzhou, 510120, China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Oliveira MS, Carvalho JL, Campos ACDA, Gomes DA, de Goes AM, Melo MM. Doxorubicin has in vivo toxicological effects on ex vivo cultured mesenchymal stem cells. Toxicol Lett 2013; 224:380-6. [PMID: 24291741 DOI: 10.1016/j.toxlet.2013.11.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 11/19/2013] [Accepted: 11/20/2013] [Indexed: 12/13/2022]
Abstract
Doxorubicin (dox) is an effective chemotherapeutic agent that leads to cardiotoxicity. An alternative treatment for dox-cardiotoxicity is autologous mesenchymal stem cells (MSCs) transplantation. It remains unclear if dox has deleterious effects on MSCs from subjects under chemotherapy, therefore this study aimed to evaluate dox in vivo toxicological effects on ex vivo cultured MSCs, inferring whether autologous transplantation may be an alternative treatment in patients who are exposed to the drug. Wistar rats received either dox or saline. Following treatments, animals were sacrificed and bone marrow MSCs were isolated, characterized for cell surface markers and assessed according to their viability, alkaline phosphatase production, and proliferation kinetics. Moreover, MSCs were primed to cardiac differentiation and troponin T and connexin 43 expressions were evaluated. Compared to control, undifferentiated MSCs from dox group kept the pattern for surface marker and had similar viability results. In contrast, they showed lower alkaline phosphatase production, proliferation rate, and connexin 43 expression. Primed MSCs from dox group showed lower troponin T levels. It was demonstrated a toxic effect of dox in host MSCs. This result renders the possibility of autologous MSCs transplantation to treat dox-cardiotoxicity, which could be a non-suitable option for subjects receiving such antineoplastic agent.
Collapse
Affiliation(s)
- Maira Souza Oliveira
- College of Veterinary Medicine, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Caixa Postal 567, 30123-970 Belo Horizonte, MG, Brazil.
| | - Juliana Lott Carvalho
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Caixa Postal 567, 30123-970 Belo Horizonte, MG, Brazil
| | - Ana Carolina De Angelis Campos
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Caixa Postal 567, 30123-970 Belo Horizonte, MG, Brazil
| | - Dawidson Assis Gomes
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Caixa Postal 567, 30123-970 Belo Horizonte, MG, Brazil
| | - Alfredo Miranda de Goes
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Caixa Postal 567, 30123-970 Belo Horizonte, MG, Brazil
| | - Marília Martins Melo
- College of Veterinary Medicine, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Caixa Postal 567, 30123-970 Belo Horizonte, MG, Brazil
| |
Collapse
|
21
|
Caspases and p38 MAPK regulate endothelial cell adhesiveness for mesenchymal stem cells. PLoS One 2013; 8:e73929. [PMID: 24069252 PMCID: PMC3771880 DOI: 10.1371/journal.pone.0073929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 07/26/2013] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells natively circulating or delivered into the blood stream home to sites of injury. The mechanism of mesenchymal stem cell homing to sites of injury is poorly understood. We have shown that the development of apoptosis in endothelial cells stimulates endothelial cell adhesiveness for mesenchymal stem cells. Adhesion of mesenchymal stem cells to apoptotic endothelial cells depends on the activation of endothelial caspases and p38 MAPK. Activation of p38 MAPK in endothelial cells has a primary effect while the activation of caspases potentiates the mesenchymal stem cell adhesion. Overall, our study of the mesenchymal stem cell interaction with endothelial cells indicates that mesenchymal stem cells recognize and specifically adhere to distressed/apoptotic endothelial cells.
Collapse
|
22
|
Kholodenko IV, Konieva AA, Kholodenko RV, Yarygin KN. Molecular mechanisms of migration and homing of intravenously transplanted mesenchymal stem cells. ACTA ACUST UNITED AC 2013. [DOI: 10.7243/2050-1218-2-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
23
|
Porada CD, Almeida-Porada G. Treatment of Hemophilia A in Utero and Postnatally using Sheep as a Model for Cell and Gene Delivery. ACTA ACUST UNITED AC 2013; S1. [PMID: 23264887 DOI: 10.4172/2157-7412.s1-011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hemophilia A represents the most common inheritable deficiency of the coagulation proteins. Current state-of- the-art treatment consists of frequent prophylactic infusions of plasma-derived or recombinant FVIII protein to maintain hemostasis, and has greatly increased life expectancy and quality of life for many hemophilia A patients. This treatment approach is, however, far from ideal, due to the need for lifelong intravenous infusions, the high treatment cost, and the fact that it is unavailable to a large percentage of the world's hemophiliacs. There is thus a need for novel treatments that can promise long-term or permanent correction. In contrast to existing protein based therapeutics, gene therapy offers to provide a permanent cure following few, or even a single, treatment. In the present paper, we review ongoing work towards this end, focusing on studies we have performed in a large animal model. Some of the key topics covered in this review include the unique opportunities sheep offer as a model system, the re-establishment and clinical and molecular characterization of a line of sheep with severe hemophilia A, the advantages and feasibility of treating a disease like hemophilia A in utero, and the use of Mesenchymal Stem Cells (MSC) as cellular delivery vehicles for the FVIII gene. The review finishes with a brief discussion of our recent success correcting ovine hemophilia A with a postnatal transplant with gene-modified MSC, and the limitations of this approach that remain to be overcome.
Collapse
|
24
|
Effects of bone marrow mesenchymal stem cells in a rat model of myocardial infarction. Resuscitation 2012; 83:1391-6. [DOI: 10.1016/j.resuscitation.2012.02.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 02/15/2012] [Accepted: 02/21/2012] [Indexed: 01/14/2023]
|
25
|
Haneef K, Lila N, Benadda S, Legrand F, Carpentier A, Chachques JC. Development of bioartificial myocardium by electrostimulation of 3D collagen scaffolds seeded with stem cells. Heart Int 2012. [PMID: 23185681 PMCID: PMC3504306 DOI: 10.4081/hi.2012.e14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Electrostimulation (ES) can be defined as a safe physical method to induce stem cell differentiation. The aim of this study is to evaluate the effectiveness of ES on bone marrow mesenchymal stem cells (BMSCs) seeded in collagen scaffolds in terms of proliferation and differentiation into cardiomyocytes. BMSCs were isolated from Wistar rats and seeded into 3D collagen type 1 templates measuring 25 × 25 × 6 mm. Bipolar in vitro ES was performed during 21 days. Electrical impedance and cell proliferation were measured. Expression of cardiac markers was assessed by immunocytochemistry. Viscoelasticity of collagen matrix was evaluated. Electrical impedance assessments showed a low resistance of 234±41 Ohms which indicates good electrical conductivity of collagen matrix. Cell proliferation at 570 nm as significantly increased in ES groups after seven day (ES 0.129±0.03 vs non-stimulated control matrix 0.06±0.01, P=0.002) and after 21 days, (ES 0.22±0.04 vs control 0.13±0.01, P=0.01). Immunocytoche mistry of BMSCs after 21 days ES showed positive staining of cardiac markers, troponin I, connexin 43, sarcomeric alpha-actinin, slow myosin, fast myosin and desmin. Staining for BMSCs marker CD29 after 21 days was negative. Electrostimulation of cell-seeded collagen matrix changed stem cell morphology and biochemical characteristics, increasing the expression of cardiac markers. Thus, MSC-derived differentiated cells by electrostimulation grafted in biological scaffolds might result in a convenient tissue engineering source for myocardial diseases.
Collapse
Affiliation(s)
- Kanwal Haneef
- University Paris Descartes, Pompidou Hospital, Laboratory of Biosurgical Research (LRB), Paris
| | | | | | | | | | | |
Collapse
|
26
|
Wei F, Wang TZ, Zhang J, Yuan ZY, Tian HY, Ni YJ, Zhuo XZ, Han K, Liu Y, Lu Q, Bai HY, Ma AQ. Mesenchymal stem cells neither fully acquire the electrophysiological properties of mature cardiomyocytes nor promote ventricular arrhythmias in infarcted rats. Basic Res Cardiol 2012; 107:274. [PMID: 22744762 DOI: 10.1007/s00395-012-0274-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 04/30/2012] [Accepted: 05/25/2012] [Indexed: 12/28/2022]
Abstract
Electrophysiological properties of implanted mesenchymal stem cells (MSCs) in infarcted hearts remain unclear, and their proarrhythmic effect is still controversial. The intent of this study was to investigate electrophysiological properties and proarrhythmic effects of MSCs in infarcted hearts. Rats were randomly divided into a myocardial infarction (MI) group, a MI-DMEM group (received DMEM medium injection) and MI-MSCs group (received MSCs injection). Survival analysis showed that the majority of engrafted MSCs died at day 9 after transplantation. Engrafted MSCs expressed cardiac markers (MYH, cTnI, Cx43), cardiac ion channel genes (Kv1.4, Kv4.2 and Kir2.1) and potassium currents (I (to), I (K1) and I (KDR)), but did not express Nav1.5, Cav1.2, Na(+) current and Ca(2+) current during their survival. When induced by Ca(2+), implanted MSCs exhibited no contraction ability after being isolated from the heart. Following 8-week electrocardiography monitoring, the cumulative occurrence of ventricular arrhythmias (VAs) was not different among the three groups. However, the prolonged QRS duration in infarcted rats without VAs was significantly decreased in the MI-MSCs group compared with the other two groups. The inducibility of VAs in the MI-MSCs group was much lower than that in the MI and MI-DMEM groups (41.20 vs. 86.67 % and 92.86 %; P < 0.0125). The ventricular effective refractory period in MI-MSCs group was prolonged in comparison with that in the MI and MI-DMEM groups (56.0 ± 8.8 vs. 47.7 ± 8.8 ms and 45.7 ± 6.2 ms; P < 0.01). These results demonstrate that MSCs do not acquire the electrophysiological properties of mature cardiomyocytes during the survival period in the infarcted hearts. However, they can alleviate the electrical vulnerability and do not promote ventricular arrhythmias.
Collapse
Affiliation(s)
- Feng Wei
- Department of Cardiovascular Medicine, First Affiliated Hospital of the Xi'an Jiaotong University School of Medicine, Shaanxi, 710061, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Journey of mesenchymal stem cells for homing: strategies to enhance efficacy and safety of stem cell therapy. Stem Cells Int 2012; 2012:342968. [PMID: 22754575 PMCID: PMC3382267 DOI: 10.1155/2012/342968] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/06/2012] [Accepted: 04/17/2012] [Indexed: 12/11/2022] Open
Abstract
Human mesenchymal stem cells (MSCs) communicate with other cells in the human body and appear to "home" to areas of injury in response to signals of cellular damage, known as homing signals. This review of the state of current research on homing of MSCs suggests that favorable cellular conditions and the in vivo environment facilitate and are required for the migration of MSCs to the site of insult or injury in vivo. We review the current understanding of MSC migration and discuss strategies for enhancing both the environmental and cellular conditions that give rise to effective homing of MSCs. This may allow MSCs to quickly find and migrate to injured tissues, where they may best exert clinical benefits resulting from improved homing and the presence of increased numbers of MSCs.
Collapse
|
28
|
Singh JA. Stem cells and other innovative intra-articular therapies for osteoarthritis: what does the future hold? BMC Med 2012; 10:44. [PMID: 22551396 PMCID: PMC3364907 DOI: 10.1186/1741-7015-10-44] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 05/02/2012] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA), the most common type of arthritis in the world, is associated with suffering due to pain, productivity loss, decreased mobility and quality of life. Systemic therapies available for OA are mostly symptom modifying and have potential gastrointestinal, renal, hepatic, and cardiac side effects. BMC Musculoskeletal Disorders recently published a study showing evidence of reparative effects demonstrated by homing of intra-articularly injected autologous bone marrow stem cells in damaged cartilage in an animal model of OA, along with clinical and radiographic benefit. This finding adds to the growing literature showing the potential benefit of intra-articular (IA) bone marrow stem cells. Other emerging potential IA therapies include IL-1 receptor antagonists, conditioned autologous serum, botulinum toxin, and bone morphogenetic protein-7. For each of these therapies, trial data in humans have been published, but more studies are needed to establish that they are safe and effective. Several additional promising new OA treatments are on the horizon, but challenges remain to finding safe and effective local and systemic therapies for OA.Please see related article: http://www.biomedcentral.com/1471-2474/12/259.
Collapse
Affiliation(s)
- Jasvinder A Singh
- Medicine Service, Birmingham VA Medical Center and Department of Medicine, University of Alabama, Faculty Office Tower 805B, 510 20th Street S, Birmingham, AL 35294, USA.
| |
Collapse
|
29
|
Wang J, Cui W, Ye J, Ji S, Zhao X, Zhan L, Feng J, Zhang Z, Zhao Y. A cellular delivery system fabricated with autologous BMSCs and collagen scaffold enhances angiogenesis and perfusion in ischemic hind limb. J Biomed Mater Res A 2012; 100:1438-47. [PMID: 22378701 DOI: 10.1002/jbm.a.34081] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 12/30/2011] [Accepted: 01/17/2012] [Indexed: 01/07/2023]
Abstract
Although therapeutic cellular angiogenesis is effective for chronic ischemia, the optimal mode of cellular administration is still under exploration. This study aimed to develop a cellular delivery system to enhance the perfusion and angiogenesis in the ischemic hind limb. Collagen scaffold (CS) was prepared, and for morphology and toxicity analysis, bone marrow-derived mesenchymal stem cells (BMSCs) were isolated, expanded, filtrated, and seeded onto CS to construct BMSCs-CS. The ischemic hind limbs of rabbit models were implanted with autologous BMSCs-CS, CS, and autologous BMSCs; the untreated ischemic or normal animals were considered as the ischemic or normal control groups. Oxygen saturation parameters were regularly measured to determine the perfusion in the extremities. Histological examinations with hematoxylin and eosin immunostaining against von Willebrand factor and smooth muscle (SM) α-actin were performed for capillary and mature vessel evaluation. CS was a multiporous structure without cytotoxicity. At several intervals, the oxygen saturation ratio (OSR) in normal control was the highest. The OSRs in BMSCs-CS and CS were higher than that in BMSCs and ischemic control (p < 0.05); the OSR in BMSCs-CS group was higher than that in CS at 6 and 8 weeks (p < 0.05). The capillaries in BMSCs-CS and CS were higher than that in CS, BMSCs, and the ischemic or normal control (p < 0.05). The mature vessels in BMSCs-CS were higher than that in CS, BMSCs, and the ischemic or normal control (p < 0.05). The autologous cellular delivery system proved to be an effective approach for improving higher ischemic hind limb perfusion and angiogenesis as opposed to cellular therapy alone.
Collapse
Affiliation(s)
- Jinling Wang
- Department of Emergency, Zhongshan Hospital, Xiamen University, Xiamen, China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
|
31
|
Zhang H, Albersen M, Jin X, Lin G. Stem cells: novel players in the treatment of erectile dysfunction. Asian J Androl 2012; 14:145-55. [PMID: 22002437 PMCID: PMC3735142 DOI: 10.1038/aja.2011.79] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 06/30/2011] [Accepted: 07/19/2011] [Indexed: 01/09/2023] Open
Abstract
Stem cells are defined by their capacity for both self-renewal and directed differentiation; thus, they represent great promise for regenerative medicine. Historically, stem cells have been categorized as either embryonic stem cells (ESCs) or adult stem cells (ASCs). It was previously believed that only ESCs hold the ability to differentiate into any cell type, whereas ASCs have the capacity to give rise only to cells of a given germ layer. More recently, however, numerous studies demonstrated the ability of ASCs to differentiate into cell types beyond their tissue origin. The aim of this review was to summarize contemporary evidence regarding stem cell availability, differentiation, and more specifically, the potential of these cells in the diagnosis and treatment of erectile dysfunction (ED) in both animal models and human research. We performed a search on PubMed for articles related to definition, localisation and circulation of stem cells as well as the application of stem cells in both diagnosis and treatment of ED. Strong evidence supports the concept that stem cell therapy is potentially the next therapeutic approach for ED. To date, a large spectrum of stem cells, including bone marrow mesenchymal stem cells, adipose tissue-derived stem cells and muscle-derived stem cells, have been investigated for neural, vascular, endothelial or smooth muscle regeneration in animal models for ED. In addition, several subtypes of ASCs are localized in the penis, and circulating endogenous stem cells can be employed to predict the outcome of ED and ED-related cardiovascular diseases.
Collapse
Affiliation(s)
- Haiyang Zhang
- Minimally Invasive Urology Center, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | | | | | | |
Collapse
|
32
|
Cao X, Deng W, Wei Y, Su W, Yang Y, Wei Y, Yu J, Xu X. Encapsulation of plasmid DNA in calcium phosphate nanoparticles: stem cell uptake and gene transfer efficiency. Int J Nanomedicine 2011; 6:3335-49. [PMID: 22229000 PMCID: PMC3252680 DOI: 10.2147/ijn.s27370] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background The purpose of this study was to develop calcium phosphate nanocomposite particles encapsulating plasmid DNA (CP-pDNA) nanoparticles as a nonviral vector for gene delivery. Methods CP-pDNA nanoparticles employing plasmid transforming growth factor beta 1 (TGF-β1) were prepared and characterized. The transfection efficiency and cell viability of the CP-pDNA nanoparticles were evaluated in mesenchymal stem cells, which were identified by immunofluorescence staining. Cytotoxicity of plasmid TGF-β1 and calcium phosphate to mesenchymal stem cells were evaluated by MTT assay. Results The integrity of TGF-β1 encapsulated in the CP-pDNA nanoparticles was maintained. The well dispersed CP-pDNA nanoparticles exhibited an ultralow particle size (20–50 nm) and significantly lower cytotoxicity than Lipofectamine™ 2000. Immunofluorescence staining revealed that the cultured cells in this study were probably mesenchymal stem cells. The cellular uptake and transfection efficiency of the CP-pDNA nanoparticles into the mesenchymal stem cells were higher than that of needle-like calcium phosphate nanoparticles and a standard calcium phosphate transfection kit. Furthermore, live cell imaging and confocal laser microscopy vividly showed the transportation process of the CP-pDNA nanoparticles in mesenchymal stem cells. The results of a cytotoxicity assay found that both plasmid TGF-β1 and calcium phosphate were not toxic to mesenchymal stem cells. Conclusion CP-pDNA nanoparticles can be developed into an effective alternative as a nonviral gene delivery system that is highly efficient and has low cytotoxicity.
Collapse
Affiliation(s)
- Xia Cao
- Department of Pharmaceutics, School of Pharmacy, and Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Jingkou District, Zhenjiang, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Wen Z, Zheng S, Zhou C, Wang J, Wang T. Repair mechanisms of bone marrow mesenchymal stem cells in myocardial infarction. J Cell Mol Med 2011; 15:1032-43. [PMID: 21199333 PMCID: PMC3822616 DOI: 10.1111/j.1582-4934.2010.01255.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The prognosis of patients with myocardial infarction (MI) and resultant chronic heart failure remains extremely poor despite advances in optimal medical therapy and interventional procedures. Animal experiments and clinical trials using adult stem cell therapy following MI have shown a global improvement of myocardial function. Bone marrow-derived mesenchymal stem cells (MSCs) hold promise for cardiac repair following MI, due to their multilineage, self-renewal and proliferation potential. In addition, MSCs can be easily isolated, expanded in culture, and have immunoprivileged properties to the host tissue. Experimental studies and clinical trials have revealed that MSCs not only differentiate into cardiomyocytes and vascular cells, but also secrete amounts of growth factors and cytokines which may mediate endogenous regeneration via activation of resident cardiac stem cells and other stem cells, as well as induce neovascularization, anti-inflammation, anti-apoptosis, anti-remodelling and cardiac contractility in a paracrine manner. It has also been postulated that the anti-arrhythmic and cardiac nerve sprouting potential of MSCs may contribute to their beneficial effects in cardiac repair. Most molecular and cellular mechanisms involved in the MSC-based therapy after MI are still unclear at present. This article reviews the potential repair mechanisms of MSCs in the setting of MI.
Collapse
Affiliation(s)
- Zhuzhi Wen
- The Sun Yat-sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | |
Collapse
|
34
|
Mokbel AN, El Tookhy OS, Shamaa AA, Rashed LA, Sabry D, El Sayed AM. Homing and reparative effect of intra-articular injection of autologus mesenchymal stem cells in osteoarthritic animal model. BMC Musculoskelet Disord 2011; 12:259. [PMID: 22085445 PMCID: PMC3232438 DOI: 10.1186/1471-2474-12-259] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Accepted: 11/15/2011] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND This work aimed to study the homing evidence and the reparative effect of mesenchymal stem cells (MSCs) in the healing process of induced osteoarthritis in experimental animal model (donkeys). METHODS Twenty-seven donkeys were equally divided into 3 groups based on the observation period after induction of arthritis (3, 6 and 9 weeks) to achieve different degrees of osteoarthritis. Each group was subdivided into three subgroups of three animals each based on the follow-up period (1, 2 and 6 months) after treatment. The induction was done through intra-articular (IA) injection of 2 ml of Amphotericin-B in both carpal joints. MSCs were harvested in a separate procedure, labeled with green fluorescent protein (GFP) using monster GFP vector and suspended in hyaluronic acid for IA injection. Treatment approaches consisted of cell-treatment using MSCs suspended in 3 ml of hyaluronic acid (HA) for the right carpal joint; and using the same amount of (HA) but without MSCs for the left contralateral carpal joint to serve as a control. Animals were assessed clinically and radiologically before and after treatment. Synovial fluid was also evaluated. Histopathologically; articular cartilage structural changes, reduction of articular cartilage matrix staining, osteophyte formation, and subchondral bone plate thickening were graded. Data was summarized using median and percentile for scores of histopathologic grading. Comparison between groups was done using non-parametric Mann Whitney test. RESULTS The reparative effect of MSCs was significant both clinically and radiologically in all treated groups (P < 0.05) compared to the control groups. Fluorescence microscopy of sections of the cell-treated joints of all animals indicated that the GFP-transduced injected cells have participated effectively in the reparative process of the damaged articular surface and have integrated within the existing articular cartilage. The cells were associated with the surface of the cartilage and, were also detected in the interior. CONCLUSIONS Homing was confirmed by the incorporation of injected GFP-labeled MSCs within the repaired newly formed cartilage. Significant recovery proves that the use of IA injection of autologous MSCs is a viable and a practical option for treating different degrees of osteoarthritis.
Collapse
Affiliation(s)
- Abir N Mokbel
- Department of rheumatology and rehabilitation, Faculty of Medicine. Cairo University, Egypt
| | - Omar S El Tookhy
- Department of surgery, anesthesiology and radiology, Faculty of Veterinary Medicine. Cairo University, Egypt
| | - Ashraf A Shamaa
- Department of surgery, anesthesiology and radiology, Faculty of Veterinary Medicine. Cairo University, Egypt
| | - Laila A Rashed
- Department of medical biochemistry and molecular biology, Faculty of Medicine. Cairo University, Egypt
| | - Dina Sabry
- Department of medical biochemistry and molecular biology, Faculty of Medicine. Cairo University, Egypt
| | - Abeer M El Sayed
- Department of pathology, National Cancer Institute. Cairo University, Egypt
| |
Collapse
|
35
|
Simpson DL, Boyd NL, Kaushal S, Stice SL, Dudley SC. Use of human embryonic stem cell derived-mesenchymal cells for cardiac repair. Biotechnol Bioeng 2011; 109:274-83. [PMID: 21837664 DOI: 10.1002/bit.23301] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 06/03/2011] [Accepted: 08/03/2011] [Indexed: 01/09/2023]
Abstract
Human mesenchymal stem cells (hMSC) have proven beneficial in the repair and preservation of infarcted myocardium. Unfortunately, MSCs represent a small portion of the bone marrow and require ex vivo expansion. To further advance the clinical usefulness of cellular cardiomyoplasty, derivation of "MSC-like" cells that can be made available "off-the-shelf" are desirable. Recently, human embryonic stem cell-derived mesenchymal cells (hESC-MC) were described. We investigated the efficacy of hESC-MC for cardiac repair after myocardial infarction (MI) compared to hMSC. Because of increased efficacy of cell delivery, cells were embedded into collagen patches and delivered to infarcted myocardium. Culture of hMSC and hESC-MCs in collagen patches did not induce differentiation or significant loss in viability. Transplantation of hMSC and hES-MC patches onto infarcted myocardium of athymic nude rats prevented adverse changes in infarct wall thickness and fractional area change compared to a non-viable patch control. Hemodynamic assessment showed that hMSCs and hES-MC patch application improved end diastolic pressure equivalently. There were no changes in systolic function. hES-MC and hMSC construct application enhanced neovessel formation compared to a non-viable control, and each cell type had similar efficacy in stimulating endothelial cell growth in vitro. In summary, the use of hES-MC provides similar efficacy for cellular cardiomyoplasty as compared to hMSC and may be considered a suitable alternative for cell therapy.
Collapse
Affiliation(s)
- David L Simpson
- University of Illinois at Chicago/Jesse Brown VA Medical Center, 840 S. Wood Street, MC715, Chicago, Illinois 60612, USA
| | | | | | | | | |
Collapse
|
36
|
Tong Y, Xu W, Han H, Chen Y, Yang J, Qiao H, Hong D, Wu Y, Zhou C. Tanshinone IIA increases recruitment of bone marrow mesenchymal stem cells to infarct region via up-regulating stromal cell-derived factor-1/CXC chemokine receptor 4 axis in a myocardial ischemia model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2011; 18:443-450. [PMID: 21146968 DOI: 10.1016/j.phymed.2010.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2010] [Revised: 09/07/2010] [Accepted: 10/12/2010] [Indexed: 05/30/2023]
Abstract
Systemic administration with bone marrow mesenchymal stem cells (BMSCs) is a promising approach to cure myocardial ischemia (MI), while the efficacy of cell transplantation is limited by the low engraftment of BMSCs. Tanshinone IIA (Tan IIA) has been reported many times for the treatment of MI. Therefore, the present study was performed to investigate whether Tan IIA could increase the migration of BMSCs to ischemic region and its potential mechanisms. In our study, we found that combination treatment with Tan IIA and BMSCs significantly alleviated the infarct size when compared with control group (31.46 ± 3.00% vs. 46.95 ± 6.51%, p<0.05). Results of real-time PCR showed that Tanshinone IIA (Tan IIA) did increase the migration of BMSCs to ischemic region in vivo, which was correlated with cardiac function recovery after MI. Furthermore, 2 μM Tan IIA could enhance the migration capability of BMSCs in vitro (3.69-fold of control), and this enhancement could be blocked by AMD3100 (a CXC chemokine receptor 4 blocker). CXCR4, together with its specific receptor, stromal cell-derived factor-1 (SDF-1) plays a critical role in the stem cell recruitment. Our experiment indicated that Tan IIA could promote SDF-1α expression in the infarct area and enhance the CXCR4 expression of BMSCs in vitro. Therefore, we postulated that Tan IIA could increase the BMSCs migration via up-regulating SDF1/CXCR4 axis.
Collapse
Affiliation(s)
- Yinghui Tong
- College of Pharmaceutical Sciences, Zhejiang University, No. 358 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Deng W, Chen QW, Li XS, Liu H, Niu SQ, Zhou Y, Li GQ, Ke DZ, Mo XG. Bone marrow mesenchymal stromal cells with support of bispecific antibody and ultrasound-mediated microbubbles prevent myocardial fibrosis via the signal transducer and activators of transcription signaling pathway. Cytotherapy 2011; 13:431-40. [DOI: 10.3109/14653249.2010.542458] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
Liu X, Sun H, Yan D, Zhang L, Lv X, Liu T, Zhang W, Liu W, Cao Y, Zhou G. In vivo ectopic chondrogenesis of BMSCs directed by mature chondrocytes. Biomaterials 2010; 31:9406-14. [DOI: 10.1016/j.biomaterials.2010.08.052] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 08/23/2010] [Indexed: 01/07/2023]
|
39
|
Porada CD, Almeida-Porada G. Mesenchymal stem cells as therapeutics and vehicles for gene and drug delivery. Adv Drug Deliv Rev 2010; 62:1156-66. [PMID: 20828588 DOI: 10.1016/j.addr.2010.08.010] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2010] [Revised: 08/26/2010] [Accepted: 08/27/2010] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) possess a set of several fairly unique properties which make them ideally suited both for cellular therapies/regenerative medicine, and as vehicles for gene and drug delivery. These include: 1) relative ease of isolation; 2) the ability to differentiate into a wide variety of seemingly functional cell types of both mesenchymal and non-mesenchymal origin; 3) the ability to be extensively expanded in culture without a loss of differentiative capacity; 4) they are not only hypoimmunogenic, but they produce immunosuppression upon transplantation; 5) their pronounced anti-inflammatory properties; and 6) their ability to home to damaged tissues, tumors, and metastases following in vivo administration. In this review, we summarize the latest research in the use of mesenchymal stem cells in regenerative medicine, as immunomodulatory/anti-inflammatory agents, and as vehicles for transferring both therapeutic genes in genetic disease and genes designed to destroy malignant cells.
Collapse
|
40
|
Haack-Sorensen M, Friis T, Kastrup J. Mesenchymal stromal cell and mononuclear cell therapy in heart disease. Future Cardiol 2010; 4:481-94. [PMID: 19804342 DOI: 10.2217/14796678.4.5.481] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Despite progress in percutaneous coronary intervention, bypass surgery and drug therapy, rates of mortality and morbidity after acute coronary syndrome are high due to ventricular remodeling and heart failure. Mesenchymal stromal cells (MSCs) from adult bone marrow or adipose tissue are considered potential candidates for therapeutic regenerative treatment in cardiovascular disease. Recent animal studies have demonstrated that MSCs can induce neovascularization and improve myocardial function in postinfarction myocardial ischemic hearts. This review will focus on the present preclinical and clinical knowledge about the use of mononuclear cells and MSCs for cardiac regenerative medicine, the source of MSCs for clinical use and problems to consider when conducting clinical MSC therapy.
Collapse
Affiliation(s)
- Mandana Haack-Sorensen
- Rigshospitalet University Hospital, Cardiac Stem Cell Laboratory, The Heart Centre, Copenhagen, Denmark.
| | | | | |
Collapse
|
41
|
Mathiasen AB, Haack-Sørensen M, Kastrup J. Mesenchymal stromal cells for cardiovascular repair: current status and future challenges. Future Cardiol 2010; 5:605-17. [PMID: 19886787 DOI: 10.2217/fca.09.42] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Ischemic heart disease is the most common cause of death in most industrialized countries. Early treatment with stabilizing drugs and mechanical revascularization by percutaneous coronary intervention or coronary bypass surgery has reduced the mortality significantly. In spite of improved offers of treatments in patients with heart failure, the 1-year mortality is still approximately 20% after the diagnosis has been established. Treatment with stem cells with the potential to regenerate the damaged myocardium is a relatively new approach. Mesenchymal stromal cells are a promising source of stem cells for regenerative therapy. Clinical studies on stem cell therapy for cardiac regeneration have shown significant improvements in ventricular pump function, ventricular remodeling, myocardial perfusion, exercise potential and clinical symptoms compared with conventionally treated control groups. The results of most studies are promising, but there are still many unanswered questions. In this review, we explore present preclinical and clinical knowledge regarding the use of stem cells in cardiovascular regenerative medicine, with special focus on mesenchymal stromal cells. We take a closer look at sources of stem cells, delivery method and methods for tracking injected cells.
Collapse
Affiliation(s)
- Anders Bruun Mathiasen
- Cardiac Stem Cell laboratory & Cardiac Catheterization Laboratory 2014, The Heart Centre, Rigshospitalet, Copenhagen University Hospital & Faculty of Health Sciences, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.
| | | | | |
Collapse
|
42
|
|
43
|
Simpson D, Dudley SC. Lost in translation: what is limiting cardiomyoplasty and can tissue engineering help? Curr Stem Cell Res Ther 2009; 4:210-23. [PMID: 19492979 DOI: 10.2174/157488809789057437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Accepted: 03/19/2009] [Indexed: 01/16/2023]
Abstract
Heart failure accounts for more deaths in the United States than any other detrimental human pathology. Recently, repairing the heart after seemingly irreversible injury leading to heart failure appears to have come within reach. Cellular cardiomyoplasty, transplanting viable cell alternatives into the diseased myocardium, has emerged as a promising possible solution. Translating this approach from the laboratory to the clinic, however, has been met with several challenges, leaving many questions unanswered. This review assesses the state of investigation of several progenitor cell sources, including induced pluripotent stem cells, embryonic stem cells, bone marrow stem cells, adipose-derived adult stem cells, amniotic fluid stem cells, skeletal muscle progenitors, induced pluripotent stem cells and cardiac progenitors. Several current roadblocks to maximum success are discussed. These include understanding the need for cardiomyocyte differentiation, appreciating the role of paracrine factors, and addressing the low engraftment rates using current techniques. Tissue engineering strategies to address these obstacles and to help maximize cellular cardiomyoplasty success are reviewed.
Collapse
Affiliation(s)
- David Simpson
- Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | |
Collapse
|
44
|
Assis ACM, Carvalho JL, Jacoby BA, Ferreira RLB, Castanheira P, Diniz SOF, Cardoso VN, Goes AM, Ferreira AJ. Time-dependent migration of systemically delivered bone marrow mesenchymal stem cells to the infarcted heart. Cell Transplant 2009; 19:219-30. [PMID: 19906330 DOI: 10.3727/096368909x479677] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In this study the time course of homing and the body distribution of systemically delivered bone marrow mesenchymal stem cells (BM-MSCs) after myocardial infarction (MI) were evaluated. BM-MSCs were isolated from Wistar rats, expanded in vitro, and their phenotypical characterization was performed by flow cytometer. Rats were randomly divided into three groups: control, sham MI, and MI. BM-MSCs (5 x 10(6)) were labeled with (99m)Tc-HMPAO and injected through the tail vein 7 days after MI. Gamma camera imaging was performed at 5, 15, 30, and 60 min after cell inoculation. Due to the (99m)Tc short half-life, cell migration and location were also evaluated in heart sections using DAPI-labeled cells 7 days after transplantation. Phenotypical characterization showed that BM-MSCs were CD90(+), CD73(+), CD54(+), and CD45(-). Five minutes after (99m)Tc-HMPAO-labeled cell injection, they were detected in various tissues. The cells migrated mainly to the lungs (approximately 70%) and, in small amounts, to the heart, kidneys, spleen, and bladder. The number of cells in the heart and lungs decreased after 60 min. MI markedly increased the amount of cells in the heart, but not in the lungs, during the period of observation (4.55 +/- 0.32 vs. 6.34 +/- 0.67% of uptake in infarcted hearts). No significant differences were observed between control and sham groups. Additionally, 7 days after DAPI-labeled cells injection, they were still detected in the heart but only in infarcted areas. These results suggest that the migration of systemically delivered BM-MSCs to the heart is time dependent and MI specifically increases BM-MSCs homing to injured hearts. However, the systemic delivery is limited by cell entrapment in the lungs.
Collapse
Affiliation(s)
- Ana Carolina M Assis
- Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Paul A, Ge Y, Prakash S, Shum-Tim D. Microencapsulated stem cells for tissue repairing: implications in cell-based myocardial therapy. Regen Med 2009; 4:733-45. [DOI: 10.2217/rme.09.43] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Stem cells have the unique properties of self-renewal, pluripotency and a high proliferative capability, which contributes to a large biomass potential. Hence, these cells act as a useful source for acquiring renewable adult cell lines. This, in turn, acts as a potent therapeutic tool to treat various diseases related to the heart, liver and kidney, as well as neurodegenerative diseases such as Parkinson’s and Alzheimer’s disease. However, a major problem that must be overcome before it can be effectively implemented into the clinical setting is a suitable delivery system that can retain an optimal quantity of the cells at the targeted site for a maximal clinical benefit; a system that will give a mechanical as well as an immune protection to the foreign cells, while at the same time enhancing the yields of differentiated cells, maintaining cell microenvironments and sustaining the differentiated cell functions. To address this issue we opted for a novel delivery system, termed the ‘artificial cells’, which are semipermeable microcapsules with strong and thin multilayer membrane components with specific mass transport properties. Here, we briefly introduce the concept of artificial cells for encapsulation of stem cells and investigate the application of microencapsulation technology as an ideal tool for all stem transplantations and relate their role to the emerging field of cellular cardiomyoplasty.
Collapse
Affiliation(s)
- Arghya Paul
- Biomedical Technology & Cell Therapy Research Laboratory, Department of Biomedical Engineering & Artificial Cells & Organs Research Centre, Faculty of Medicine, McGill University, 3775 University Street, Montreal, Quebec, H3A 2B4, Canada
| | - Yin Ge
- Divisions of Cardiac Surgery & Surgical Research, The Montreal General Hospital, MUHC, 1650 Cedar Avenue, Suite C9–169, Montreal, Quebec, H3G 1A4, Canada
| | - Satya Prakash
- Biomedical Technology & Cell Therapy Research Laboratory, Department of Biomedical Engineering & Artificial Cells & Organs Research Centre, Faculty of Medicine, McGill University, 3775 University Street, Montreal, Quebec, H3A 2B4, Canada
| | - Dominique Shum-Tim
- Divisions of Cardiac Surgery & Surgical Research, The Montreal General Hospital, MUHC, 1650 Cedar Avenue, Suite C9–169, Montreal, Quebec, H3G 1A4, Canada
| |
Collapse
|
46
|
Molecular mechanisms involved in mesenchymal stem cell migration to the site of acute myocardial infarction. Int J Cell Biol 2009; 2009:904682. [PMID: 20130773 PMCID: PMC2809335 DOI: 10.1155/2009/904682] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 04/17/2009] [Accepted: 05/19/2009] [Indexed: 01/16/2023] Open
Abstract
Mesenchymal stem cells or multipotent mesenchymal stromal cells (both referred to as MSC) have been shown in some studies to have a beneficial effect on myocardial recovery after infarct. Current strategies for MSC delivery to heart involve intravenous, intraarterial, and intramuscular delivery. Different routes of MSC delivery and a lack of knowledge of the mechanisms that MSC utilise to migrate in vivo has most likely led to the marked variations in results that have been found. This review aims to summarise the current knowledge of MSC migratory mechanisms and looks to future methods of MSC manipulation prior to delivery in order to enhance MSC migration and engraftment.
Collapse
|
47
|
Enoki C, Otani H, Sato D, Okada T, Hattori R, Imamura H. Enhanced mesenchymal cell engraftment by IGF-1 improves left ventricular function in rats undergoing myocardial infarction. Int J Cardiol 2009; 138:9-18. [PMID: 19411121 DOI: 10.1016/j.ijcard.2009.04.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Revised: 03/31/2009] [Accepted: 04/05/2009] [Indexed: 11/17/2022]
Abstract
BACKGROUND We hypothesized that enhanced mesenchymal cell (MC) engraftment with insulin-like growth factor-1 (IGF-1) improves left ventricular (LV) function and survival. METHODS AND RESULTS IGF-1 (10 microg/ml) increased adhesion and inhibited apoptosis under hypoxia in vitro through activation of phosphatidylinositol 3-kinase (PI3K) in bone marrow-derived MCs obtained from transgenic rats expressing green fluorescence protein. Myocardial infarction (MI) in rats was produced by ligature of the left coronary artery. One month after MI, rat hearts were injected with MCs in the presence or absence of 10 microg/ml IGF-1 with or without PI3K inhibitor, 5 microM LY294002. IGF-1 significantly increased engraftment of MCs between 6 h and 3 days after transplantation associated with the increase in stromal cell-derived factor-1alpha in the infracted LV. The transplanted MCs had disappeared 1 month after transplantation in all groups. MC transplantation with IGF-1 significantly increased neovascularization and inhibited cardiomyocyte apoptosis 3 days and 1 month after MC transplantation. This was associated with improved LV function 1 month after MC transplantation and eventually survival. LY294002 abrogated all of the beneficial effects of MC transplantation with IGF-1. IGF-1 alone had no effect on neovascularization and did not improve LV function and/or survival. CONCLUSIONS These results suggest that IGF-1 improves engraftment of MCs at the time of transplantation via activation of PI3K and this improved engraftment of MCs may be attributed to an increased neovascularization and inhibition of cardiomyocyte death, leading to improvement of LV function and prolongation of survival despite the eventual loss of the transplanted MCs.
Collapse
Affiliation(s)
- Chiharu Enoki
- Department of Thoracic and Cardiovascular Surgery, Kansai Medical University, Moriguchi City, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Chacko SM, Khan M, Kuppusamy ML, Pandian RP, Varadharaj S, Selvendiran K, Bratasz A, Rivera BK, Kuppusamy P. Myocardial oxygenation and functional recovery in infarct rat hearts transplanted with mesenchymal stem cells. Am J Physiol Heart Circ Physiol 2009; 296:H1263-73. [PMID: 19286938 DOI: 10.1152/ajpheart.01311.2008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Stem cell therapy for myocardial tissue repair is limited by the poor survival of transplanted cells, possibly because of inadequate supply of oxygen and nutrients. The purpose of this study was to assess the oxygenation level and functional recovery after allogenic transplantation of mesenchymal stem cells (MSC) in a rat model of myocardial infarction (MI). Myocardial oxygen tension (Po(2)) was measured by electron paramagnetic resonance oximetry using an implantable oxygen-sensing spin probe (OxySpin). MSCs incubated with OxySpins showed substantial uptake of the probe without affecting its oxygen sensitivity or calibration. The cells internalized with OxySpins were able to differentiate into osteogenic, adipogenic, cardiomyocyte, and endothelial cell lineages. The labeled cells tested positive for CD44 and CD29 markers and negative for the hematopoietic markers CD14 and CD45. For the in vivo studies, MI was induced in rats by permanently ligating the left anterior descending coronary artery. MSCs with OxySpins were transplanted in the infarct region of hearts. A significant increase in Po(2) was observed in the MSC group compared with the untreated MI group (18.1 +/- 2.6 vs. 13.0 +/- 1.8 mmHg, n = 4, P < 0.05) at 4 wk after transplantation. Echocardiography showed a significant improvement in ejection fraction and fraction shortening, which inversely correlated with the magnitude of fibrosis in the treated hearts. The cell-transplanted hearts also showed an increase in vascular endothelial growth factor level and capillary density in the infarct region. The study established our ability to measure and correlate changes in myocardial tissue oxygenation with cardiac function in infarcted rat hearts treated with MSCs.
Collapse
Affiliation(s)
- Simi M Chacko
- Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State Univ., 420 W. 12th Ave, Rm. 114, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lee BC, Hsu HC, Tseng WYI, Chen CY, Lin HJ, Ho YL, Su MJ, Chen MF. Cell therapy generates a favourable chemokine gradient for stem cell recruitment into the infarcted heart in rabbits. Eur J Heart Fail 2009; 11:238-45. [PMID: 19147447 DOI: 10.1093/eurjhf/hfn035] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Stem cell recruitment into the heart is determined by a concentration gradient of stromal-derived factor 1 (SDF-1) from bone marrow to peripheral blood and from blood to injured myocardium. However, this gradient is decreased in chronic myocardial infarction (MI). This study evaluated the effect of cell therapy using bone marrow stromal cells (BMSCs) on an SDF-1 gradient in post-infarction rabbits. METHODS AND RESULTS Myocardial infarction was induced in male New Zealand white rabbits (2.5-3 kg) by ligation of the left anterior descending coronary artery. Two months later, the rabbits were randomized to either saline or BMSC (2 x 10(6) autologous BMSCs injected into the left ventricular cavity) treatment. Four weeks after therapy, the SDF-1 gradients from bone marrow to blood and from blood to myocardium increased in the BMSC group compared with the saline group. This was accompanied by an increase in cells positive for CD34, CD117, and STRO-1 in the myocardium, resulting in more capillary density, better cardiac function, and a decrease in infarct size. CONCLUSION Generation of an SDF-1 gradient towards the heart is a novel effect of BMSC-based cell therapy. This effect facilitates stem cell recruitment into remodelled myocardium and supports improvement in cardiac function.
Collapse
Affiliation(s)
- Bai-Chin Lee
- Department of Internal Medicine, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei, Taiwan, Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kim YJ, Huh YM, Choe KO, Choi BW, Choi EJ, Jang Y, Lee JM, Suh JS. In vivo magnetic resonance imaging of injected mesenchymal stem cells in rat myocardial infarction; simultaneous cell tracking and left ventricular function measurement. Int J Cardiovasc Imaging 2009; 25 Suppl 1:99-109. [PMID: 19132547 DOI: 10.1007/s10554-008-9407-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 12/17/2008] [Indexed: 02/06/2023]
Abstract
To determine whether magnetic resonance imaging (MRI) can enable magnetically labeled mesenchymal stem cell (MSC) tracking and simultaneous in vivo functional data acquisition in rat models of myocardial infarction. Superparamagnetic iron oxide-laden human MSCs were injected into rat myocardium infarcted by cryoinjury 3 weeks after myocardial infarction. The control group received cell-free media injection. Before injection and for 3 months after, in vivo serial MRI was performed. Electrocardiography-gated gradient echo sequence MRI and cine MRI were performed for in vivo cell tracking and assessing cardiac function using left ventricular ejection fraction (LVEF), respectively. MRI revealed a persistent signal-void representing iron-laden MSCs until ten post-injection weeks. Serial follow-up MRI revealed that LVEF was significantly higher in the MSC injection group than in the control group. We conclude that MRI enables in vivo tracking of injected cells and evaluation of the long-term therapeutic potential of MSCs for myocardial infarction.
Collapse
Affiliation(s)
- Young Jin Kim
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemun-gu, 120-752 Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|