1
|
Wang X, Wang Q, Xia Z, Yang Y, Dai X, Zhang C, Wang J, Xu Y. Mesenchymal stromal cell therapies for traumatic neurological injuries. J Transl Med 2024; 22:1055. [PMID: 39578845 PMCID: PMC11583761 DOI: 10.1186/s12967-024-05725-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/01/2024] [Indexed: 11/24/2024] Open
Abstract
Improved treatment options are urgently needed for neurological injuries resulting from trauma or iatrogenic events causing long-term disabilities that severely impact patients' quality of life. In vitro and animal studies have provided promising proof-of-concept examples of regenerative therapies using mesenchymal stromal cells (MSC) for a wide range of pathological conditions. Over the previous decade, various MSC-based therapies have been investigated in clinical trials to treat traumatic neurological injuries. However, while the safety and feasibility of MSC treatments has been established, the patient outcomes in these studies have not demonstrated significant success in the translation of MSC regenerative therapy for the treatment of human brain and spinal cord injuries. Herein, we have reviewed the literature and ongoing registered trials on the application of MSC for the treatment of traumatic brain injury, traumatic spinal cord injury, and peripheral nerve injury. We have focused on the shortcomings and technological hurdles that must be overcome to further advance clinical research to phase 3 trials, and we discuss recent advancements that represent potential solutions to these obstacles to progress.
Collapse
Affiliation(s)
- Xiujuan Wang
- Technology Department, Tianjin Everunion Biotechnology Co., Ltd, SOHO Nexus Center, No. 19A East 3rd Ring North Road, Chaoyang District, Beijing, 100020, China
| | - Qian Wang
- HELP Therapeutics Co., Ltd, No. 568 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, E12 Avenida da Universidade, Macau, 519000, SAR, China
| | - Ziyao Xia
- Department of Ophthalmology, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Ying Yang
- Technology Department, Tianjin Everunion Biotechnology Co., Ltd, SOHO Nexus Center, No. 19A East 3rd Ring North Road, Chaoyang District, Beijing, 100020, China
| | - Xunan Dai
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Chun Zhang
- Department of Ophthalmology, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
| | - Jiaxian Wang
- HELP Therapeutics Co., Ltd, No. 568 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China.
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, E12 Avenida da Universidade, Macau, 519000, SAR, China.
| | - Yongsheng Xu
- Technology Department, Tianjin Everunion Biotechnology Co., Ltd, SOHO Nexus Center, No. 19A East 3rd Ring North Road, Chaoyang District, Beijing, 100020, China.
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
2
|
Huang X, Henck J, Qiu C, Sreenivasan VKA, Balachandran S, Amarie OV, Hrabě de Angelis M, Behncke RY, Chan WL, Despang A, Dickel DE, Duran M, Feuchtinger A, Fuchs H, Gailus-Durner V, Haag N, Hägerling R, Hansmeier N, Hennig F, Marshall C, Rajderkar S, Ringel A, Robson M, Saunders LM, da Silva-Buttkus P, Spielmann N, Srivatsan SR, Ulferts S, Wittler L, Zhu Y, Kalscheuer VM, Ibrahim DM, Kurth I, Kornak U, Visel A, Pennacchio LA, Beier DR, Trapnell C, Cao J, Shendure J, Spielmann M. Single-cell, whole-embryo phenotyping of mammalian developmental disorders. Nature 2023; 623:772-781. [PMID: 37968388 PMCID: PMC10665194 DOI: 10.1038/s41586-023-06548-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/16/2023] [Indexed: 11/17/2023]
Abstract
Mouse models are a critical tool for studying human diseases, particularly developmental disorders1. However, conventional approaches for phenotyping may fail to detect subtle defects throughout the developing mouse2. Here we set out to establish single-cell RNA sequencing of the whole embryo as a scalable platform for the systematic phenotyping of mouse genetic models. We applied combinatorial indexing-based single-cell RNA sequencing3 to profile 101 embryos of 22 mutant and 4 wild-type genotypes at embryonic day 13.5, altogether profiling more than 1.6 million nuclei. The 22 mutants represent a range of anticipated phenotypic severities, from established multisystem disorders to deletions of individual regulatory regions4,5. We developed and applied several analytical frameworks for detecting differences in composition and/or gene expression across 52 cell types or trajectories. Some mutants exhibit changes in dozens of trajectories whereas others exhibit changes in only a few cell types. We also identify differences between widely used wild-type strains, compare phenotyping of gain- versus loss-of-function mutants and characterize deletions of topological associating domain boundaries. Notably, some changes are shared among mutants, suggesting that developmental pleiotropy might be 'decomposable' through further scaling of this approach. Overall, our findings show how single-cell profiling of whole embryos can enable the systematic molecular and cellular phenotypic characterization of mouse mutants with unprecedented breadth and resolution.
Collapse
Affiliation(s)
- Xingfan Huang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA
| | - Jana Henck
- Institute of Human Genetics, University Medical Center Schleswig-Holstein, University of Lübeck & Kiel University, Lübeck, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Chengxiang Qiu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Varun K A Sreenivasan
- Institute of Human Genetics, University Medical Center Schleswig-Holstein, University of Lübeck & Kiel University, Lübeck, Germany
| | - Saranya Balachandran
- Institute of Human Genetics, University Medical Center Schleswig-Holstein, University of Lübeck & Kiel University, Lübeck, Germany
| | - Oana V Amarie
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Rose Yinghan Behncke
- Institute of Medical Genetics and Human Genetics of the Charité, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BCRT, Berlin, Germany
| | - Wing-Lee Chan
- Institute of Medical Genetics and Human Genetics of the Charité, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BCRT, Berlin, Germany
| | - Alexandra Despang
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BCRT, Berlin, Germany
| | - Diane E Dickel
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Madeleine Duran
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Annette Feuchtinger
- Core Facility Pathology & Tissue Analytics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Natja Haag
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Rene Hägerling
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Medical Genetics and Human Genetics of the Charité, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BCRT, Berlin, Germany
| | - Nils Hansmeier
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Medical Genetics and Human Genetics of the Charité, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BCRT, Berlin, Germany
| | | | - Cooper Marshall
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA
| | | | - Alessa Ringel
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Medical Genetics and Human Genetics of the Charité, Berlin, Germany
| | - Michael Robson
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Lauren M Saunders
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Patricia da Silva-Buttkus
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Nadine Spielmann
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Sanjay R Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Sascha Ulferts
- Institute of Medical Genetics and Human Genetics of the Charité, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BCRT, Berlin, Germany
| | - Lars Wittler
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Yiwen Zhu
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | | | - Daniel M Ibrahim
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BCRT, Berlin, Germany
| | - Ingo Kurth
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Uwe Kornak
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Axel Visel
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | | | - David R Beier
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Junyue Cao
- Laboratory of Single-Cell Genomics and Population Dynamics, The Rockefeller University, New York, NY, USA.
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
- Howard Hughes Medical Institute, Seattle, WA, USA.
| | - Malte Spielmann
- Institute of Human Genetics, University Medical Center Schleswig-Holstein, University of Lübeck & Kiel University, Lübeck, Germany.
- Max Planck Institute for Molecular Genetics, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany.
| |
Collapse
|
3
|
Nguyen QT, Thanh LN, Hoang VT, Phan TTK, Heke M, Hoang DM. Bone Marrow-Derived Mononuclear Cells in the Treatment of Neurological Diseases: Knowns and Unknowns. Cell Mol Neurobiol 2023; 43:3211-3250. [PMID: 37356043 PMCID: PMC11410020 DOI: 10.1007/s10571-023-01377-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/14/2023] [Indexed: 06/27/2023]
Abstract
Bone marrow-derived mononuclear cells (BMMNCs) have been used for decades in preclinical and clinical studies to treat various neurological diseases. However, there is still a knowledge gap in the understanding of the underlying mechanisms of BMMNCs in the treatment of neurological diseases. In addition, prerequisite factors for the efficacy of BMMNC administration, such as the optimal route, dose, and number of administrations, remain unclear. In this review, we discuss known and unknown aspects of BMMNCs, including the cell harvesting, administration route and dose; mechanisms of action; and their applications in neurological diseases, including stroke, cerebral palsy, spinal cord injury, traumatic brain injury, amyotrophic lateral sclerosis, autism spectrum disorder, and epilepsy. Furthermore, recommendations on indications for BMMNC administration and the advantages and limitations of BMMNC applications for neurological diseases are discussed. BMMNCs in the treatment of neurological diseases. BMMNCs have been applied in several neurological diseases. Proposed mechanisms for the action of BMMNCs include homing, differentiation and paracrine effects (angiogenesis, neuroprotection, and anti-inflammation). Further studies should be performed to determine the optimal cell dose and administration route, the roles of BMMNC subtypes, and the indications for the use of BMMNCs in neurological conditions with and without genetic abnormalities.
Collapse
Affiliation(s)
- Quyen Thi Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, 458 Minh Khai, Hai Ba Trung, Hanoi, 11622, Vietnam
| | - Liem Nguyen Thanh
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, 458 Minh Khai, Hai Ba Trung, Hanoi, 11622, Vietnam.
- College of Health Science, Vin University, Vinhomes Ocean Park, Gia Lam District, Hanoi, 12400, Vietnam.
- Vinmec International Hospital-Times City, Vinmec Healthcare System, 458 Minh Khai, Hanoi, 11622, Vietnam.
| | - Van T Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, 458 Minh Khai, Hai Ba Trung, Hanoi, 11622, Vietnam
| | - Trang T K Phan
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, 458 Minh Khai, Hai Ba Trung, Hanoi, 11622, Vietnam
| | - Michael Heke
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Duc M Hoang
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, 458 Minh Khai, Hai Ba Trung, Hanoi, 11622, Vietnam
| |
Collapse
|
4
|
Bakhtiary N, Pezeshki-Modaress M, Najmoddin N. Wet-electrospinning of nanofibrous magnetic composite 3-D scaffolds for enhanced stem cells neural differentiation. Chem Eng Sci 2022. [DOI: 10.1016/j.ces.2022.118144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
5
|
Pandey RS, Krebs MP, Bolisetty MT, Charette JR, Naggert JK, Robson P, Nishina PM, Carter GW. Single-Cell RNA Sequencing Reveals Molecular Features of Heterogeneity in the Murine Retinal Pigment Epithelium. Int J Mol Sci 2022; 23:10419. [PMID: 36142331 PMCID: PMC9499471 DOI: 10.3390/ijms231810419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/09/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Transcriptomic analysis of the mammalian retinal pigment epithelium (RPE) aims to identify cellular networks that influence ocular development, maintenance, function, and disease. However, available evidence points to RPE cell heterogeneity within native tissue, which adds complexity to global transcriptomic analysis. Here, to assess cell heterogeneity, we performed single-cell RNA sequencing of RPE cells from two young adult male C57BL/6J mice. Following quality control to ensure robust transcript identification limited to cell singlets, we detected 13,858 transcripts among 2667 and 2846 RPE cells. Dimensional reduction by principal component analysis and uniform manifold approximation and projection revealed six distinct cell populations. All clusters expressed transcripts typical of RPE cells; the smallest (C1, containing 1-2% of total cells) exhibited the hallmarks of stem and/or progenitor (SP) cells. Placing C1-6 along a pseudotime axis suggested a relative decrease in melanogenesis and SP gene expression and a corresponding increase in visual cycle gene expression upon RPE maturation. K-means clustering of all detected transcripts identified additional expression patterns that may advance the understanding of RPE SP cell maintenance and the evolution of cellular metabolic networks during development. This work provides new insights into the transcriptome of the mouse RPE and a baseline for identifying experimentally induced transcriptional changes in future studies of this tissue.
Collapse
Affiliation(s)
- Ravi S. Pandey
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr., Farmington, CT 06032, USA
| | - Mark P. Krebs
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Mohan T. Bolisetty
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr., Farmington, CT 06032, USA
| | | | | | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Dr., Farmington, CT 06032, USA
| | - Patsy M. Nishina
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | | |
Collapse
|
6
|
Xie JL, Wang XR, Li MM, Tao ZH, Teng WW, Saijilafu. Mesenchymal Stromal Cell Therapy in Spinal Cord Injury: Mechanisms and Prospects. Front Cell Neurosci 2022; 16:862673. [PMID: 35722621 PMCID: PMC9204037 DOI: 10.3389/fncel.2022.862673] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) often leads to severe motor, sensory, and autonomic dysfunction in patients and imposes a huge economic cost to individuals and society. Due to its complicated pathophysiological mechanism, there is not yet an optimal treatment available for SCI. Mesenchymal stromal cells (MSCs) are promising candidate transplant cells for use in SCI treatment. The multipotency of MSCs, as well as their rich trophic and immunomodulatory abilities through paracrine signaling, are expected to play an important role in neural repair. At the same time, the simplicity of MSCs isolation and culture and the bypassing of ethical barriers to stem cell transplantation make them more attractive. However, the MSCs concept has evolved in a specific research context to encompass different populations of cells with a variety of biological characteristics, and failure to understand this can undermine the quality of research in the field. Here, we review the development of the concept of MSCs in order to clarify misconceptions and discuss the controversy in MSCs neural differentiation. We also summarize a potential role of MSCs in SCI treatment, including their migration and trophic and immunomodulatory effects, and their ability to relieve neuropathic pain, and we also highlight directions for future research.
Collapse
Affiliation(s)
- Ji-Le Xie
- Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, China,Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China
| | - Xing-Ran Wang
- Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China
| | - Mei-Mei Li
- Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China
| | - Zi-Han Tao
- Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China
| | - Wen-Wen Teng
- Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China
| | - Saijilafu
- Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, China,Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China,*Correspondence: Saijilafu,
| |
Collapse
|
7
|
Shaimardanova AA, Chulpanova DS, Mullagulova AI, Afawi Z, Gamirova RG, Solovyeva VV, Rizvanov AA. Gene and Cell Therapy for Epilepsy: A Mini Review. Front Mol Neurosci 2022; 15:868531. [PMID: 35645733 PMCID: PMC9132249 DOI: 10.3389/fnmol.2022.868531] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/30/2022] [Indexed: 11/16/2022] Open
Abstract
Epilepsy is a chronic non-infectious disease of the brain, characterized primarily by recurrent unprovoked seizures, defined as an episode of disturbance of motor, sensory, autonomic, or mental functions resulting from excessive neuronal discharge. Despite the advances in the treatment achieved with the use of antiepileptic drugs and other non-pharmacological therapies, about 30% of patients suffer from uncontrolled seizures. This review summarizes the currently available methods of gene and cell therapy for epilepsy and discusses the development of these approaches. Currently, gene therapy for epilepsy is predominantly adeno-associated virus (AAV)-mediated delivery of genes encoding neuro-modulatory peptides, neurotrophic factors, enzymes, and potassium channels. Cell therapy for epilepsy is represented by the transplantation of several types of cells such as mesenchymal stem cells (MSCs), bone marrow mononuclear cells, neural stem cells, and MSC-derived exosomes. Another approach is encapsulated cell biodelivery, which is the transplantation of genetically modified cells placed in capsules and secreting various therapeutic agents. The use of gene and cell therapy approaches can significantly improve the condition of patient with epilepsy. Therefore, preclinical, and clinical studies have been actively conducted in recent years to prove the benefits and safety of these strategies.
Collapse
Affiliation(s)
| | - Daria S. Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Aysilu I. Mullagulova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Zaid Afawi
- Center for Neuroscience, Ben Gurion University of the Negev, Be’er Sheva, Israel
| | - Rimma G. Gamirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- *Correspondence: Albert A. Rizvanov,
| |
Collapse
|
8
|
Issa SS, Shaimardanova AA, Valiullin VV, Rizvanov AA, Solovyeva VV. Mesenchymal Stem Cell-Based Therapy for Lysosomal Storage Diseases and Other Neurodegenerative Disorders. Front Pharmacol 2022; 13:859516. [PMID: 35308211 PMCID: PMC8924473 DOI: 10.3389/fphar.2022.859516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/17/2022] [Indexed: 12/11/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of approximately 50 genetic disorders caused by mutations in genes coding enzymes that are involved in cell degradation and transferring lipids and other macromolecules. Accumulation of lipids and other macromolecules in lysosomes leads to the destruction of affected cells. Although the clinical manifestations of different LSDs vary greatly, more than half of LSDs have symptoms of central nervous system neurodegeneration, and within each disorder there is a considerable variation, ranging from severe, infantile-onset forms to attenuated adult-onset disease, sometimes with distinct clinical features. To date, treatment options for this group of diseases remain limited, which highlights the need for further development of innovative therapeutic approaches, that can not only improve the patients' quality of life, but also provide full recovery for them. In many LSDs stem cell-based therapy showed promising results in preclinical researches. This review discusses using mesenchymal stem cells for different LSDs therapy and other neurodegenerative diseases and their possible limitations.
Collapse
Affiliation(s)
- Shaza S Issa
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Faculty of Biology, Saint Petersburg State University, Saint Petersburg, Russia
| | - Alisa A Shaimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Victor V Valiullin
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Valeriya V Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
9
|
Kadkhodaeian HA. Mesenchymal Stem Cells: Signaling Pathways in Transdifferentiation Into Retinal Progenitor Cells. Basic Clin Neurosci 2021; 12:29-42. [PMID: 33995925 PMCID: PMC8114861 DOI: 10.32598/bcn.9.10.510] [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: 07/16/2018] [Revised: 08/25/2018] [Accepted: 02/02/2020] [Indexed: 11/29/2022] Open
Abstract
Several signaling pathways and transcription factors control the cell fate in its in vitro development and differentiation. The orchestrated use of these factors results in cell specification. In coculture methods, many of these factors secrete from host cells but control the process. Today, transcription factors required for retinal progenitor cells are well known, but the generation of these cells from mesenchymal stem cells is an ideal goal. The purpose of the paper is to review novel methods for retinal progenitor cell production and selecting a set of signaling molecules in the presence of adult retinal pigment epithelium and extraocular mesenchyme acting as inducers of retinal cell differentiation.
Collapse
|
10
|
Karakaş N, Bay S, Türkel N, Öztunç N, Öncül M, Bilgen H, Shah K, Şahin F, Öztürk G. Neurons from human mesenchymal stem cells display both spontaneous and stimuli responsive activity. PLoS One 2020; 15:e0228510. [PMID: 32407317 PMCID: PMC7224507 DOI: 10.1371/journal.pone.0228510] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells have the ability to transdifferentiate into neurons and therefore one of the potential adult stem cell source for neuronal tissue regeneration applications and understanding neurodevelopmental processes. In many studies on human mesenchymal stem cell (hMSC) derived neurons, success in neuronal differentiation was limited to neuronal protein expressions which is not statisfactory in terms of neuronal activity. Established neuronal networks seen in culture have to be investigated in terms of synaptic signal transmission ability to develop a culture model for human neurons and further studying the mechanism of neuronal differentiation and neurological pathologies. Accordingly, in this study, we analysed the functionality of bone marrow hMSCs differentiated into neurons by a single step cytokine-based induction protocol. Neurons from both primary hMSCs and hMSC cell line displayed spontaneous activity (≥75%) as demonstrated by Ca++ imaging. Furthermore, when electrically stimulated, hMSC derived neurons (hMd-Neurons) matched the response of a typical neuron in the process of maturation. Our results reveal that a combination of neuronal inducers enhance differentiation capacity of bone marrow hMSCs into high yielding functional neurons with spontaneous activity and mature into electrophysiologically active state. Conceptually, we suggest these functional hMd-Neurons to be used as a tool for disease modelling of neuropathologies and neuronal differentiation studies.
Collapse
Affiliation(s)
- Nihal Karakaş
- Medical Biology Department, School of Medicine, İstanbul Medipol University, İstanbul, Turkey
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, İstanbul, Turkey
- * E-mail:
| | - Sadık Bay
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, İstanbul, Turkey
| | - Nezaket Türkel
- Genetics and Bioengineering Department, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| | - Nurşah Öztunç
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, İstanbul, Turkey
- Medical Biology and Genetics Program, Graduate School of Health Sciences, İstanbul Medipol University, İstanbul, Turkey
| | - Merve Öncül
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, İstanbul, Turkey
| | - Hülya Bilgen
- Center for Bone Marrow Transplantation, İstanbul Medipol University Hospital, İstanbul, Turkey
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging, Brigham and Woman’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Fikrettin Şahin
- Genetics and Bioengineering Department, Faculty of Engineering, Yeditepe University, İstanbul, Turkey
| | - Gürkan Öztürk
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), İstanbul Medipol University, İstanbul, Turkey
- Physiology Department, International School of Medicine, İstanbul Medipol University, İstanbul, Turkey
| |
Collapse
|
11
|
Weiss JN, Levy S. Stem Cell Ophthalmology Treatment Study (SCOTS): bone marrow derived stem cells in the treatment of Dominant Optic Atrophy. Stem Cell Investig 2019; 6:41. [PMID: 32039263 DOI: 10.21037/sci.2019.11.01] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/14/2019] [Indexed: 01/01/2023]
Abstract
Background We report the results of 6 patients with Dominant Optic Atrophy (DOA) who met inclusion criteria and were treated in the Stem Cell Ophthalmology Treatment Study (SCOTS). SCOTS/SCOTS 2 is an Institutional Review Board approved and NIH registered (NCT03011541) clinical study that uses autologous bone marrow derived stem cells (BMSC) in the treatment of optic nerve and retinal disease. Methods This is an open label, non-randomized clinical study using natural history of the disease as the comparator. BMSC were separated from aspirated autologous bone marrow with minimal manipulation using an FDA cleared Class II medical device. Patients were treated with combinations of retrobulbar, subtenons, intravitreal or subretinal placement of BMSC followed by intravenous injection of BMSC depending on the arm of the study chosen. There were no surgical complications. Results Of the patients treated, 83.3% (5 of 6 patients) experienced visual improvements and in all of these cases both eyes improved. Ten eyes or 83.3% experienced gains in visual acuity with a median improvement of 2.125 Snellen lines, or approximately 10.63 letters. Two eyes were considered unchanged compared to longstanding measurements. Using LogMAR, the average improvement in vision for all eyes was 29.5%. The averagevisual acuity increasein eyes that improved was 33.3%. Findings were statistically significant with P<0.001. Conclusions Using autologous BMSC per protocols developed in the SCOTS/SCOTS 2 clinical studies resulted in statistically significant visual acuity improvements in patients with DOA or Kjers Optic Neuropathy. Improvements occurred in 83.3% of eyes and averaged 29.5%. Mitochondrial transfer and neuroprotective exosome secretions from the BMSC may have been key to the improvements observed in this mitochondrial disease.
Collapse
|
12
|
Mukhamedshina Y, Shulman I, Ogurcov S, Kostennikov A, Zakirova E, Akhmetzyanova E, Rogozhin A, Masgutova G, James V, Masgutov R, Lavrov I, Rizvanov A. Mesenchymal Stem Cell Therapy for Spinal Cord Contusion: A Comparative Study on Small and Large Animal Models. Biomolecules 2019; 9:E811. [PMID: 31805639 PMCID: PMC6995633 DOI: 10.3390/biom9120811] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/05/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022] Open
Abstract
Here, we provide a first comparative study of the therapeutic potential of allogeneic mesenchymal stem cells derived from bone marrow (BM-MSCs), adipose tissue (AD-MSCs), and dental pulp (DP-MSCs) embedded in fibrin matrix, in small (rat) and large (pig) spinal cord injury (SCI) models during subacute period of spinal contusion. Results of behavioral, electrophysiological, and histological assessment as well as immunohistochemistry and real-time polymerase chain reaction analysis suggest that application of AD-MSCs combined with a fibrin matrix within the subacute period in rats (2 weeks after injury), provides significantly higher post-traumatic regeneration compared to a similar application of BM-MSCs or DP-MSCs. Within the rat model, use of AD-MSCs resulted in a marked change in: (1) restoration of locomotor activity and conduction along spinal axons; (2) reduction of post-traumatic cavitation and enhancing tissue retention; and (3) modulation of microglial and astroglial activation. The effect of an autologous application of AD-MSCs during the subacute period after spinal contusion was also confirmed in pigs (6 weeks after injury). Effects included: (1) partial restoration of the somatosensory spinal pathways; (2) reduction of post-traumatic cavitation and enhancing tissue retention; and (3) modulation of astroglial activation in dorsal root entry zone. However, pigs only partially replicated the findings observed in rats. Together, these results indicate application of AD-MSCs embedded in fibrin matrix at the site of SCI during the subacute period can facilitate regeneration of nervous tissue in rats and pigs. These results, for the first time, provide robust support for the use of AD-MSC to treat subacute SCI.
Collapse
Affiliation(s)
- Yana Mukhamedshina
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Department of Histology, Cytology, and Embryology, Kazan State Medical University, 420012 Kazan, Russia
| | - Iliya Shulman
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Republic Clinical Hospital, 420138 Kazan, Russia
| | - Sergei Ogurcov
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Republic Clinical Hospital, 420138 Kazan, Russia
| | - Alexander Kostennikov
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
| | - Elena Zakirova
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
| | - Elvira Akhmetzyanova
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
| | - Alexander Rogozhin
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Department of Neurology, Kazan State Medical Academy–Branch Campus of the Federal State Budgetary Edicational Institution of Father Professional Education «Russian Medical Academy of Continuous Professional Education», 420012 Kazan, Russia
| | - Galina Masgutova
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
| | - Victoria James
- Division of Biomedical Science, School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham Biodiscovery Institute, University Park, Nottingham NG7 2RD, UK;
| | - Ruslan Masgutov
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Republic Clinical Hospital, 420138 Kazan, Russia
| | - Igor Lavrov
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Albert Rizvanov
- Clinical Research Center for Precision and Regenerative Medicine, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (I.S.); (S.O.); (A.K.); (E.Z.); (E.A.); (A.R.); (G.M.); (R.M.); (I.L.); (A.R.)
| |
Collapse
|
13
|
Brboric A, Vasylovska S, Saarimäki-Vire J, Espes D, Caballero-Corbalan J, Larfors G, Otonkoski T, Lau J. Characterization of neural crest-derived stem cells isolated from human bone marrow for improvement of transplanted islet function. Ups J Med Sci 2019; 124:228-237. [PMID: 31623497 PMCID: PMC6968573 DOI: 10.1080/03009734.2019.1658661] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: Murine boundary cap-derived neural crest stem cells (NCSCs) are capable of enhancing islet function by stimulating beta cell proliferation as well as increasing the neural and vascular density in the islets both in vitro and in vivo. This study aimed to isolate NCSC-like cells from human bone marrow.Methods: CD271 magnetic cell separation and culture techniques were used to purify a NCSC-enriched population of human bone marrow. Analyses of the CD271+ and CD271- fractions in terms of protein expression were performed, and the capacity of the CD271+ bone marrow cells to form 3-dimensional spheres when grown under non-adherent conditions was also investigated. Moreover, the NCSC characteristics of the CD271+ cells were evaluated by their ability to migrate toward human islets as well as human islet-like cell clusters (ICC) derived from pluripotent stem cells.Results: The CD271+ bone marrow population fulfilled the criterion of being multipotent stem cells, having the potential to differentiate into glial cells, neurons as well as myofibroblasts in vitro. They had the capacity to form 3-dimensional spheres as well as an ability to migrate toward human islets, further supporting their NCSC identity. Additionally, we demonstrated similar migration features toward stem cell-derived ICC.Conclusion: The results support the NCSC identity of the CD271-enriched human bone marrow population. It remains to investigate whether the human bone marrow-derived NCSCs have the ability to improve transplantation efficacy of not only human islets but stem cell-derived ICC as well.
Collapse
Affiliation(s)
- Anja Brboric
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Jonna Saarimäki-Vire
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Daniel Espes
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Gunnar Larfors
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Timo Otonkoski
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- CONTACT Joey Lau Department of Medical Cell Biology, Uppsala University, Husargatan 3, Box 571, SE-751 23 Uppsala, Sweden
| |
Collapse
|
14
|
Human orbital adipose tissue-derived mesenchymal stem cells possess neuroectodermal differentiation and repair ability. Cell Tissue Res 2019; 378:531-542. [PMID: 31377878 DOI: 10.1007/s00441-019-03072-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 07/08/2019] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) are used extensively in cell therapy for repair and regeneration of several organs and tissues. Cell therapy is a valuable option to treat neurodegenerative diseases and MSCs have been shown to improve neuronal function through direct differentiation or secretion of neurotrophic factors. In the present study, we isolated and characterized stem cells from medial and central orbital adipose tissue and found that they could be grown in a monolayer culture. The orbital adipose tissue-derived cells were identical to bone marrow-derived MSCs in their cell surface marker expression, gene expression and multilineage differentiation abilities. The orbital adipose-derived MSCs (OAMSCs) express several neurotrophic factors, possess neuroectodermal differentiation ability and secreted factors from OAMSCs abrogated neuronal cell damage induced by oxidative stress. Thus, OAMSCs might be a valuable cell source for treatment of neurological diseases and to reverse oxidative damage in the neuronal cells.
Collapse
|
15
|
Pinchi E, Frati A, Cantatore S, D'Errico S, Russa RL, Maiese A, Palmieri M, Pesce A, Viola RV, Frati P, Fineschi V. Acute Spinal Cord Injury: A Systematic Review Investigating miRNA Families Involved. Int J Mol Sci 2019; 20:1841. [PMID: 31013946 PMCID: PMC6515063 DOI: 10.3390/ijms20081841] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/06/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023] Open
Abstract
Acute traumatic spinal cord injury (SCI) involves primary and secondary injury mechanisms. The primary mechanism is related to the initial traumatic damage caused by the damaging impact and this damage is irreversible. Secondary mechanisms, which begin as early as a few minutes after the initial trauma, include processes such as spinal cord ischemia, cellular excitotoxicity, ionic dysregulation, and free radical-mediated peroxidation. SCI is featured by different forms of injury, investigating the pathology and degree of clinical diagnosis and treatment strategies, the animal models that have allowed us to better understand this entity and, finally, the role of new diagnostic and prognostic tools such as miRNA could improve our ability to manage this pathological entity. Autopsy could benefit from improvements in miRNA research: the specificity and sensitivity of miRNAs could help physicians in determining the cause of death, besides the time of death.
Collapse
Affiliation(s)
- Enrica Pinchi
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
| | - Alessandro Frati
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
- NESMOS Department ⁻ Neurosurgery Division, "Sapienza" University of Roma, 00189 Rome, Italy.
| | - Santina Cantatore
- Forensic Pathology Institute, University of Foggia, 71122 Foggia, Italy.
| | - Stefano D'Errico
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
- Legal Medicine Division, Ospedale Sant'Andrea, 00189 Rome, Italy.
| | - Raffaele La Russa
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
| | - Aniello Maiese
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
| | - Mauro Palmieri
- NESMOS Department ⁻ Neurosurgery Division, "Sapienza" University of Roma, 00189 Rome, Italy.
| | - Alessandro Pesce
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
- NESMOS Department ⁻ Neurosurgery Division, "Sapienza" University of Roma, 00189 Rome, Italy.
| | | | - Paola Frati
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
| | - Vittorio Fineschi
- Department SAIMLAL, "Sapienza" University of Roma, 00161 Rome, Italy.
- IRCCS "Neuromed" ⁻ Neurosurgery Division, 86077 Pozzilli. (IS) Italy.
| |
Collapse
|
16
|
Urrutia DN, Caviedes P, Mardones R, Minguell JJ, Vega-Letter AM, Jofre CM. Comparative study of the neural differentiation capacity of mesenchymal stromal cells from different tissue sources: An approach for their use in neural regeneration therapies. PLoS One 2019; 14:e0213032. [PMID: 30856179 PMCID: PMC6437714 DOI: 10.1371/journal.pone.0213032] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/13/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can trans/differentiate to neural precursors and/or
mature neurons and promote neuroprotection and neurogenesis. The above could
greatly benefit neurodegenerative disorders as well as in the treatment of
post-traumatic and hereditary diseases of the central nervous system (CNS). In
order to attain an ideal source of adult MSCs for the treatment of CNS diseases,
adipose tissue, bone marrow, skin and umbilical cord derived MSCs were isolated
and studied to explore differences with regard to neural differentiation
capacity. In this study, we demonstrated that MSCs from several tissues can
differentiate into neuron-like cells and differentially express progenitors and
mature neural markers. Adipose tissue MSCs exhibited significantly higher
expression of neural markers and had a faster proliferation rate. Our results
suggest that adipose tissue MSCs are the best candidates for the use in
neurological diseases.
Collapse
Affiliation(s)
| | - Pablo Caviedes
- Program of Molecular & Clinical Pharmacology, ICBM, Faculty of
Medicine, Universidad de Chile, Santiago, Chile
- Centro de Biotecnología y Bioingeniería (CeBiB), Departamento de
Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y
Matemáticas, Universidad de Chile, Santiago, Chile
| | - Rodrigo Mardones
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
- Orthopedic Department, Clinica Las Condes, Santiago,
Chile
| | - José J. Minguell
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
| | - Ana Maria Vega-Letter
- Program of Traslational Immunology ICIM, Faculty of Medicine, Clinica
Alemana Universidad del Desarrollo, Santiago, Chile
| | - Claudio M. Jofre
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
- * E-mail:
| |
Collapse
|
17
|
Ramli K, Aminath Gasim I, Ahmad AA, Hassan S, Law ZK, Tan GC, Baharuddin A, Naicker AS, Htwe O, Mohammed Haflah NH, B H Idrus R, Abdullah S, Ng MH. Human bone marrow-derived MSCs spontaneously express specific Schwann cell markers. Cell Biol Int 2019; 43:233-252. [PMID: 30362196 DOI: 10.1002/cbin.11067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/07/2018] [Indexed: 12/15/2022]
Abstract
In peripheral nerve injuries, Schwann cells (SC) play pivotal roles in regenerating damaged nerve. However, the use of SC in clinical cell-based therapy is hampered due to its limited availability. In this study, we aim to evaluate the effectiveness of using an established induction protocol for human bone marrow derived-MSC (hBM-MSCs) transdifferentiation into a SC lineage. A relatively homogenous culture of hBM-MSCs was first established after serial passaging (P3), with profiles conforming to the minimal criteria set by International Society for Cellular Therapy (ISCT). The cultures (n = 3) were then subjected to a series of induction media containing β-mercaptoethanol, retinoic acid, and growth factors. Quantitative RT-PCR, flow cytometry, and immunocytochemistry analyses were performed to quantify the expression of specific SC markers, that is, S100, GFAP, MPZ and p75 NGFR, in both undifferentiated and transdifferentiated hBM-MSCs. Based on these analyses, all markers were expressed in undifferentiated hBM-MSCs and MPZ expression (mRNA transcripts) was consistently detected before and after transdifferentiation across all samples. There was upregulation at the transcript level of more than twofolds for NGF, MPB, GDNF, p75 NGFR post-transdifferentiation. This study highlights the existence of spontaneous expression of specific SC markers in cultured hBM-MSCs, inter-donor variability and that MSC transdifferentiation is a heterogenous process. These findings strongly oppose the use of a single marker to indicate SC fate. The heterogenous nature of MSC may influence the efficiency of SC transdifferentiation protocols. Therefore, there is an urgent need to re-define the MSC subpopulations and revise the minimal criteria for MSC identification.
Collapse
Affiliation(s)
- Khairunnisa Ramli
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Ifasha Aminath Gasim
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Amir Adham Ahmad
- Department of Orthopaedics, School of Medicine, International Medical University, Negeri Sembilan, Malaysia
| | - Shariful Hassan
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Zhe Kang Law
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Azmi Baharuddin
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Amaramalar Selvi Naicker
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ohnmar Htwe
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nor Hazla Mohammed Haflah
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ruszymah B H Idrus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shalimar Abdullah
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Min Hwei Ng
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
18
|
Kaneko T, Gu B, Sone PP, Zaw SYM, Murano H, Zaw ZCT, Okiji T. Dental Pulp Tissue Engineering Using Mesenchymal Stem Cells: a Review with a Protocol. Stem Cell Rev Rep 2018; 14:668-676. [PMID: 29804171 DOI: 10.1007/s12015-018-9826-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells that can be isolated from human and animal sources such as rats. Recently, an in vivo protocol for pulp tissue engineering using implantation of bone marrow MSCs into rat pulpotomized molars was established by our research group. This coronal pulp regeneration model showed almost complete regeneration/healing with dentin bridge formation when the cavity was sealed with mineral trioxide aggregate (MTA) to create a biocompatible seal of the pulp. This method is a powerful tool for elucidating the processes of dental pulp tissue regeneration following implantation of MSCs. In the present review, we discuss the literature in the field of dental pulp tissue engineering using MSCs including dental pulp stem cells and stem cells from exfoliated deciduous teeth. In addition, we present a brief step-by-step protocol of the coronal pulp regeneration model focusing on the implantation of rat bone marrow MSCs, biodegradable scaffolds, and hydrogels in pulpotomized rat molars. The protocol may lay the foundation for studies aiming at defining further histological and molecular mechanism of the rat pulp tissue engineering.
Collapse
Affiliation(s)
- Tomoatsu Kaneko
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan.
| | - Bin Gu
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Phyo Pyai Sone
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Su Yee Myo Zaw
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Hiroki Murano
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Zar Chi Thein Zaw
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| | - Takashi Okiji
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-Ku, Tokyo, 113-8549, Japan
| |
Collapse
|
19
|
Ji W, Álvarez Z, Edelbrock AN, Sato K, Stupp SI. Bioactive Nanofibers Induce Neural Transdifferentiation of Human Bone Marrow Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2018; 10:41046-41055. [PMID: 30475573 DOI: 10.1021/acsami.8b13653] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The combination of biomaterials with stem cells is a promising therapeutic strategy to repair traumatic injuries in the central nervous system, and human bone marrow mesenchymal stem cells (BMSCs) offer a clinically translatable option among other possible sources of stem cells. We report here on the use of a supramolecular bioactive material based on a peptide amphiphile (PA), displaying a laminin-mimetic IKVAV sequence to drive neural transdifferentiation of human BMSCs. The IKVAV-PA self-assembles into supramolecular nanofibers that induce neuroectodermal lineage commitment after 1 week, as evidenced by the upregulation of the neural progenitor gene nestin ( NES) and glial fibrillary acidic protein ( GFAP). After 2 weeks, the bioactive IKVAV-PA nanofibers induce significantly higher expression of neuronal markers β-III tubulin (TUJ-1), microtubule-associated protein-2 (MAP-2), and neuronal nuclei (NEUN), as well as the extracellular matrix laminin (LMN). Furthermore, the human BMSCs exposed to the biomaterial reveal a polarized cytoskeletal architecture and a decrease in cellular size, resembling neuron-like cells. We conclude that the investigated supramolecular biomaterial opens the opportunity to transdifferentiate adult human BMSCs into neuronal lineage.
Collapse
Affiliation(s)
- Wei Ji
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration , KU Leuven , Leuven 3000 , Belgium
| | | | | | | | | |
Collapse
|
20
|
Soleimani M, Mashayekhan S, Baniasadi H, Ramazani A, Ansarizadeh M. Design and fabrication of conductive nanofibrous scaffolds for neural tissue engineering: Process modeling via response surface methodology. J Biomater Appl 2018; 33:619-629. [PMID: 30388384 DOI: 10.1177/0885328218808917] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Peripheral nervous system in contrary to central one has the potential for regeneration, but its regrowth requires proper environmental conditions and supporting growth factors. The aim of this study is to design and fabricate a conductive polyaniline/graphene nanoparticles incorporated gelatin nanofibrous scaffolds suitable for peripheral nervous system regeneration. The scaffolds were fabricated with electrospinning and the fabrication process was designed with Design-Expert software via response surface methodology. The effect of process parameters including applied voltage (kV), syringe pump flow rate (cm3/h), and PAG concentration (wt%), on the scaffold conductivity, nanofibers diameter, and cell viability were investigated. The obtained results showed that the scaffold conductivity and cell viability are affected by polyaniline/graphene concentration while nanofiber diameter is more affected by the applied voltage and syringe pump flow rate. Optimum scaffold with maximum conductivity (0.031 ± 0.0013 S/cm) and cell compatibility and suitable diameter were electrospun according to the software introduced values for the process parameters (voltage of 13 kV, flow rate of 0.1 cm3/h, and PAG wt.% of 1.3) and its morphology, cell compatibility, and biodegradability were further investigated, which showed its potential for applying in peripheral nervous system injury regeneration.
Collapse
Affiliation(s)
- Maryam Soleimani
- 1 Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Shohreh Mashayekhan
- 1 Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Hossein Baniasadi
- 2 Department of Petroleum and Chemical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ahmad Ramazani
- 1 Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Mohamadhasan Ansarizadeh
- 1 Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
21
|
Oliveira Miranda C, Marcelo A, Silva TP, Barata J, Vasconcelos-Ferreira A, Pereira D, Nóbrega C, Duarte S, Barros I, Alves J, Sereno J, Petrella LI, Castelhano J, Paiva VH, Rodrigues-Santos P, Alves V, Nunes-Correia I, Nobre RJ, Gomes C, Castelo-Branco M, Pereira de Almeida L. Repeated Mesenchymal Stromal Cell Treatment Sustainably Alleviates Machado-Joseph Disease. Mol Ther 2018; 26:2131-2151. [PMID: 30087083 DOI: 10.1016/j.ymthe.2018.07.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 06/19/2018] [Accepted: 07/03/2018] [Indexed: 12/16/2022] Open
Abstract
Machado-Joseph disease (MJD) or spinocerebellar ataxia type 3, the most common dominant spinocerebellar ataxia (SCA) worldwide, is caused by over-repetition of a CAG repeat in the ATXN3/MJD1 gene, which translates into a polyglutamine tract within the ataxin-3 protein. There is no treatment for this fatal disorder. Despite evidence of the safety and efficacy of mesenchymal stromal cells (MSCs) in delaying SCA disease progression in exploratory clinical trials, unanticipated regression of patients to the status prior to treatment makes the investigation of causes and solutions urgent and imperative. In the present study, we compared the efficacy of a single intracranial injection with repeated systemic MSC administration in alleviating the MJD phenotype of two strongly severe genetic rodent models. We found that a single MSC transplantation only produces transient effects, whereas periodic administration promotes sustained motor behavior and neuropathology alleviation, suggesting that MSC therapies should be re-designed to get sustained beneficial results in clinical practice. Furthermore, MSC promoted neuroprotection, increased the levels of GABA and glutamate, and decreased the levels of Myo-inositol, which correlated with motor improvements, indicating that these metabolites may serve as valid neurospectroscopic biomarkers of disease and treatment. This study makes important contributions to the design of new clinical approaches for MJD and other SCAs/polyglutamine disorders.
Collapse
Affiliation(s)
- Catarina Oliveira Miranda
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Adriana Marcelo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Teresa Pereira Silva
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - João Barata
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Ana Vasconcelos-Ferreira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Doctoral Programme of Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Dina Pereira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Doctoral Programme in Experimental Biology and Biomedicine, CNC - University of Coimbra, Rua Larga, Faculdade de Medicina, Pólo I, 1° andar, 3004-504 Coimbra, Portugal
| | - Clévio Nóbrega
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Sónia Duarte
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Inês Barros
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Joana Alves
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - José Sereno
- Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research, Edifício do ICNAS, Polo 3, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Institute of Nuclear Science Applied to Health, University of Coimbra, Polo 3, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Lorena Itatí Petrella
- Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research, Edifício do ICNAS, Polo 3, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Institute of Nuclear Science Applied to Health, University of Coimbra, Polo 3, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - João Castelhano
- Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research, Edifício do ICNAS, Polo 3, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Institute of Nuclear Science Applied to Health, University of Coimbra, Polo 3, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Vitor Hugo Paiva
- Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal; MARE - Marine and Environmental Sciences Centre, Department of Life Sciences, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Paulo Rodrigues-Santos
- Immunology Institute, Faculty of Medicine, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal; Immunology and Oncology Laboratory, Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Rua Larga, 3004-504, Portugal; Center of Investigation in Environment, Genetics and Oncobiology, Apartado 9015, 3001-301, Coimbra, Portugal
| | - Vera Alves
- Immunology Institute, Faculty of Medicine, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal; Immunology and Oncology Laboratory, Center for Neurosciences and Cell Biology (CNC), University of Coimbra, Rua Larga, 3004-504, Portugal; Center of Investigation in Environment, Genetics and Oncobiology, Apartado 9015, 3001-301, Coimbra, Portugal
| | - Isabel Nunes-Correia
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Rui Jorge Nobre
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão - Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal; Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Célia Gomes
- Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Polo 3, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Miguel Castelo-Branco
- Centre for Neuroscience and Cell Biology - Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research, Edifício do ICNAS, Polo 3, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Institute of Nuclear Science Applied to Health, University of Coimbra, Polo 3, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Luís Pereira de Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Faculdade de Medicina, Rua Larga, Pólo I, 1° andar, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Polo 3, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
22
|
Muniswami DM, Kanthakumar P, Kanakasabapathy I, Tharion G. Motor Recovery after Transplantation of Bone Marrow Mesenchymal Stem Cells in Rat Models of Spinal Cord Injury. Ann Neurosci 2018; 25:126-140. [PMID: 30814821 DOI: 10.1159/000487069] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 12/26/2018] [Indexed: 12/25/2022] Open
Abstract
Background Neuronal tissue has a limited potential to self-renew or get repaired after damage. Cell therapies using stem cells are promising approaches for the treatment of central nervous system (CNS) injuries. However, the clinical use of embryonic stem cells is limited by ethical concerns and other scientific consequences. Bone marrow mesenchymal stromal cells (BM-MSC) could represent an alternative source of stem cells for replacement therapy. Indeed, many studies have demonstrated that MSCs can give rise to neuronal cells as well as many tissue-specific cell phenotypes. Purpose Motor recovery by transplantation of bone marrow MSCs in rat models of spinal cord injury (SCI). Methods Bone marrow was collected from the femur of albino Wistar rats. MSCs were separated using the Ficoll-Paque density gradient method and cultured in Dulbecco's Modified Eagle Medium supplemented with 20% fetal bovine serum. Cultured MSC was characterized by immunohistochemistry and flow cytometry and neuronal-induced cells were further characterized for neural markers. Cultured MSCs were transplanted into the experimentally injured spinal cord of Wistar rats. Control (injured, but without cell transplantation) and transplanted rats were followed up to 8 weeks, analyzed using the Basso, Beattie, Bresnahan (BBB) scale and electromyography (EMG) for behavioral and physiological status of the injured spinal cord. Finally, the tissue was evaluated histologically. Results Rat MSCs expressed positivity for a panel of MSC markers CD29, CD54, CD90, CD73, and CD105, and negativity for hematopoietic markers CD34, CD14, and CD45. In vitro neuronal transdifferentiated MSCs express positivity for β III tubulin, MAP2, NF, NeuN, Nav1.1, oligodendrocyte (O4), and negativity for glial fibrillary acid protein. All the treated groups show promising hind-limb motor recovery BBB score, except the control group. There was increased EMG amplitude in treated groups as compared to the control group. Green fluorescent protein (GFP)-labeled MSC survived and differentiated into neurons in the injured spinal cord, which is responsible for functional recovery. Conclusion Our results demonstrate that BM-MSC has the potential to repair the injured cord in rat models of SCI. Thus, BM-MSC appears to be a promising candidate for cell-based therapy in CNS injury.
Collapse
Affiliation(s)
- Durai Murugan Muniswami
- Department of Physical Medicine and Rehabilitation, Christian Medical College, Vellore, India
| | | | | | - George Tharion
- Department of Physical Medicine and Rehabilitation, Christian Medical College, Vellore, India
| |
Collapse
|
23
|
George S, Hamblin MR, Abrahamse H. Current and Future Trends in Adipose Stem Cell Differentiation into Neuroglia. Photomed Laser Surg 2018; 36:230-240. [PMID: 29570423 DOI: 10.1089/pho.2017.4411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Neurological diseases and disorders pose a challenge for treatment and rehabilitation due to the limited capacity of the nervous system to repair itself. Adipose stem cells (ASCs) are more pliable than any adult stem cells and are capable of differentiating into non-mesodermal tissues, including neurons. Transdifferentiating ASCs to specific neuronal lineage cells enables us to deliver the right type of cells required for a replacement therapy into the nervous system. METHODS Several methodologies are being explored and tested to differentiate ASCs to functional neurons and glia with cellular factors and chemical compounds. However, none of these processes and prototypes has been wholly successful in changing the cellular structure and functional status of ASCs to become identical to neuroglial cells. In addition, successful integration and functional competence of these cells for use in clinical applications remain problematic. Photobiomodulation or low-level laser irradiation has been successfully applied to not only improve ASC viability and proliferation but has also shown promise as a possible enhancer of ASC differentiation. CONCLUSIONS Studies have shown that photobiomodulation improves the use of stem cell transplantation for neurological applications. This review investigates current neuro-differentiation inducers and suitable methodologies, including photobiomodulation, utilizing ASCs for induction of differentiation into neuronal lineages.
Collapse
Affiliation(s)
- Sajan George
- 1 Laser Research Centre, Faculty of Health Sciences, University of Johannesburg , Doornfontein, South Africa
| | - Michael R Hamblin
- 2 Wellman Centre for Photomedicine, Massachusetts General Hospital , Boston, Massachusetts.,3 Department of Dermatology, Harvard Medical School , Boston, Massachusetts.,4 Harvard-MIT Division of Health Sciences and Technology , Cambridge, Massachusetts
| | - Heidi Abrahamse
- 1 Laser Research Centre, Faculty of Health Sciences, University of Johannesburg , Doornfontein, South Africa
| |
Collapse
|
24
|
Srivastava A, Singh S, Pandey A, Kumar D, Rajpurohit CS, Khanna VK, Pant AB. Secretome of Differentiated PC12 Cells Enhances Neuronal Differentiation in Human Mesenchymal Stem Cells Via NGF-Like Mechanism. Mol Neurobiol 2018. [PMID: 29527653 DOI: 10.1007/s12035-018-0981-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The secretome-mediated responses over cellular physiology are well documented. Stem cells have been ruling the field of secretomics and its role in regenerative medicine since the past few years. However, the mechanistic aspects of secretome-mediated responses and the role of other cells in this area remain somewhat elusive. Here, we investigate the effects of secretome-enriched conditioned medium (CM) of neuronally differentiated PC12 cells on the neuronal differentiation of human mesenchymal stem cells (hMSCs). The exposure to CM at a ratio of 1:1 (CM: conditioned medium of PC12 cells) led to neuronal induction in hMSCs. This neuronal induction was compared with a parallel group of cells exposed to nerve growth factor (NGF). There was a marked increase in neurite length and expression of neuronal markers (β-III tubulin, neurofilament-M (NF-M), synaptophysin, NeuN in exposed hMSCs). Experimental group co-exposed to NGF and CM showed an additive response via MAPK signaling and directed the cells particularly towards cholinergic lineage. The ability of CM to enhance the neuronal properties of stem cells could aid in their rapid differentiation into neuronal subtypes in case of stem cell transplantation for neuronal injuries, thus broadening the scope of non-stem cell-based applications in the area of secretomics.
Collapse
Affiliation(s)
- A Srivastava
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India
| | - S Singh
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - A Pandey
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India
| | - D Kumar
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - C S Rajpurohit
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - V K Khanna
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India.,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India
| | - A B Pant
- System Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg, Lucknow, Uttar Pradesh, -226001, India. .,Academy of Scientific and Innovative Research, CSIR-IITR Campus, Lucknow, India.
| |
Collapse
|
25
|
Weiss JN, Levy S, Benes SC. Stem Cell Ophthalmology Treatment Study: bone marrow derived stem cells in the treatment of non-arteritic ischemic optic neuropathy (NAION). Stem Cell Investig 2017; 4:94. [PMID: 29270420 DOI: 10.21037/sci.2017.11.05] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/13/2017] [Indexed: 12/17/2022]
Abstract
Background Ten patients with bilateral visual loss due to sequential non-arteritic ischemic optic neuropathy (NAION) underwent autologous Bone Marrow Derived Stem Cell (BMSC) therapy within the Stem Cell Ophthalmology Treatment Study (SCOTS). SCOTS is an Institutional Review Board approved clinical study utilizing autologous BMSC in the treatment of optic nerve and retinal diseases that meet inclusion criteria. Methods The average age of the patients treated was 69.8 years. The average duration of visual loss in eyes treated was 9.8 years and ranged from 1 to 35 years. Affected eyes were treated with either retrobulbar, subtenons and intravenous BMSC or, following vitrectomy, intra-optic nerve, subtenons and intravenous BMSC. The primary outcome was visual acuity as measured by Snellen or converted to LogMAR. Results Following therapy in SCOTS, 80% of patients experienced improvement in Snellen binocular vision (P=0.029) with 20% remaining stable; 73.6% of eyes treated gained vision (P=0.019) and 15.9% remained stable in the post-operative period. There was an average of 3.53 Snellen lines of vision improvement per eye with an average 22.74% and maximum 83.3% improvement in LogMAR acuity per eye. The average LogMAR change in treated eyes was a gain of 0.364 (P=0.0089). Improvements typically manifested no later than 6 months post procedure. Conclusions The use of BMSC in the Stem Cell Ophthalmology Treatment Study achieved meaningful visual improvements in a significant percentage of the NAION patients reported. Improvements typically manifested no later than 6 months post-procedure. Duration of visual loss did not appear to affect the ability of the eyes to respond to treatment. Possible mechanisms by which visual improvement occurred may include BMSC paracrine secretion of proteins and hormones, transfer of mitochondria, release of messenger RNA or other compounds via exosomes or microvesicles and neuronal transdifferentiation of the stem cells.
Collapse
Affiliation(s)
- Jeffrey N Weiss
- The Healing Institute, 1308 North State Road 7, Margate, FL 33063, USA
| | - Steven Levy
- MD Stem Cells, 3 Sylvan Road South, Westport, CT 06880, USA
| | - Susan C Benes
- The Eye Center of Columbus, 9262 Neil Avenue, The Ohio State University, Columbus, OH 43205, USA
| |
Collapse
|
26
|
Yu C, Griffiths LR, Haupt LM. Exploiting Heparan Sulfate Proteoglycans in Human Neurogenesis-Controlling Lineage Specification and Fate. Front Integr Neurosci 2017; 11:28. [PMID: 29089873 PMCID: PMC5650988 DOI: 10.3389/fnint.2017.00028] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 09/25/2017] [Indexed: 12/26/2022] Open
Abstract
Unspecialized, self-renewing stem cells have extraordinary application to regenerative medicine due to their multilineage differentiation potential. Stem cell therapies through replenishing damaged or lost cells in the injured area is an attractive treatment of brain trauma and neurodegenerative neurological disorders. Several stem cell types have neurogenic potential including neural stem cells (NSCs), embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs). Currently, effective use of these cells is limited by our lack of understanding and ability to direct lineage commitment and differentiation of neural lineages. Heparan sulfate proteoglycans (HSPGs) are ubiquitous proteins within the stem cell microenvironment or niche and are found localized on the cell surface and in the extracellular matrix (ECM), where they interact with numerous signaling molecules. The glycosaminoglycan (GAG) chains carried by HSPGs are heterogeneous carbohydrates comprised of repeating disaccharides with specific sulfation patterns that govern ligand interactions to numerous factors including the fibroblast growth factors (FGFs) and wingless-type MMTV integration site family (Wnts). As such, HSPGs are plausible targets for guiding and controlling neural stem cell lineage fate. In this review, we provide an overview of HSPG family members syndecans and glypicans, and perlecan and their role in neurogenesis. We summarize the structural changes and subsequent functional implications of heparan sulfate as cells undergo neural lineage differentiation as well as outline the role of HSPG core protein expression throughout mammalian neural development and their function as cell receptors and co-receptors. Finally, we highlight suitable biomimetic approaches for exploiting the role of HSPGs in mammalian neurogenesis to control and tailor cell differentiation into specific lineages. An improved ability to control stem cell specific neural lineage fate and produce abundant cells of lineage specificity will further advance stem cell therapy for the development of improved repair of neurological disorders. We propose a deeper understanding of HSPG-mediated neurogenesis can potentially provide novel therapeutic targets of neurogenesis.
Collapse
Affiliation(s)
- Chieh Yu
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Larisa M Haupt
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
27
|
Human bone marrow harbors cells with neural crest-associated characteristics like human adipose and dermis tissues. PLoS One 2017; 12:e0177962. [PMID: 28683107 PMCID: PMC5500284 DOI: 10.1371/journal.pone.0177962] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 05/05/2017] [Indexed: 12/13/2022] Open
Abstract
Adult neural crest stem-derived cells (NCSC) are of extraordinary high plasticity and promising candidates for use in regenerative medicine. Several locations such as skin, adipose tissue, dental pulp or bone marrow have been described in rodent, as sources of NCSC. However, very little information is available concerning their correspondence in human tissues, and more precisely for human bone marrow. The main objective of this study was therefore to characterize NCSC from adult human bone marrow. In this purpose, we compared human bone marrow stromal cells to human adipose tissue and dermis, already described for containing NCSC. We performed comparative analyses in terms of gene and protein expression as well as functional characterizations. It appeared that human bone marrow, similarly to adipose tissue and dermis, contains NESTIN+ / SOX9+ / TWIST+ / SLUG+ / P75NTR+/ BRN3A+/ MSI1+/ SNAIL1+ cells and were able to differentiate into melanocytes, Schwann cells and neurons. Moreover, when injected into chicken embryos, all those cells were able to migrate and follow endogenous neural crest migration pathways. Altogether, the phenotypic characterization and migration abilities strongly suggest the presence of neural crest-derived cells in human adult bone marrow.
Collapse
|
28
|
Song XQ, Su LN, Wei HP, Liu YH, Yin HF. Protective Effects of Hydroxysafflor Yellow A against Oxidative Damage of β-Mercaptoethanol During Neural Differentiation of Mesenchymal Stem Cells. CHINESE HERBAL MEDICINES 2017. [DOI: 10.1016/s1674-6384(17)60105-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
29
|
Singh SP, Jadhav SH, Chaturvedi CP, Nityanand S. Therapeutic efficacy of multipotent adult progenitor cells versus mesenchymal stem cells in experimental autoimmune encephalomyelitis. Regen Med 2017. [PMID: 28621170 DOI: 10.2217/rme-2016-0109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIM In this study, we have evaluated the therapeutic efficacy of mouse multipotent adult progenitor cells (mMAPCs) in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, and compared it with mouse mesenchymal stem cells (mMSCs). MATERIALS & METHODS We administered PKH26-labeled mMAPC and mMSC into EAE mice and evaluated their therapeutic efficacy. RESULTS The mMAPC-treated mice in comparison with the mMSC group exhibited a higher suppression of EAE (p < 0.05), and a higher fold expression of neuronal genes GAP43, NG2, PDGFR, Nestin, SMI 32, BDNF and NT 3 in spinal cord (p < 0.05), suggesting a better neuroprotective and regenerative potential of mMAPC than mMSC. CONCLUSION MAPC may be a potential cell type, which is superior to mesenchymal stem cell for the treatment of EAE/multiple sclerosis.
Collapse
Affiliation(s)
- Saurabh Pratap Singh
- Stem Cell Research Facility, Department of Hematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow-226014, India
| | - Sachin Hanumantrao Jadhav
- Stem Cell Research Facility, Department of Hematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow-226014, India
| | - Chandra Prakash Chaturvedi
- Stem Cell Research Facility, Department of Hematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow-226014, India
| | - Soniya Nityanand
- Stem Cell Research Facility, Department of Hematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow-226014, India
| |
Collapse
|
30
|
Li D, Secher JO, Juhl M, Mashayekhi K, Nielsen TT, Holst B, Hyttel P, Freude KK, Hall VJ. Identification of SSEA-1 expressing enhanced reprogramming (SEER) cells in porcine embryonic fibroblasts. Cell Cycle 2017; 16:1070-1084. [PMID: 28426281 DOI: 10.1080/15384101.2017.1315490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Previous research has shown that a subpopulation of cells within cultured human dermal fibroblasts, termed multilineage-differentiating stress enduring (Muse) cells, are preferentially reprogrammed into induced pluripotent stem cells. However, controversy exists over whether these cells are the only cells capable of being reprogrammed from a heterogeneous population of fibroblasts. Similarly, there is little research to suggest such cells may exist in embryonic tissues or other species. To address if such a cell population exists in pigs, we investigated porcine embryonic fibroblast populations (pEFs) and identified heterogeneous expression of several key cell surface markers. Strikingly, we discovered a small population of stage-specific embryonic antigen 1 positive cells (SSEA-1+) in Danish Landrace and Göttingen minipig pEFs, which were absent in the Yucatan pEFs. Furthermore, reprogramming of SSEA-1+ sorted pEFs led to higher reprogramming efficiency. Subsequent transcriptome profiling of the SSEA-1+ vs. the SSEA-1neg cell fraction revealed highly comparable gene signatures. However several genes that were found to be upregulated in the SSEA-1+ cells were similarly expressed in mesenchymal stem cells (MSCs). We therefore termed these cells SSEA-1 Expressing Enhanced Reprogramming (SEER) cells. Interestingly, SEER cells were more effective at differentiating into osteocytes and chondrocytes in vitro. We conclude that SEER cells are more amenable for reprogramming and that the expression of mesenchymal stem cell genes is advantageous in the reprogramming process. This data provides evidence supporting the elite theory and helps to delineate which cell types and specific genes are important for reprogramming in the pig.
Collapse
Affiliation(s)
- Dong Li
- a Department of Veterinary and Animal Sciences , Faculty of Health and Medical Sciences, University of Copenhagen , Denmark
| | - Jan O Secher
- a Department of Veterinary and Animal Sciences , Faculty of Health and Medical Sciences, University of Copenhagen , Denmark
| | - Morten Juhl
- b Cardiology Stem Cell Centre , The Heart Centre, Copenhagen University Hospital , Copenhagen , Denmark
| | - Kaveh Mashayekhi
- a Department of Veterinary and Animal Sciences , Faculty of Health and Medical Sciences, University of Copenhagen , Denmark.,c BioTalentum Ltd. , Gödöllő , Hungary.,d Chief Scientific Officer , Sandor Life Sciences, Sandor Medicaids Group Pvt. Ltd. , Hyderabad , India
| | - Troels T Nielsen
- e Danish Dementia Research Centre , Copenhagen University Hospital , Copenhagen , Denmark
| | | | - Poul Hyttel
- a Department of Veterinary and Animal Sciences , Faculty of Health and Medical Sciences, University of Copenhagen , Denmark
| | - Kristine K Freude
- a Department of Veterinary and Animal Sciences , Faculty of Health and Medical Sciences, University of Copenhagen , Denmark
| | - Vanessa J Hall
- a Department of Veterinary and Animal Sciences , Faculty of Health and Medical Sciences, University of Copenhagen , Denmark
| |
Collapse
|
31
|
Redondo-Castro E, Cunningham C, Miller J, Martuscelli L, Aoulad-Ali S, Rothwell NJ, Kielty CM, Allan SM, Pinteaux E. Interleukin-1 primes human mesenchymal stem cells towards an anti-inflammatory and pro-trophic phenotype in vitro. Stem Cell Res Ther 2017; 8:79. [PMID: 28412968 PMCID: PMC5393041 DOI: 10.1186/s13287-017-0531-4] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/01/2017] [Accepted: 03/08/2017] [Indexed: 12/25/2022] Open
Abstract
Background Inflammation is a key contributor to central nervous system (CNS) injury such as stroke, and is a major target for therapeutic intervention. Effective treatments for CNS injuries are limited and applicable to only a minority of patients. Stem cell-based therapies are increasingly considered for the treatment of CNS disease, because they can be used as in-situ regulators of inflammation, and improve tissue repair and recovery. One promising option is the use of bone marrow-derived mesenchymal stem cells (MSCs), which can secrete anti-inflammatory and trophic factors, can migrate towards inflamed and injured sites or can be implanted locally. Here we tested the hypothesis that pre-treatment with inflammatory cytokines can prime MSCs towards an anti-inflammatory and pro-trophic phenotype in vitro. Methods Human MSCs from three different donors were cultured in vitro and treated with inflammatory mediators as follows: interleukin (IL)-1α, IL-1β, tumour necrosis factor alpha (TNF-α) or interferon-γ. After 24 h of treatment, cell supernatants were analysed by ELISA for expression of granulocyte-colony stimulating factor (G-CSF), IL-10, brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), IL-1 receptor antagonist (IL-1Ra) and vascular endothelial growth factor (VEGF). To confirm the anti-inflammatory potential of MSCs, immortalised mouse microglial BV2 cells were treated with bacterial lipopolysaccharide (LPS) and exposed to conditioned media (CM) of naïve or IL-1-primed MSCs, and levels of secreted microglial-derived inflammatory mediators including TNF-α, IL-10, G-CSF and IL-6 were measured by ELISA. Results Unstimulated MSCs constitutively expressed anti-inflammatory cytokines and trophic factors (IL-10, VEGF, BDNF, G-CSF, NGF and IL-1Ra). MSCs primed with IL-1α or IL-1β showed increased secretion of G-CSF, which was blocked by IL-1Ra. Furthermore, LPS-treated BV2 cells secreted less inflammatory and apoptotic markers, and showed increased secretion of the anti-inflammatory IL-10 in response to treatment with CM of IL-1-primed MSCs compared with CM of unprimed MSCs. Conclusions Our results demonstrate that priming MSCs with IL-1 increases expression of trophic factor G-CSF through an IL-1 receptor type 1 (IL-1R1) mechanism, and induces a reduction in the secretion of inflammatory mediators in LPS-activated microglial cells. The results therefore support the potential use of preconditioning treatments of stem cells in future therapies. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0531-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elena Redondo-Castro
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Catriona Cunningham
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jonjo Miller
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Licia Martuscelli
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sarah Aoulad-Ali
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Nancy J Rothwell
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Cay M Kielty
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
32
|
Sueyama Y, Kaneko T, Ito T, Okiji T. Effect of lipopolysaccharide stimulation on stem cell-associated marker-expressing cells. Int Endod J 2017; 51 Suppl 2:e107-e114. [DOI: 10.1111/iej.12740] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 12/08/2016] [Indexed: 01/05/2023]
Affiliation(s)
- Y. Sueyama
- Division of Cariology; Operative Dentistry and Endodontics; Niigata University Graduate School of Medical and Dental Sciences; Niigata Japan
| | - T. Kaneko
- Pulp Biology and Endodontics; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| | - T. Ito
- Division of Cariology; Operative Dentistry and Endodontics; Niigata University Graduate School of Medical and Dental Sciences; Niigata Japan
| | - T. Okiji
- Pulp Biology and Endodontics; Graduate School; Tokyo Medical and Dental University; Tokyo Japan
| |
Collapse
|
33
|
Lv B, Li F, Fang J, Xu L, Sun C, Han J, Hua T, Zhang Z, Feng Z, Wang Q, Jiang X. Activated Microglia Induce Bone Marrow Mesenchymal Stem Cells to Produce Glial Cell-Derived Neurotrophic Factor and Protect Neurons Against Oxygen-Glucose Deprivation Injury. Front Cell Neurosci 2016; 10:283. [PMID: 28018176 PMCID: PMC5160383 DOI: 10.3389/fncel.2016.00283] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/28/2016] [Indexed: 12/16/2022] Open
Abstract
In this study, we investigated interactions among microglia (MG), bone marrow mesenchymal stem cells (BMSCs) and neurons in cerebral ischemia and the potential mechanisms using an in vitro oxygen-glucose deprivation (OGD) model. Rat BMSCs were incubated with conditioned medium (CM) from in vitro cultures of OGD-activated rat MG and murine BV2 MG cells. Effects of glial cell-derived neurotrophic factor (GDNF) on rat neuron viability, apoptosis, lactate dehydrogenase (LDH) leakage and mitochondrial membrane potential (MMP) were analyzed in this model. OGD-activated MG promoted GDNF production by BMSCs (P < 0.01). Tumor necrosis factor-α (TNFα), but not interleukin-6 (IL6) or interleukin 1β (IL1β), promoted GDNF production by BMSCs (P < 0.001). GDNF or CM pre-treated BMSCs elevated neuronal viability and suppressed apoptosis (P < 0.05 or P < 0.01); these effects were inhibited by the RET antibody. GDNF activated MEK/ERK and phosphoinositide-3-kinase (PI3K)/AKT signaling but not JNK/c-JUN. Furthermore, GDNF upregulated B cell lymphoma 2 (BCL2) and heat shock 60 kDa protein 1 (HSP60) levels, suppressed LDH leakage, and promoted MMP. Thus, activated MG produce TNFα to stimulate GDNF production by BMSCs, which prevents and repairs OGD-induced neuronal injury, possibly via regulating MEK/ERK and PI3K/AKT signaling. These findings will facilitate the prevention and treatment of neuronal injury by cerebral ischemia.
Collapse
Affiliation(s)
- Bingke Lv
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Feng Li
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Jie Fang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Limin Xu
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Chengmei Sun
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Jianbang Han
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Tian Hua
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Zhongfei Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Zhiming Feng
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Qinghua Wang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| | - Xiaodan Jiang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration Guangzhou, China
| |
Collapse
|
34
|
G-CSF-mobilized Bone Marrow Mesenchymal Stem Cells Replenish Neural Lineages in Alzheimer's Disease Mice via CXCR4/SDF-1 Chemotaxis. Mol Neurobiol 2016; 54:6198-6212. [PMID: 27709493 DOI: 10.1007/s12035-016-0122-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 09/12/2016] [Indexed: 10/20/2022]
Abstract
Recent studies reported granulocyte colony-stimulating factor (G-CSF) treatment can improve the cognitive function of Alzheimer's disease (AD) mice, and the mobilized hematopoietic stem cells (HSCs) or bone marrow mesenchymal stem cells (BM-MSCs) are proposed to be involved in this recovery effect. However, the exact role of mobilized HSC/BM-MSC in G-CSF-based therapeutic effects is still unknown. Here, we report that C-X-C chemokine receptor type 4 (CXCR4)/stromal cell-derived factor 1 (SDF-1) chemotaxis was a key mediator in G-CSF-based therapeutic effects, which was involved in the recruitment of repair-competent cells. Furthermore, we found both mobilized HSCs and BM-MSCs were able to infiltrate into the brain, but only BM-MSCs replenished the neural lineage cells and contributed to neurogenesis in the brains of AD mice. Together, our data show that mobilized BM-MSCs are involved in the replenishment of neural lineages following G-CSF treatment via CXCR4/SDF-1 chemotaxis and further support the potential use of BM-MSCs for further autogenically therapeutic applications.
Collapse
|
35
|
Mediano DR, Sanz-Rubio D, Bolea R, Marín B, Vázquez FJ, Remacha AR, López-Pérez Ó, Fernández-Borges N, Castilla J, Zaragoza P, Badiola JJ, Rodellar C, Martín-Burriel I. Characterization of mesenchymal stem cells in sheep naturally infected with scrapie. J Gen Virol 2016; 96:3715-3726. [PMID: 26431976 DOI: 10.1099/jgv.0.000292] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be infected with prions and have been proposed as in vitro cell-based models for prion replication. In addition, autologous MSCs are of interest for cell therapy in neurodegenerative diseases. To the best of our knowledge, the effect of prion diseases on the characteristics of these cells has never been investigated. Here, we analysed the properties of MSCs obtained from bone marrow (BM-MSCs) and peripheral blood (PB-MSCs) of sheep naturally infected with scrapie — a large mammal model for the study of prion diseases. After three passages of expansion, MSCs derived from scrapie animals displayed similar adipogenic, chondrogenic and osteogenic differentiation ability as cells from healthy controls, although a subtle decrease in the proliferation potential was observed. Exceptionally, mesenchymal markers such as CD29 were significantly upregulated at the transcript level compared with controls. Scrapie MSCs were able to transdifferentiate into neuron-like cells, but displayed lower levels of neurogenic markers at basal conditions, which could limit this potential .The expression levels of cellular prion protein (PrPC) were highly variable between cultures, and no significant differences were observed between control and scrapie-derived MSCs. However, during neurogenic differentiation the expression of PrPC was upregulated in MSCs. This characteristic could be useful for developing in vitro models for prion replication. Despite the infectivity reported for MSCs obtained from scrapie-infected mice and Creutzfeldt–Jakob disease patients, protein misfolding cyclic amplification did not detect PrPSc in BM- or PB-MSCs from scrapie-infected sheep, which limits their use for in vivo diagnosis for scrapie.
Collapse
Affiliation(s)
- Diego R Mediano
- Laboratorio de Genética Bioquímica, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - David Sanz-Rubio
- Laboratorio de Genética Bioquímica, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Rosa Bolea
- Centro de Investigación en Encefalopatías y Enfermedades Transmisibles Emergentes, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Belén Marín
- Centro de Investigación en Encefalopatías y Enfermedades Transmisibles Emergentes, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Francisco J Vázquez
- Laboratorio de Genética Bioquímica, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Ana R Remacha
- Laboratorio de Genética Bioquímica, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Óscar López-Pérez
- Laboratorio de Genética Bioquímica, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain.,Centro de Investigación en Encefalopatías y Enfermedades Transmisibles Emergentes, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | | | - Joaquín Castilla
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, Spain.,IKERBasque, Basque Foundation for Science, Bilbao, Spain
| | - Pilar Zaragoza
- Laboratorio de Genética Bioquímica, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Juan J Badiola
- Centro de Investigación en Encefalopatías y Enfermedades Transmisibles Emergentes, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Clementina Rodellar
- Laboratorio de Genética Bioquímica, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Inmaculada Martín-Burriel
- Centro de Investigación en Encefalopatías y Enfermedades Transmisibles Emergentes, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain.,Laboratorio de Genética Bioquímica, Instituto de Investigación Agroalimentaria (IA2), IIS Aragón, Universidad de Zaragoza, Zaragoza, Spain
| |
Collapse
|
36
|
Kim DY, Lee J, Kang D, Lee DH, Kim YJ, Hwang SG, Kim DI, Lee CW, Lee KH. Multipotent neurogenic fate of mesenchymal stem cell is determined by Cdk4-mediated hypophosphorylation of Smad-STAT3. Cell Cycle 2016; 15:1787-95. [PMID: 27192561 DOI: 10.1080/15384101.2016.1188230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Cyclin-dependent kinase (Cdk) in complex with a corresponding cyclin plays a pivotal role in neurogenic differentiation. In particular, Cdk4 activity acts as a signaling switch to direct human mesenchymal stem cells (MSCs) to neural transdifferentiation. However, the molecular evidence of how Cdk4 activity converts MSCs to neurogenic lineage remains unknown. Here, we found that Cdk4 inhibition in human MSCs enriches the populations of neural stem and progenitor pools rather than differentiated glial and neuronal cell pools. Interestingly, Cdk4 inhibition directly inactivates Smads and subsequently STAT3 signaling by hypophosphorylation, and both Cdk4 and Smads levels are linked during the processes of neural transdifferentiation and differentiation. In summary, our results provide novel molecular evidence in which Cdk4 inhibition leads to directing human MSCs to a multipotent neurogenic fate by inactivating Smads-STAT3 signaling.
Collapse
Affiliation(s)
- Dong-Young Kim
- a Research Core Facility, Center for Molecular Medicine, Samsung Biomedical Research Institute , Seoul , Republic of Korea
| | - Janet Lee
- b Department of Molecular Cell Biology , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea
| | - Dongrim Kang
- b Department of Molecular Cell Biology , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea
| | - Do-Hyeong Lee
- b Department of Molecular Cell Biology , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea
| | - Yoon-Ja Kim
- a Research Core Facility, Center for Molecular Medicine, Samsung Biomedical Research Institute , Seoul , Republic of Korea
| | - Sang-Gu Hwang
- c Division of Radiation Cancer Biology , Korea Institute of Radiological & Medical Sciences , Seoul , Republic of Korea
| | - Dong-Ik Kim
- d Department of Vascular Surgery , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea.,e Samsung Seoul Hospital , Seoul , Republic of Korea
| | - Chang-Woo Lee
- a Research Core Facility, Center for Molecular Medicine, Samsung Biomedical Research Institute , Seoul , Republic of Korea.,b Department of Molecular Cell Biology , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea
| | - Kyung-Hoon Lee
- a Research Core Facility, Center for Molecular Medicine, Samsung Biomedical Research Institute , Seoul , Republic of Korea.,f Department of Anatomy , Sungkyunkwan University School of Medicine , Suwon, Gyeonggi , Republic of Korea
| |
Collapse
|
37
|
Katsiani E, Garas A, Skentou C, Tsezou A, Messini CI, Dafopoulos K, Daponte A, Messinis IE. Chorionic villi derived mesenchymal like stem cells and expression of embryonic stem cells markers during long-term culturing. Cell Tissue Bank 2016; 17:517-29. [PMID: 27139894 DOI: 10.1007/s10561-016-9559-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 04/26/2016] [Indexed: 01/28/2023]
Abstract
Mesenchymal stem cells (MSCs) can be obtained from a variety of human tissues. MSCs derived from placental chorionic villi of the first trimester are likely to resemble, biologically, embryonic stem cells (ESC), due to the earlier development stage of placenta. In the present study long-term cultures of MSC-like cells were assessed in order to evaluate MSCs multipotent characteristics and molecular features during the period of culture. CV-cells obtained from 10 samples of chorionic villus displayed typical fibroblastoid morphology, undergone 20 passages during a period of 120 days, maintaining a stable karyotype throughout long term expansion. The cells were positive, for CD90, CD73, CD105, CD29, CD44, HLA ABC antigens and negative for CD14, CD34, AC133, and HLA DR antigens as resulted from the flow cytometry analysis. CV-cells were differentiated in adipocytes, osteoblasts, chondrocytes and neuronal cells under specific culture conditions. The expression of the ESC-gene markers POU5F1 (Oct-4) and NANOG was observed at earliest stages (4-12 passages) and not at the late stages (14-20 passages) by RT-PCR analysis. ZFP42 and SOX2 expression were not detected. Moreover, CV-cells were found to express GATA4 but not NES (Nestin). Chorionic villi-derived cells possess multipotent properties, display high proliferation rate and self-renew capacity, share common surface antigens with adult MSCs and express certain embryonics stem cells gene markers. These characteristics highlight chorionic villi as an attractive source of MSCs for the needs of regenerative medicine.
Collapse
Affiliation(s)
- E Katsiani
- Department of Obstetrics and Gynaecology, Medical School, University Hospital, University of Thessaly, Larissa, Greece
| | - A Garas
- Department of Obstetrics and Gynaecology, Medical School, University Hospital, University of Thessaly, Larissa, Greece
| | - C Skentou
- Department of Obstetrics and Gynaecology, Medical School, University Hospital, University of Thessaly, Larissa, Greece
| | - A Tsezou
- Department of Biology and Laboratory of Cytogenetics and Molecular Genetics, Medical School, University Hospital, University of Thessaly, Larissa, Greece
| | - C I Messini
- Department of Obstetrics and Gynaecology, Medical School, University Hospital, University of Thessaly, Larissa, Greece
| | - K Dafopoulos
- Department of Obstetrics and Gynaecology, Medical School, University Hospital, University of Thessaly, Larissa, Greece
| | - A Daponte
- Department of Obstetrics and Gynaecology, Medical School, University Hospital, University of Thessaly, Larissa, Greece
| | - I E Messinis
- Department of Obstetrics and Gynaecology, Medical School, University Hospital, University of Thessaly, Larissa, Greece. .,Department of Obstetrics and Gynaecology, School of Health Sciences, Faculty of Medicine, University of Thessaly, 41110, Viopolis, Larissa, Greece.
| |
Collapse
|
38
|
Weiss JN, Levy S, Benes SC. Stem Cell Ophthalmology Treatment Study (SCOTS): bone marrow-derived stem cells in the treatment of Leber's hereditary optic neuropathy. Neural Regen Res 2016; 11:1685-1694. [PMID: 27904503 PMCID: PMC5116851 DOI: 10.4103/1673-5374.193251] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Stem Cell Ophthalmology Treatment Study (SCOTS) is currently the largest-scale stem cell ophthalmology trial registered at ClinicalTrials.gov (identifier: NCT01920867). SCOTS utilizes autologous bone marrow-derived stem cells (BMSCs) to treat optic nerve and retinal diseases. Treatment approaches include a combination of retrobulbar, subtenon, intravitreal, intra-optic nerve, subretinal, and intravenous injection of autologous BMSCs according to the nature of the disease, the degree of visual loss, and any risk factors related to the treatments. Patients with Leber's hereditary optic neuropathy had visual acuity gains on the Early Treatment Diabetic Retinopathy Study (ETDRS) of up to 35 letters and Snellen acuity improvements from hand motion to 20/200 and from counting fingers to 20/100. Visual field improvements were noted. Macular and optic nerve head nerve fiber layer typically thickened. No serious complications were seen. The increases in visual acuity obtained in our study were encouraging and suggest that the use of autologous BMSCs as provided in SCOTS for ophthalmologic mitochondrial diseases including Leber's hereditary optic neuropathy may be a viable treatment option.
Collapse
Affiliation(s)
- Jeffrey N Weiss
- Retina Associates of South Florida, 5800 Colonial Drive, Suite 300, Margate, FL, USA
| | - Steven Levy
- MD Stem Cells, 3 Sylvan Road South, Westport, CT, USA
| | - Susan C Benes
- The Eye Center of Columbus, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
39
|
Chromatographically isolated CD63+CD81+ extracellular vesicles from mesenchymal stromal cells rescue cognitive impairments after TBI. Proc Natl Acad Sci U S A 2015; 113:170-5. [PMID: 26699510 DOI: 10.1073/pnas.1522297113] [Citation(s) in RCA: 350] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) secreted by cells present an attractive strategy for developing new therapies, but progress in the field is limited by several issues: The quality of the EVs varies with the type and physiological status of the producer cells; protocols used to isolate the EVs are difficult to scale up; and assays for efficacy are difficult to develop. In the present report, we have addressed these issues by using human mesenchymal stem/stromal cells (MSCs) that produce EVs when incubated in a protein-free medium, preselecting the preparations of MSCs with a biomarker for their potency in modulating inflammation, incubating the cells in a chemically defined protein-free medium that provided a stable environment, isolating the EVs with a scalable chromatographic procedure, and developing an in vivo assay for efficacy of the cells in suppressing neuroinflammation after traumatic brain injury (TBI) in mice. In addition, we demonstrate that i.v. infusion of the isolated EVs shortly after induction of TBI rescued pattern separation and spatial learning impairments 1 mo later.
Collapse
|
40
|
Stem cells in canine spinal cord injury--promise for regenerative therapy in a large animal model of human disease. Stem Cell Rev Rep 2015; 11:180-93. [PMID: 25173879 DOI: 10.1007/s12015-014-9553-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The use of cell transplantation for spinal cord injury is a rapidly evolving field in regenerative medicine. Numerous animal models are currently being used. However, translation to human patients is still a challenging step. Dogs are of increasing importance as a translational model for human disease since there is a greater awareness of the need to increase the quality of preclinical data. The use of dogs ultimately brings benefit to both human and veterinary medicine. In this review we analyze experimental and clinical studies using cell transplantation for canine spinal cord injury. Overall, in experimental studies, transplantation groups showed improvement over control groups. Improvements were measured at the functional, electrophysiological, histological, RNA and protein levels. Most clinical studies support beneficial effects of cell transplantation despite the fact that methodological limitations preclude definitive conclusions. However, the mechanisms of action and underlying the behavior of transplanted cells in the injured spinal cord remain unclear. Overall, we conclude here that stem cell interventions are a promising avenue for the treatment of spinal cord injury. Canines are a promising model that may help bridge the gap between translational research and human clinical trials.
Collapse
|
41
|
PERIPHERAL NERVE REGENERATION: CELL THERAPY AND NEUROTROPHIC FACTORS. Rev Bras Ortop 2015; 46:643-9. [PMID: 27027067 PMCID: PMC4799329 DOI: 10.1016/s2255-4971(15)30319-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 06/16/2011] [Indexed: 12/25/2022] Open
Abstract
Peripheral nerve trauma results in functional loss in the innervated organ, and recovery without surgical intervention is rare. Many surgical techniques can be used for nerve repair. Among these, the tubulization technique can be highlighted: this allows regenerative factors to be introduced into the chamber. Cell therapy and tissue engineering have arisen as an alternative for stimulating and aiding peripheral nerve regeneration. Therefore, the aim of this review was to provide a survey and analysis on the results from experimental and clinical studies that used cell therapy and tissue engineering as tools for optimizing the regeneration process. The articles used came from the LILACS, Medline and SciELO scientific databases. Articles on the use of stem cells, Schwann cells, growth factors, collagen, laminin and platelet-rich plasma for peripheral nerve repair were summarized over the course of the review. Based on these studies, it could be concluded that the use of stem cells derived from different sources presents promising results relating to nerve regeneration, because these cells have a capacity for neuronal differentiation, thus demonstrating effective functional results. The use of tubes containing bioactive elements with controlled release also optimizes the nerve repair, thus promoting greater myelination and axonal growth of peripheral nerves. Another promising treatment is the use of platelet-rich plasma, which not only releases growth factors that are important in nerve repair, but also serves as a carrier for exogenous factors, thereby stimulating the proliferation of specific cells for peripheral nerve repair.
Collapse
|
42
|
Al Ghrbawy NM, Afify RAAM, Dyaa N, El Sayed AA. Differentiation of Bone Marrow: Derived Mesenchymal Stem Cells into Hepatocyte-like Cells. Indian J Hematol Blood Transfus 2015; 32:276-83. [PMID: 27429519 DOI: 10.1007/s12288-015-0581-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 08/17/2015] [Indexed: 02/06/2023] Open
Abstract
Cirrhosis is the end-stage liver fibrosis, whereby normal liver architecture is disrupted by fibrotic bands, parenchymal nodules and vascular distortion. Portal hypertension and hepatocyte dysfunction are the end results and give rise to major systemic complications and premature death. Mesenchymal stem cells (MSC) have the capacity of self-renew and to give rise to cells of various lineages, so MSC can be isolated from bone marrow (BM) and induced to differentiate into hepatocyte-like cells. MSC were induced to differentiate into hepatocyte-like cells by hepatotic growth factor (HGF) and fibroblast growth factor-4 (FGF-4). Differentiated cells were examined for the expression of hepatocyte-specific markers and hepatocyte functions. MSC were isolated. Flow cytometry analysis showed that they expressed the MSC-specific markers, reverse transcriptase-polymerase chain reaction (RT-PCR) demonstrated that MSC expressed the hepatocyte-specific marker cytokeratin 18 (CK-18) following hepatocyte induction. This study demonstrates that BM-derived-MSC can differentiate into functional hepatocyte-like cells following the induction of HGF and FGF-4. MSC can serve as a favorable cell source for tissue engineering in the treatment of liver disease.
Collapse
Affiliation(s)
- Nesrien M Al Ghrbawy
- Clinical Pathology Department, Faculty of Medicine, Al kaser Al Aini, Cairo University, Cairo, Egypt
| | | | - Nehal Dyaa
- Clinical Pathology Department, Faculty of Medicine, Al kaser Al Aini, Cairo University, Cairo, Egypt
| | | |
Collapse
|
43
|
Choudhery MS, Badowski M, Muise A, Pierce J, Harris DT. Subcutaneous Adipose Tissue-Derived Stem Cell Utility Is Independent of Anatomical Harvest Site. Biores Open Access 2015; 4:131-145. [PMID: 26309790 PMCID: PMC4497709 DOI: 10.1089/biores.2014.0059] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
One of the challenges for tissue engineering and regenerative medicine is to obtain suitably large cell numbers for therapy. Mesenchymal stem cells (MSCs) can easily be expanded in vitro to obtain large numbers of cells, but this approach may induce cellular senescence. The characteristics of cells are dependent on variables like age, body mass index (BMI), and disease conditions, however, and in the case of adipose tissue-derived stem cells (ASCs), anatomical harvest site is also an important variable that can affect the regenerative potential of isolated cells. We therefore had kept the parameters (age, BMI, disease conditions) constant in this study to specifically assess influence of anatomical sites of individual donors on utility of ASCs. Adipose tissue was obtained from multiple anatomical sites in individual donors, and viability and nucleated cell yield were determined. MSC frequency was enumerated using colony forming unit assay and cells were characterized by flow cytometry. Growth characteristics were determined by long-term population doubling analysis of each sample. Finally, MSCs were induced to undergo adipogenic, osteogenic, and chondrogenic differentiation. To validate the findings, these results were compared with similar single harvest sites from multiple individual patients. The results of the current study indicated that MSCs obtained from multiple harvest sites in a single donor have similar morphology and phenotype. All adipose depots in a single donor exhibited similar MSC yield, viability, frequency, and growth characteristics. Equivalent differentiation capacity into osteocytes, adipocytes, and chondrocytes was also observed. On the basis of results, we conclude that it is acceptable to combine MSCs obtained from various anatomical locations in a single donor to obtain suitably large cell numbers required for therapy, avoiding in vitro senescence and lengthy and expensive in vitro culturing and expansion steps.
Collapse
Affiliation(s)
- Mahmood S. Choudhery
- Tissue Engineering and Regenerative Medicine Laboratory, Advance Research Center of Biomedical Sciences, King Edward Medical University, Lahore, Pakistan
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Michael Badowski
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - Angela Muise
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | - John Pierce
- Aesthetic Surgery of Tucson, Tucson, Arizona
| | - David T. Harris
- Department of Immunobiology, College of Medicine, The University of Arizona, Tucson, Arizona
| |
Collapse
|
44
|
Scuteri A, Donzelli E, Foudah D, Caldara C, Redondo J, D’Amico G, Tredici G, Miloso M. Mesengenic differentiation: comparison of human and rat bone marrow mesenchymal stem cells. Int J Stem Cells 2014; 7:127-134. [PMID: 25473450 PMCID: PMC4249895 DOI: 10.15283/ijsc.2014.7.2.127] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Cellular therapies using Mesenchymal Stem Cells (MSCs) represent a promising approach for the treatment of degenerative diseases, in particular for mesengenic tissue regeneration. However, before the approval of clinical trials in humans, in vitro studies must be performed aimed at investigating MSCs' biology and the mechanisms regulating their proliferation and differentiation abilities. Besides studies on human MSCs (hMSCs), MSCs derived from rodents have been the most used cellular type for in vitro studies. Nevertheless, the transfer of the results obtained using animal MSCs to hMSCs has been hindered by the limited knowledge regarding the similarities existing between cells of different origins. Aim of this paper is to highlight similarities and differences and to clarify the sometimes reported different results obtained using these cells. METHODS AND RESULTS We compare the differentiation ability into mesengenic lineages of rat and human MSCs cultured in their standard conditions. Our results describe in which way the source from which MSCs are derived affects their differentiation potential, depending on the mesengenic lineage considered. For osteogenic and chondrogenic lineages, the main difference between human and rat MSCs is represented by differentiation time, while for adipogenesis hMSCs have a greater differentiation potential. CONCLUSIONS These results on the one hand suggest to carefully evaluate the transfer of results obtained with animal MSCs, on the other hand they offer a clue to better apply MSCs into clinical practice.
Collapse
Affiliation(s)
- Arianna Scuteri
- Dipartimento di Chirurgia e Medicina Traslazionale, Università Milano-Bicocca, via Cadore
- NeuroMi, Milan Center for Neurosciences, Milano, Italy
| | - Elisabetta Donzelli
- Dipartimento di Chirurgia e Medicina Traslazionale, Università Milano-Bicocca, via Cadore
- NeuroMi, Milan Center for Neurosciences, Milano, Italy
| | - Dana Foudah
- Dipartimento di Chirurgia e Medicina Traslazionale, Università Milano-Bicocca, via Cadore
| | - Cristina Caldara
- Dipartimento di Chirurgia e Medicina Traslazionale, Università Milano-Bicocca, via Cadore
| | - Juliana Redondo
- Dipartimento di Chirurgia e Medicina Traslazionale, Università Milano-Bicocca, via Cadore
| | - Giovanna D’Amico
- Centro Ricerca Tettamanti, Clinica Pediatrica, Università Milano-Bicocca, Monza
| | - Giovanni Tredici
- Dipartimento di Chirurgia e Medicina Traslazionale, Università Milano-Bicocca, via Cadore
- NeuroMi, Milan Center for Neurosciences, Milano, Italy
| | - Mariarosaria Miloso
- Dipartimento di Chirurgia e Medicina Traslazionale, Università Milano-Bicocca, via Cadore
- NeuroMi, Milan Center for Neurosciences, Milano, Italy
| |
Collapse
|
45
|
Identifying the role of microRNAs in spinal cord injury. Neurol Sci 2014; 35:1663-71. [PMID: 25231644 DOI: 10.1007/s10072-014-1940-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 08/06/2014] [Indexed: 02/07/2023]
Abstract
Spinal cord injury (SCI) is medically and socioeconomically debilitating, and effective treatments are lacking. The elucidation of the pathophysiological mechanisms underlying SCI is essential for developing effective treatments for SCI. MicroRNAs (miRNAs) are small non-coding RNA molecules (18-24 nucleotides long) that regulate gene expression by interacting with specific target sequences. Recent studies suggest that miRNAs can act as post-transcriptional regulators to inhibit mRNA translation. Bioinformatic analyses indicate that the altered expression of miRNAs has an effect on critical processes of SCI physiopathology, including astrogliosis, oxidative stress, inflammation, apoptosis, and neuroplasticity. Therefore, the study of miRNAs may provide new insights into the molecular mechanisms of SCI. Current studies have also provided potential therapeutic clinical applications that involve targeting mRNAs to treat SCI. This review summarizes the biogenesis and function of miRNAs and the roles of miRNAs in SCI. We also discuss the potential therapeutic applications of miRNA-based interventions for SCI.
Collapse
|
46
|
Allers C, Jones JA, Lasala GP, Minguell JJ. Mesenchymal stem cell therapy for the treatment of amyotrophic lateral sclerosis: signals for hope? Regen Med 2014; 9:637-47. [DOI: 10.2217/rme.14.30] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Based on the distinctive cellular, molecular and immunomodulatory traits of mesenchymal stem cells (MSC), it has been postulated that these cells may play a critical role in regenerative medicine. In addition to the participation of MSC in the repair of mesodermal-derived tissues (bone, cartilage), robust data have suggested that MSC may also play a reparative role in conditions involving damage of cells of ectodermal origin. The above content has been supported by the capability of MSC to differentiate into neuron-like cells as well as by a competence to generate a ‘neuroprotective’ environment. In turn, several preclinical studies have put forward the concept that MSC therapy may represent an option for the treatment of several neurological disorders and injuries, including amyotrophic lateral sclerosis. We expect that the above foundations, which have inspired this review, may result in the founding of an effective and/or palliative therapy for amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Carolina Allers
- TCA Cellular Therapy, LLC, 101 Judge Tanner Blvd, Suite 502, Covington, LA 70433, USA
| | - Janet A Jones
- TCA Cellular Therapy, LLC, 101 Judge Tanner Blvd, Suite 502, Covington, LA 70433, USA
- School of Nursing, Southeastern Louisiana University, Hammond, LA 70402, USA
| | - Gabriel P Lasala
- TCA Cellular Therapy, LLC, 101 Judge Tanner Blvd, Suite 502, Covington, LA 70433, USA
| | - José J Minguell
- TCA Cellular Therapy, LLC, 101 Judge Tanner Blvd, Suite 502, Covington, LA 70433, USA
| |
Collapse
|
47
|
Zhao B, Xu B, Hu W, Song C, Wang F, Liu Z, Ye M, Zou H, Miao QR. Comprehensive proteome quantification reveals NgBR as a new regulator for epithelial-mesenchymal transition of breast tumor cells. J Proteomics 2014; 112:38-52. [PMID: 25173099 DOI: 10.1016/j.jprot.2014.08.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 07/22/2014] [Accepted: 08/04/2014] [Indexed: 12/13/2022]
Abstract
UNLABELLED Nogo-B receptor (NgBR) is a type I receptor and specifically binds to ligand Nogo-B. Our previous work has shown that NgBR is highly expressed in human breast invasive ductal carcinoma. Here, comprehensive proteome quantification was performed to examine the alteration of protein expression profile in MDA-MB-231 breast tumor cells after knocking down NgBR using lentivirus-mediated shRNA approach. Among a total of 1771 proteins feasibly quantified, 994 proteins were quantified in two biological replicates with RSD <50%. There are 122 proteins significantly down-regulated in NgBR knockdown MDA-MB-231 breast tumor cells, such as vimentin and S100A4, well-known markers for mesenchymal cells, and CD44, a stemness indicator. The decrease of vimentin, S100A4 and CD44 protein expression levels was further confirmed by Western blot analysis. MDA-MB-231 cells are typical breast invasive ductal carcinoma cells showing mesenchymal phenotype. Cell morphology analysis demonstrates NgBR knockdown in MDA-MB-231 cells results in reversibility of epithelial-mesenchymal transition (EMT), which is one of the major mechanisms involved in breast cancer metastasis. Furthermore, we demonstrated that NgBR knockdown in MCF-7 cells significantly prevented the TGF-β-induced EMT process as determined by the morphology change, and staining of E-cadherin intercellular junction as well as the decreased expression of vimentin. BIOLOGICAL SIGNIFICANCE Our previous publication showed that NgBR is highly expressed in human breast invasive ductal carcinoma. However, the roles of NgBR and NgBR-mediated signaling pathway in breast tumor cells are still unclear. Here, we not only demonstrated that the quantitative proteomics analysis is a powerful tool to investigate the global biological function of NgBR, but also revealed that NgBR is involved in the transition of breast epithelial cells to mesenchymal stem cells, which is one of the major mechanisms involved in breast cancer metastasis. These findings provide new insights for understanding the roles of NgBR in regulating breast epithelial cell transform during the pathogenesis of breast cancer.
Collapse
Affiliation(s)
- Baofeng Zhao
- Division of Pediatric Surgery, Department of Surgery, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Bo Xu
- Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Wenquan Hu
- Division of Pediatric Surgery, Department of Surgery, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Chunxia Song
- Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Fangjun Wang
- Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Zhong Liu
- Division of Pediatric Surgery, Department of Surgery, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mingliang Ye
- Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Hanfa Zou
- Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| | - Qing R Miao
- Division of Pediatric Surgery, Department of Surgery, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Division of Pediatric Pathology, Department of Pathology, Children's Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
48
|
Immunological barriers to stem cell therapy in the central nervous system. Stem Cells Int 2014; 2014:507905. [PMID: 25165476 PMCID: PMC4138731 DOI: 10.1155/2014/507905] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/16/2014] [Indexed: 12/21/2022] Open
Abstract
The central nervous system is vulnerable to many neurodegenerative disorders such as Alzheimer's disease that result in the extensive loss of neuronal cells. Stem cells have the ability to differentiate into many types of cells, which make them ideal for treating such disorders. Although stem cell therapy has shown some promising results in animal models for many brain disorders it has yet to translate into the clinic. A major hurdle to the translation of stem cell therapy into the clinic is the immune response faced by stem cell transplants. Here, we focus on immunological and related hurdles to stem cell therapies for central nervous system disorders.
Collapse
|
49
|
Ganz J, Arie I, Buch S, Zur TB, Barhum Y, Pour S, Araidy S, Pitaru S, Offen D. Dopaminergic-like neurons derived from oral mucosa stem cells by developmental cues improve symptoms in the hemi-parkinsonian rat model. PLoS One 2014; 9:e100445. [PMID: 24945922 PMCID: PMC4063966 DOI: 10.1371/journal.pone.0100445] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 05/28/2014] [Indexed: 11/23/2022] Open
Abstract
Achieving safe and readily accessible sources for cell replacement therapy in Parkinson’s disease (PD) is still a challenging unresolved issue. Recently, a primitive neural crest stem cell population (hOMSC) was isolated from the adult human oral mucosa and characterized in vitro and in vivo. In this study we assessed hOMSC ability to differentiate into dopamine-secreting cells with a neuronal-dopaminergic phenotype in vitro in response to dopaminergic developmental cues and tested their therapeutic potential in the hemi-Parkinsonian rat model. We found that hOMSC express constitutively a repertoire of neuronal and dopaminergic markers and pivotal transcription factors. Soluble developmental factors induced a reproducible neuronal-like morphology in the majority of hOMSC, downregulated stem cells markers, upregulated the expression of the neuronal and dopaminergic markers that resulted in dopamine release capabilities. Transplantation of these dopaminergic-induced hOMSC into the striatum of hemi-Parkinsonian rats improved their behavioral deficits as determined by amphetamine-induced rotational behavior, motor asymmetry and motor coordination tests. Human TH expressing cells and increased levels of dopamine in the transplanted hemispheres were observed 10 weeks after transplantation. These results demonstrate for the first time that soluble factors involved in the development of DA neurons, induced a DA phenotype in hOMSC in vitro that significantly improved the motor function of hemiparkinsonian rats. Based on their neural-related origin, their niche accessibility by minimal-invasive procedures and their propensity for DA differentiation, hOMSC emerge as an attractive tool for autologous cell replacement therapy in PD.
Collapse
Affiliation(s)
- Javier Ganz
- Neurosciences Laboratory, Felsenstein Medical Research Center-Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Ina Arie
- Oral Biology Dept., School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sigal Buch
- Oral Biology Dept., School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tali Ben Zur
- Neurosciences Laboratory, Felsenstein Medical Research Center-Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Yael Barhum
- Neurosciences Laboratory, Felsenstein Medical Research Center-Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Sammy Pour
- Oral & Maxillofacial Dept., Baruch Padeh Medical Center, Poria, Lower Galilee, Israel
| | - Shareef Araidy
- Oral & Maxillofacial Dept., Baruch Padeh Medical Center, Poria, Lower Galilee, Israel
| | - Sandu Pitaru
- Oral Biology Dept., School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daniel Offen
- Neurosciences Laboratory, Felsenstein Medical Research Center-Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
50
|
Mostafavi FS, Razavi S, Mardani M, Esfandiari E, Esfahani HZ, Kazemi M. Comparative Study of Microtubule-associated Protein-2 and Glial Fibrillary Acidic Proteins during Neural Induction of Human Bone Marrow Mesenchymal Stem Cells and Adipose-Derived Stem Cells. Int J Prev Med 2014; 5:584-95. [PMID: 24932390 PMCID: PMC4050679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 02/16/2014] [Indexed: 10/25/2022] Open
Abstract
BACKGROUND In recent years, adipose tissue, due to the stem cells contained within, has found a new special place in laboratory and clinical applications. These adipose-derived stem cells (ADSCs) have the same characteristics of bone marrow mesenchymal stem cells (BMSCs). Although bone marrow (BM) is not easily accessible and its procurements may be painful, most patients possess excess fat which can be obtained by less invasive methods; this makes adipose tissue ubiquitous, available and an ideal large-scale source for research on clinical applications. METHODS BMSCs and ADSCs were harvested from three healthy human and were characterized using flow-cytometry. After they were treated for neurosphere formation using basic fibroblast growth factor, epidermal growth factor, B 27; terminal differentiation was performed. In this study, we used immunocytochemistry, real time-polymerase chain reaction and western blotting techniques for detection and comparison of Nestin, microtubule-associated protein-2 (MAP-2) and glial fibrillary acidic protein (GFAP) markers in human ADSCs and BMSCs. RESULTS Under appropriate conditions ADSCs can differentiate into neuron-like cells and express neural markers the same as BMSCs, also the expression of GFAP marker in differentiated cells derived from ADSCs was significantly lower than the cells derived from BMSCs (P < 0.05). While the expression of MAP-2 marker in both groups was the same. CONCLUSIONS However, due to its advantages and according to our results based on the expression levels of GFAP and MAP-2, adipose tissue rather than BM could represent a more appropriate stem cell source for investigating the application of these cells in understanding the pathophysiology and in treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Fatemeh Sadat Mostafavi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahnaz Razavi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran,Correspondence to: Prof. Shahnaz Razavi, Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan 81744-176, Iran. E-mail:
| | - Mohammad Mardani
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ebrahim Esfandiari
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Mohammad Kazemi
- Department of Genetic, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|