51
|
Volkman R, Offen D. Concise Review: Mesenchymal Stem Cells in Neurodegenerative Diseases. Stem Cells 2017; 35:1867-1880. [PMID: 28589621 DOI: 10.1002/stem.2651] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/06/2017] [Indexed: 12/13/2022]
Abstract
Stem cell-based therapies for neurodegenerative diseases aim at halting clinical deterioration by regeneration and by providing local support for damaged tissue. Mesenchymal stem cells (MSCs) hold great potential for cell therapy as they can be efficiently derived from adult tissue, ex vivo expanded in culture and safely transplanted autologously. MSCs were also shown to be able to differentiate toward neural fates and to secrete a broad range of factors able to promote nervous tissue maintenance and repair. Moreover, upon transplantation, MSCs were shown capable of homing toward lesioned areas, implying their potential use as vehicles for therapeutic agents administration. Indeed, various advantageous effects were reported following human MSCs transplantation into rodent models of neurodegenerative diseases, such as neurotrophic factor-mediated protection, enhanced neurogenesis, modulation of inflammation, and abnormal protein aggregate clearance. Per journal style, most nonstandard abbreviations must be used at least two times in the abstract to be retained; NTF was used once and thus has been deleted. Recent studies have also used ex vivo manipulation for enhanced expression of potentially favorable factors, by so exploiting the homing capacity of MSCs for effective expression at the lesion site. Here, we will summarize current advancements in MSCs-based therapies for neurodegenerative diseases. We will examine the roles of central mechanisms suggested to mediate the beneficial effects of MSCs-based therapy and consider the augmentation of these mechanisms for superior clinical outcomes in rodent models of neurodegeneration as well as in clinical trials. Stem Cells 2017;35:1867-1880.
Collapse
|
52
|
Jeong SG, Oh YS, Joe IS, Jeong SY, Cho HM, Lee JS, Oh WK, Cho TO, Cho GW. Functional restoration of replicative senescent mesenchymal stem cells by the brown alga Undaria pinnatifida. Anim Cells Syst (Seoul) 2017; 21:108-114. [PMID: 30460058 PMCID: PMC6138307 DOI: 10.1080/19768354.2017.1292951] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/13/2017] [Accepted: 01/24/2017] [Indexed: 12/12/2022] Open
Abstract
The brown alga Undaria pinnatifida, which is called Mi-Yoek in Korea, has been traditionally consumed as a health food in East Asian countries. Recent studies have reported that U. pinnatifida has beneficial effects on arteriosclerosis, inflammation, fat metabolism, and tumors. In this study, we examined the anti-senescence effects of ethanol extracts of U. pinnatifida (UP-Ex) in human bone marrow mesenchymal stem cells (hBM-MSCs). UP-Ex protected hBM-MSCs against oxidative injury, as determined by MTT assays. This effect was confirmed by immunoblot analysis of the oxidation-sensitive protein p53 and the apoptotic protein cleaved caspase-3. Excessive intracellular reactive oxygen species (ROS) accumulation induced by oxidative stress was moderated in UP-Ex-treated hBM-MSCs (UP-Ex-MSCs). Similarly, expression of the ROS-scavenging enzymes superoxide dismutase 1 (SOD1), SOD2, and catalase was recovered in UP-Ex-MSCs. Excessive ROS induced by long-term cell expansion (passage 17) was significantly decreased along with restoration of the senescence proteins p53, p21, and p16 in UP-Ex-MSCs. UP-Ex treatment also improved the ability of these replicative, senescent hBM-MSCs (passage 17) to differentiate into osteocytes or adipocytes, suggesting that UP-Ex ameliorates the functional decline of senescent stem cells and may provide better therapeutic efficacy in stem cell therapy. Abbreviations: hBM-MSCs: human bone marrow mesenchymal stem cells; DCF: 2′,7′-dichlorodihydrofluorescein; DCFH-DA: 2′,7′-dichlorofluorescein diacetate; MTT: 3-(4,5-dimethylthiazol-2-yl-)2,5-diphenyltetrazolium bromide; PBS: phosphate-buffered saline; PFA: paraformaldehyde; RIPA: radioimmunoprecipitation assay; ROS: reactive oxygen species; SOD1: superoxide dismutase 1; SOD2: superoxide dismutase 2.
Collapse
Affiliation(s)
- Sin-Gu Jeong
- Department of Biology, College of Natural Science, Chosun University, Gwangju, Korea.,Department of Life Science, BK21-Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju, Korea
| | - Youn Seo Oh
- Department of Biology, College of Natural Science, Chosun University, Gwangju, Korea.,Department of Life Science, BK21-Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju, Korea
| | - I-Seul Joe
- Department of Biology, College of Natural Science, Chosun University, Gwangju, Korea.,Department of Life Science, BK21-Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju, Korea
| | - So Young Jeong
- Department of Biology, College of Natural Science, Chosun University, Gwangju, Korea
| | - Hyo Moon Cho
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Jun Sik Lee
- Department of Biology, College of Natural Science, Chosun University, Gwangju, Korea.,Department of Life Science, BK21-Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju, Korea
| | - Won Keun Oh
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Science, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Tae Oh Cho
- Department of Biology, College of Natural Science, Chosun University, Gwangju, Korea
| | - Goang-Won Cho
- Department of Biology, College of Natural Science, Chosun University, Gwangju, Korea.,Department of Life Science, BK21-Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju, Korea
| |
Collapse
|
53
|
Assoni A, Coatti G, Valadares MC, Beccari M, Gomes J, Pelatti M, Mitne-Neto M, Carvalho VM, Zatz M. Different Donors Mesenchymal Stromal Cells Secretomes Reveal Heterogeneous Profile of Relevance for Therapeutic Use. Stem Cells Dev 2016; 26:206-214. [PMID: 27762666 DOI: 10.1089/scd.2016.0218] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal X-linked disorder caused by null mutations in the dystrophin gene. Although the primary defect is the deficiency of muscle dystrophin, secondary events, including chronic inflammation, fibrosis, and muscle regeneration failure are thought to actively contribute to disease progression. Despite several advances, there is still no effective therapy for DMD. Therefore, the potential regenerative capacities, and immune-privileged properties of mesenchymal stromal cells (MSCs), have been the focus of intense investigation in different animal models aiming the treatment of these disorders. However, these studies have shown different outcomes according to the sources from which MSCs were obtained, which raise the question whether stem cells from distinct sources have comparable clinical effects. Here, we analyzed the protein content of the secretome of MSCs, isolated from three different sources (adipose tissue, skeletal muscle, and uterine tubes), obtained from five donors and evaluated their in vitro properties when cocultured with DMD myoblasts. All MSC lineages showed pathways enrichment related to protein metabolic process, oxidation-reduction process, cell proliferation, and regulation of apoptosis. We found that MSCs secretome proteins and their effect in vitro vary significantly according to the tissue and donors, including opposite effects in apoptosis assay, indicating the importance of characterizing MSC secretome profile before its use in animal and clinical trials. Despite the individual differences a pool of conditioned media from all MSCs lineages was able to delay apoptosis and enhance migration when in contact with DMD myoblasts.
Collapse
Affiliation(s)
- Amanda Assoni
- 1 Human Genome and Stem Cell Research Center, Institute of Biosciences University of São Paulo , São Paulo, Brazil
| | - Giuliana Coatti
- 1 Human Genome and Stem Cell Research Center, Institute of Biosciences University of São Paulo , São Paulo, Brazil
| | - Marcos C Valadares
- 1 Human Genome and Stem Cell Research Center, Institute of Biosciences University of São Paulo , São Paulo, Brazil
| | - Melinda Beccari
- 1 Human Genome and Stem Cell Research Center, Institute of Biosciences University of São Paulo , São Paulo, Brazil
| | - Juliana Gomes
- 1 Human Genome and Stem Cell Research Center, Institute of Biosciences University of São Paulo , São Paulo, Brazil
| | - Mayra Pelatti
- 1 Human Genome and Stem Cell Research Center, Institute of Biosciences University of São Paulo , São Paulo, Brazil
| | - Miguel Mitne-Neto
- 1 Human Genome and Stem Cell Research Center, Institute of Biosciences University of São Paulo , São Paulo, Brazil .,2 Fleury Group (Research and Development Department), São Paulo, Brazil
| | | | - Mayana Zatz
- 1 Human Genome and Stem Cell Research Center, Institute of Biosciences University of São Paulo , São Paulo, Brazil
| |
Collapse
|
54
|
Noh MY, Lim SM, Oh KW, Cho KA, Park J, Kim KS, Lee SJ, Kwon MS, Kim SH. Mesenchymal Stem Cells Modulate the Functional Properties of Microglia via TGF-β Secretion. Stem Cells Transl Med 2016; 5:1538-1549. [PMID: 27400795 DOI: 10.5966/sctm.2015-0217] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 04/18/2016] [Indexed: 12/13/2022] Open
Abstract
: The regulation of microglial cell phenotype is a potential therapeutic intervention in neurodegenerative disease. Previously, we reported that transforming growth factor-β (TGF-β) levels in mesenchymal stromal cells (MSCs) could be used as potential biological markers to predict the effectiveness of autologous MSC therapy in patients with amyotrophic lateral sclerosis. However, the underlying mechanism of TGF-β in MSCs was not fully elucidated in determining the functional properties of microglia. In this study, we aimed to clarify the role of TGF-β that is involved in MSC effectiveness, especially focusing on microglia functional properties that play a pivotal role in neuroinflammation. We found that MSC-conditioned media (MSC-CM) inhibited proinflammatory cytokine expression, restored alternative activated microglia phenotype markers (fractalkine receptor, mannose receptor, CD200 receptor), and enhanced phagocytosis in lipopolysaccharide (LPS)-stimulated microglia. In addition, TGF-β in MSC-CM played a major role in these effects by inhibiting the nuclear factor-κB pathway and restoring the TGF-β pathway in LPS-stimulated microglia. Recombinant TGF-β also induced similar effects to MSC-CM in LPS-stimulated microglia. Therefore, we propose that MSCs can modulate the functional properties of microglia via TGF-β secretion, switching them from a classically activated phenotype to an inflammation-resolving phenotype. The latter role may be associated with the inhibition of neuroinflammatory processes in neurodegenerative disorders. SIGNIFICANCE The results of this study showed that microglia functional properties may be modulated depending on the composition and quantity of mesenchymal stromal cell (MSC)-secreting factors. Transforming growth factor (TGF)-β is proposed as a modulator of microglia functional properties among MSC-secreting factors, and this study aligns with a previous clinical study by these same authors. TGF-β releasing capacity could be an important factor enhancing the therapeutic efficacy of MSCs in clinical trials.
Collapse
Affiliation(s)
- Min Young Noh
- Cell Therapy Center and Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Su Min Lim
- Cell Therapy Center and Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Ki-Wook Oh
- Cell Therapy Center and Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Kyung-Ah Cho
- Cell Therapy Center and Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Department of Neural Development and Disease and Behavioral Neuroepigenetics Laboratory, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jinseok Park
- Cell Therapy Center and Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Kyung-Suk Kim
- Bioengineering Institute, CORESTEM Inc., Seoul, Republic of Korea
| | - Su-Jung Lee
- Cell Therapy Center and Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Min-Soo Kwon
- Department of Pharmacology, School of Medicine, CHA University, Gyeonggi-do, Republic of Korea
| | - Seung Hyun Kim
- Cell Therapy Center and Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
55
|
Jeong SG, Cho GW. Accumulation of apoptosis-insensitive human bone marrow-mesenchymal stromal cells after long-term expansion. Cell Biochem Funct 2016; 34:310-6. [DOI: 10.1002/cbf.3191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 03/05/2016] [Accepted: 04/13/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Sin-Gu Jeong
- Department of Biology, College of Natural Science; Chosun University; Gwangju Korea
- Department of Life Science, BK21-Plus Research Team for Bioactive Control Technology; Chosun University; Gwangju Korea
| | - Goang-Won Cho
- Department of Biology, College of Natural Science; Chosun University; Gwangju Korea
- Department of Life Science, BK21-Plus Research Team for Bioactive Control Technology; Chosun University; Gwangju Korea
| |
Collapse
|
56
|
Affiliation(s)
- Fumito Endo
- Department of Neuroscience and Pathobiology; Research Institute of Environmental Medicine; Nagoya University; Nagoya Japan
| | - Okiru Komine
- Department of Neuroscience and Pathobiology; Research Institute of Environmental Medicine; Nagoya University; Nagoya Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology; Research Institute of Environmental Medicine; Nagoya University; Nagoya Japan
| |
Collapse
|
57
|
Oh YS, Kim SH, Cho GW. Functional Restoration of Amyotrophic Lateral Sclerosis Patient-Derived Mesenchymal Stromal Cells Through Inhibition of DNA Methyltransferase. Cell Mol Neurobiol 2016; 36:613-20. [PMID: 26210997 DOI: 10.1007/s10571-015-0242-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 07/18/2015] [Indexed: 12/11/2022]
Abstract
Alteration of DNA methylation is highly associated with aging and neurodegenerative disorders, such as amyotrophic lateral sclerosis (ALS). Remedying these aberrant methylation patterns may serve to improve these diseases. Previously, we reported that human bone marrow mesenchymal stromal cells isolated from ALS patients (ALS-MSCs) have functionally decreased stem cell potency, and excessively express DNA methyltransferases (DNMTs). In this study, we examined the correlation between excessive DNMT expression and functional decline in ALS-MSCs. The DNMT inhibitor RG108 was used for this. RG108-treated ALS-MSCs exhibit increased expression of the anti-senescence genes TERT, VEGF, and ANG, and decreased expression of the senescence-related genes ATM and p21. The activity of SA-β-galactosidase and the expression of senescence proteins p53 and p16 were reduced in RG108-treated ALS-MSCs. The abilities of cell migration and protection against oxidative damage were improved in the treated ALS-MSCs. In neuronal differentiation experiments, the treated MSCs more effectively differentiated into neuron-like cells. These results suggest that ALS-MSC function can be restored by inhibiting excessively expressed DNMTs, an approach that may ultimately provide better efficacy in stem cell therapy.
Collapse
Affiliation(s)
- Youn Seo Oh
- Department of Biology, College of Natural Science, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 501-759, Korea
- Department of Life Science, BK21-Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju, 501-759, Korea
| | - Seung Hyun Kim
- Department of Neurology, Hanyang University College of Medicine, Seoul, Korea
- Cell Therapy Center, Hanyang University Hospital, Seoul, Korea
| | - Goang-Won Cho
- Department of Biology, College of Natural Science, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 501-759, Korea.
- Department of Life Science, BK21-Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju, 501-759, Korea.
| |
Collapse
|
58
|
Kim SM, Yun CK, Park JH, Hwang JW, Kim ZH, Choi YS. Efficient cryopreservation of human mesenchymal stem cells using silkworm hemolymph-derived proteins. J Tissue Eng Regen Med 2016. [DOI: 10.1002/term.2116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Sun-Mi Kim
- Department of Biotechnology; CHA University; Seongnam Republic of Korea
| | - Chang-Koo Yun
- Department of Biotechnology; CHA University; Seongnam Republic of Korea
| | - Jin-Ho Park
- Department of Biotechnology; CHA University; Seongnam Republic of Korea
| | - Jung Wook Hwang
- Department of Biotechnology; CHA University; Seongnam Republic of Korea
| | - Z-Hun Kim
- Department of Biotechnology; CHA University; Seongnam Republic of Korea
| | - Yong-Soo Choi
- Department of Biotechnology; CHA University; Seongnam Republic of Korea
| |
Collapse
|
59
|
Stem Cells for Amyotrophic Lateral Sclerosis. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
60
|
Hajra A, Bandyopadhyay D, Hajra S. Amyotrophic lateral sclerosis: promising therapeutic outcome-not far away? Neural Regen Res 2016; 11:856. [PMID: 27335576 PMCID: PMC4904483 DOI: 10.4103/1673-5374.182715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
61
|
Stavely R, Robinson AM, Miller S, Boyd R, Sakkal S, Nurgali K. Allogeneic guinea pig mesenchymal stem cells ameliorate neurological changes in experimental colitis. Stem Cell Res Ther 2015; 6:263. [PMID: 26718461 PMCID: PMC4697327 DOI: 10.1186/s13287-015-0254-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 11/12/2015] [Accepted: 12/02/2015] [Indexed: 12/13/2022] Open
Abstract
Background The use of mesenchymal stem cells (MSCs) to treat inflammatory bowel disease (IBD) is of great interest because of their immunomodulatory properties. Damage to the enteric nervous system (ENS) is implicated in IBD pathophysiology and disease progression. The most commonly used model to study inflammation-induced changes to the ENS is 2,4,6-trinitrobenzene-sulfonate acid (TNBS)-induced colitis in guinea pigs; however, no studies using guinea pig MSCs in colitis have been performed. This study aims to isolate and characterise guinea pig MSCs and then test their therapeutic potential for the treatment of enteric neuropathy associated with intestinal inflammation. Methods MSCs from guinea pig bone marrow and adipose tissue were isolated and characterised in vitro. In in vivo experiments, guinea pigs received either TNBS for the induction of colitis or sham treatment by enema. MSCs were administered at a dose of 1 × 106 cells via enema 3 h after the induction of colitis. Colon tissues were collected 24 and 72 h after TNBS administration to assess the level of inflammation and damage to the ENS. The secretion of transforming growth factor-β1 (TGF-β1) was analysed in MSC conditioned medium by flow cytometry. Results Cells isolated from both sources were adherent to plastic, multipotent and expressed some human MSC surface markers. In vitro characterisation revealed distinct differences in growth kinetics, clonogenicity and cell morphology between MSC types. In an in vivo model of TNBS-induced colitis, guinea pig bone marrow MSCs were comparatively more efficacious than adipose tissue MSCs in attenuating weight loss, colonic tissue damage and leukocyte infiltration into the mucosa and myenteric plexus. MSCs from both sources were equally neuroprotective in the amelioration of enteric neuronal loss and changes to the neurochemical coding of neuronal subpopulations. MSCs from both sources secreted TGF-β1 which exerted neuroprotective effects in vitro. Conclusions This study is the first evaluating the functional capacity of guinea pig bone marrow and adipose tissue-derived MSCs and providing evidence of their neuroprotective value in an animal model of colitis. In vitro characteristics of MSCs cannot be extrapolated to their therapeutic efficacy. TGF-β1 released by both types of MSCs might have contributed to the attenuation of enteric neuropathy associated with colitis.
Collapse
Affiliation(s)
- Rhian Stavely
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Ainsley M Robinson
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Sarah Miller
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Richard Boyd
- Department of Anatomy and Developmental Biology, Monash University, 19 Innovation Walk, Clayton, 3800, Victoria, Australia.
| | - Samy Sakkal
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Kulmira Nurgali
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| |
Collapse
|
62
|
Kim N, Cho SG. Overcoming immunoregulatory plasticity of mesenchymal stem cells for accelerated clinical applications. Int J Hematol 2015; 103:129-37. [PMID: 26662288 DOI: 10.1007/s12185-015-1918-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 11/25/2015] [Accepted: 11/30/2015] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells with the potential to differentiate into different tissue lineages. In addition to their differentiation potential, MSCs possess immunomodulatory properties that have created growing interest in both pre-clinical and clinical research. Over the years, MSCs have been applied rapidly in the clinic in a wide variety of immune-mediated disorders; however, MSC therapy has shown contradictory results, often with poor clinical outcomes. Recently, studies on MSC-based immune modulation have provided possible explanations for the conflicting clinical reports. It is now generally recognized that the immunomodulatory properties of MSCs are not constitutive but are induced by various mediators present in the inflammatory microenvironment. Different inflammatory stimuli are able to polarize MSCs to elicit distinct immunomodulatory phenotypes. Thus, the concepts of plasticity and polarization of MSC-based immune modulation may have important therapeutic implications in the clinic. In this review, we focus on the underlying mechanisms of MSC-mediated immune regulation that contribute to their therapeutic potential. Importantly, we discuss novel strategic approaches that enhance the therapeutic potential of MSCs through a consideration of MSC plasticity in immune modulation.
Collapse
Affiliation(s)
- Nayoun Kim
- Institute for Translational Research and Molecular Imaging, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, #505, Banpo-Dong, Seocho-Ku, Seoul, 137-040, Korea.,Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease (CRCID), Seoul, Korea
| | - Seok-Goo Cho
- Institute for Translational Research and Molecular Imaging, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, #505, Banpo-Dong, Seocho-Ku, Seoul, 137-040, Korea. .,Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease (CRCID), Seoul, Korea. .,Department of Hematology, Catholic Blood and Marrow Transplantation Center, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea.
| |
Collapse
|
63
|
|
64
|
Nolta JA. New advances in understanding stem cell fate and function. Stem Cells 2015; 33:313-5. [PMID: 25446041 DOI: 10.1002/stem.1905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Indexed: 01/01/2023]
|
65
|
Park CM, Kim MJ, Kim SM, Park JH, Kim ZH, Choi YS. Umbilical cord mesenchymal stem cell-conditioned media prevent muscle atrophy by suppressing muscle atrophy-related proteins and ROS generation. In Vitro Cell Dev Biol Anim 2015; 52:68-76. [DOI: 10.1007/s11626-015-9948-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/31/2015] [Indexed: 12/11/2022]
|
66
|
Kim HJ, Seo SW, Chang JW, Lee JI, Kim CH, Chin J, Choi SJ, Kwon H, Yun HJ, Lee JM, Kim ST, Choe YS, Lee KH, Na DL. Stereotactic brain injection of human umbilical cord blood mesenchymal stem cells in patients with Alzheimer's disease dementia: A phase 1 clinical trial. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2015; 1:95-102. [PMID: 29854930 PMCID: PMC5975048 DOI: 10.1016/j.trci.2015.06.007] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction We conducted a phase 1 clinical trial in nine patients with mild-to-moderate Alzheimer's disease to evaluate the safety and dose-limiting toxicity of stereotactic brain injection of human umbilical cord blood–derived mesenchymal stem cells (hUCB-MSCs). Methods The low- (n = 3) and high-dose (n = 6) groups received a total of 3.0 × 106 cells/60 μL and 6.0 × 106 cells/60 μL, respectively, into the bilateral hippocampi and right precuneus. Results No patient showed serious adverse events including fever during the 24-month follow-up period. During the 12-week follow-up period, the most common acute adverse event was wound pain from the surgical procedure (n = 9), followed by headache (n = 4), dizziness (n = 3), and postoperative delirium (n = 3). There was no dose-limiting toxicity. Discussion Administration of hUCB-MSCs into the hippocampus and precuneus by stereotactic injection was feasible, safe, and well tolerated. Further trials are warranted to test the efficacy. Clinical Trial Registration ClinicalTrial.gov identifier NCT01297218 and NCT01696591.
Collapse
Affiliation(s)
- Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Jong Wook Chang
- Stem Cell & Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea.,Biomedical Research Institute, MEDIPOST Co., Ltd, Seoul, Korea
| | - Jung Il Lee
- Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chi Hun Kim
- Department of Psychology, Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Juhee Chin
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd, Seoul, Korea
| | - Hunki Kwon
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | - Hyuk Jin Yun
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | - Jong Min Lee
- Department of Biomedical Engineering, Hanyang University, Seoul, Korea
| | - Sung Tae Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yearn Seong Choe
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Korea.,Neuroscience Center, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
67
|
Eve DJ, Ehrhart J, Zesiewicz T, Jahan I, Kuzmin-Nichols N, Sanberg CD, Gooch C, Sanberg PR, Garbuzova-Davis S. Plasma Derived From Human Umbilical Cord Blood Modulates Mitogen-Induced Proliferation of Mononuclear Cells Isolated From the Peripheral Blood of ALS Patients. Cell Transplant 2015; 25:963-71. [PMID: 26159164 DOI: 10.3727/096368915x688579] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by degeneration of motor neurons in the spinal cord and brain. This disease clinically manifests as gradual muscular weakness and atrophy leading to paralysis and death by respiratory failure. While multiple interdependent factors may contribute to the pathogenesis of ALS, increasing evidence shows the possible presence of autoimmune mechanisms that promote disease progression. The potential use of plasma derived from human umbilical cord blood (hUCB) as a therapeutic tool is currently in its infancy. The hUCB plasma is rich in cytokines and growth factors that are required for growth and survival of cells during hematopoiesis. In this study, we investigated the effects of hUCB plasma on the mitogen-induced proliferation of mononuclear cells (MNCs) isolated from the peripheral blood of ALS patients and apoptotic activity by detection of caspase 3/7 expression of the isolated MNCs in vitro. Three distinct responses to phytohemagglutinin (PHA)-induced proliferation of MNCs were observed, which were independent of age, disease duration, and the ALS rating scale: Group I responded normally to PHA, Group II showed no response to PHA, while Group III showed a hyperactive response to PHA. hUCB plasma attenuated the hyperactive response (Group III) and potentiated the normal response in Group I ALS patients, but did not alter that of the nonresponders to PHA (Group II). The elevated activity of caspase 3/7 observed in the MNCs from ALS patients was significantly reduced by hUCB plasma treatment. Thus, study results showing different cell responses to mitogen suggest alteration in lymphocyte functionality in ALS patients that may be a sign of immune deficiency in the nonresponders and autoimmunity alterations in the hyperactive responders. The ability of hUCB plasma to modulate the mitogen cell response and reduce caspase activity suggests that the use of hUCB plasma alone, or with stem cells, may prove useful as a therapeutic in ALS patients.
Collapse
Affiliation(s)
- David J Eve
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Oh KW, Moon C, Kim HY, Oh SI, Park J, Lee JH, Chang IY, Kim KS, Kim SH. Phase I trial of repeated intrathecal autologous bone marrow-derived mesenchymal stromal cells in amyotrophic lateral sclerosis. Stem Cells Transl Med 2015; 4:590-7. [PMID: 25934946 DOI: 10.5966/sctm.2014-0212] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/16/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Stem cell therapy is an emerging alternative therapeutic or disease-modifying strategy for amyotrophic lateral sclerosis (ALS). The aim of this open-label phase I clinical trial was to evaluate the safety of two repeated intrathecal injections of autologous bone marrow (BM)-derived mesenchymal stromal cells (MSCs) in ALS patients. Eight patients with definite or probable ALS were enrolled. After a 3-month lead-in period, autologous MSCs were isolated two times from the BM at an interval of 26 days and were then expanded in vitro for 28 days and suspended in autologous cerebrospinal fluid. Of the 8 patients, 7 received 2 intrathecal injections of autologous MSCs (1 × 10(6) cells per kg) 26 days apart. Clinical or laboratory measurements were recorded to evaluate the safety 12 months after the first MSC injection. The ALS Functional Rating Scale-Revised (ALSFRS-R), the Appel ALS score, and forced vital capacity were used to evaluate the patients' disease status. One patient died before treatment and was withdrawn from the study. With the exception of that patient, no serious adverse events were observed during the 12-month follow-up period. Most of the adverse events were self-limited or subsided after supportive treatment within 4 days. Decline in the ALSFRS-R score was not accelerated during the 6-month follow-up period. Two repeated intrathecal injections of autologous MSCs were safe and feasible throughout the duration of the 12-month follow-up period. SIGNIFICANCE Stem cell therapy is an emerging alternative therapeutic or disease-modifying strategy for amyotrophic lateral sclerosis (ALS). To the authors' best knowledge, there are no clinical trials to evaluate the safety of repeated intrathecal injections of autologous bone marrow mesenchymal stromal cells in ALS. After the clinical trial (phase I/II) was conducted, the stem cell (HYNR-CS, NEURONATA-R) was included in the revision of the regulations on orphan drug designation (number 160; December 31, 2013) and approved as a New Drug Application (Department of Cell and Gene Therapy 233; July 30, 2014) by the Korean Food and Drug Administration. The phase II trial is expected to be reported later.
Collapse
Affiliation(s)
- Ki-Wook Oh
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Chanil Moon
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Hyun Young Kim
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Sung-Il Oh
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Jinseok Park
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Jun Ho Lee
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - In Young Chang
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Kyung Suk Kim
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| |
Collapse
|
69
|
Burns TC, Verfaillie CM. From mice to mind: Strategies and progress in translating neuroregeneration. Eur J Pharmacol 2015; 759:90-100. [PMID: 25814255 DOI: 10.1016/j.ejphar.2015.03.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/18/2015] [Accepted: 03/12/2015] [Indexed: 12/20/2022]
Abstract
Decisions about what experimental therapies are advanced to clinical trials are based almost exclusively on findings in preclinical animal studies. Over the past 30 years, animal models have forecast the success of hundreds of neuroprotective pharmacological therapies for stroke, Alzheimer׳s disease, spinal cord injury, traumatic brain injury and amyotrophic lateral sclerosis. Yet almost without exception, all have failed. Rapid advances in stem cell technologies have raised new hopes that these neurological diseases may one day be treatable. Still, how can neuroregenerative therapies be translated into clinical realities if available animal models are such poor surrogates of human disease? To address this question we discuss human and rodent neurogenesis, evaluate mechanisms of action for cellular therapies and describe progress in translating neuroregeneration to date. We conclude that not only are appropriate animal models critical to the development of safe and effective therapies, but that the multiple mechanisms of stem cell-mediated therapies may be particularly well suited to the mechanistically diverse nature of central nervous system diseases in mice and man.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurosurgery and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, USA.
| | | |
Collapse
|
70
|
Oh J, An JW, Oh SI, Oh KW, Kim JA, Lee JS, Kim SH. Socioeconomic costs of amyotrophic lateral sclerosis according to staging system. Amyotroph Lateral Scler Frontotemporal Degener 2015; 16:202-8. [PMID: 25646865 DOI: 10.3109/21678421.2014.999791] [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] [Indexed: 11/13/2022]
Abstract
The objective of this study was to compare the cost of illness of amyotrophic lateral sclerosis (ALS) in the Korean population based on the staging system for ALS from the perspective of both patients and the government. Direct medical costs, care-related costs, and loss of productivity in patients with ALS were measured based on medical records and face-to-face interviews. The patients were divided into groups according to the staging system for ALS, and the cost of illness was analysed. A total of 151 patients with ALS were enrolled in the study. The mean monthly cost of ALS was US $7902 per patient and increased according to the disease stage (stage 2, US $5181; stage 3, US $7089; stage 4, US $10,557). Of direct medical costs (US $3436), 44.8% of the cost burden was carried by patients, and the remaining costs were paid by the government. In conclusion, although the current coverage rate of the National Health Insurance (NHI) system for rare and intractable diseases including ALS is 90%, the rate of direct medical costs paid by patients and out-of-pocket costs remain high. Moreover, coverage rates and cost of illness are closely related with disease severity.
Collapse
Affiliation(s)
- Juyeon Oh
- Cell Therapy Center for Intractable Disorders, Hanyang University Hospital , Seoul , Korea
| | | | | | | | | | | | | |
Collapse
|
71
|
Hypoxia/Reoxygenation-Preconditioned Human Bone Marrow-Derived Mesenchymal Stromal Cells Rescue Ischemic Rat Cortical Neurons by Enhancing Trophic Factor Release. Mol Neurobiol 2014; 52:792-803. [DOI: 10.1007/s12035-014-8912-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 09/28/2014] [Indexed: 02/07/2023]
|
72
|
Kim C, Lee HC, Sung JJ. Amyotrophic lateral sclerosis - cell based therapy and novel therapeutic development. Exp Neurobiol 2014; 23:207-14. [PMID: 25258567 PMCID: PMC4174611 DOI: 10.5607/en.2014.23.3.207] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease, characterized by the predominant loss of motor neurons (MNs) in primary motor cortex, the brainstem, and the spinal cord, causing premature death in most cases. Minimal delay of pathological development by available medicine has prompted the search for novel therapeutic treatments to cure ALS. Cell-based therapy has been proposed as an ultimate source for regeneration of MNs. Recent completion of non-autologous fetal spinal stem cell transplant to ALS patients brought renewed hope for further human trials to cure the disease. Autologous somatic stem cell-based human trials are now in track to reveal the outcome of the ongoing trials. Furthermore, induced pluripotent stem cell (iPSC)-based ALS disease drug screen and autologous cell transplant options will broaden therapeutic options. In this review paper, we discuss recent accomplishments in cell transplant treatment for ALS and future options with iPSC technology.
Collapse
Affiliation(s)
- Changsung Kim
- Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Korea
| | - Hee Chul Lee
- Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Korea
| | - Jung-Joon Sung
- Department of Neurology, Seoul National University Hospital, Seoul 110-774, Korea
| |
Collapse
|