1
|
Cacciamali A, Pascucci L, Villa R, Dotti S. Engineered nanoparticles toxicity on adipose tissue derived mesenchymal stem cells: A preliminary investigation. Res Vet Sci 2022; 152:134-149. [PMID: 35969916 DOI: 10.1016/j.rvsc.2022.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 07/12/2022] [Accepted: 08/03/2022] [Indexed: 11/15/2022]
Abstract
Nanoscience and nanotechnologies have recently gained importance in several fields, such as industry and medicine. A big issue of the increasing application of nanomaterials is the poor literature regarding their potential toxicity in humans and animals. Recently, adult stem cells have been proposed as putative targets of nanoparticles (NPs). This study aims to investigate the effects of zerovalent-metallic NPs on isolated and amplified equine Adipose tissue derived Mesenchymal Stem Cells (eAdMSCs). Cells were treated with Cobalt (Co-), Iron (Fe-), and Nickel (Ni-) nanoparticles (NPs) at different concentrations and were characterized for the cytotoxic and genotoxic effects of exposure. Treatment with NPs resulted in reduced cell viability and proliferative capability in comparison with untreated cells. However, this did not influence eAdMSCs potency, as treated cells were able to differentiate towards the adipogenic and osteogenic lineages. Ni- and Fe-NPs showed cytoplasmic localization, while Co-NPs entered the nucleus and mitochondria, suggesting a potential genotoxic activity. Regarding p53 expression, it was enhanced in the first 48 h after treatments, with a drastic reduction of expression within 72 h. Higher p53 expression was reported in the case of Co-NP treatment, suggesting the tumorigenic potential of these NPs. Telomerase activity was enhanced by Fe- and Ni-NP treatments in a concentration- and time-dependent way. This was not true for Co-NP treated samples, suggesting a reduced replicative capacity of eAdMSCs upon Co-NP exposure. The present study is a preliminary investigation of the influence exerted by NPs on eAdMSC physiological activity in terms of cytotoxic and genotoxic effects. The present results revealed eAdMSC physiology to be strongly influenced by NPs in a dose-, time- and NP-dependent way.
Collapse
Affiliation(s)
- Andrea Cacciamali
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Laboratorio di Controllo di Prodotti Biologici, Centro di Referenza Nazionale per i Metodi Alternativi, Benessere e Cura degli Animali da Laboratorio, 25124 Brescia, Italy.
| | - Luisa Pascucci
- Dipartimento di Medicina Veterinaria, Università degli Studi di Perugia, 06126 Perugia, Italy.
| | - Riccardo Villa
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Laboratorio di Controllo di Prodotti Biologici, Centro di Referenza Nazionale per i Metodi Alternativi, Benessere e Cura degli Animali da Laboratorio, 25124 Brescia, Italy.
| | - Silvia Dotti
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Laboratorio di Controllo di Prodotti Biologici, Centro di Referenza Nazionale per i Metodi Alternativi, Benessere e Cura degli Animali da Laboratorio, 25124 Brescia, Italy.
| |
Collapse
|
2
|
Schepici G, Gugliandolo A, Mazzon E. Serum-Free Cultures: Could They Be a Future Direction to Improve Neuronal Differentiation of Mesenchymal Stromal Cells? Int J Mol Sci 2022; 23:ijms23126391. [PMID: 35742836 PMCID: PMC9223839 DOI: 10.3390/ijms23126391] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are undifferentiated cells with multilinear potential, known for their immunomodulatory and regenerative properties. Although the scientific community is working to improve their application, concerns limit their use to repair tissues following neurological damage. One of these obstacles is represented by the use of culture media supplemented with fetal bovine serum (FBS), which, due to its xenogenic nature and the risk of contamination, has increased scientific, ethical and safety problems. Therefore, the use of serum-free media could improve MSC culture methods, avoiding infectious and immunogenic transmission problems as well as MSC bioprocesses, without the use of animal components. The purpose of our review is to provide an overview of experimental studies that demonstrate that serum-free cultures, along with the supplementation of growth factors or chemicals, can lead to a more defined and controlled environment, enhancing the proliferation and neuronal differentiation of MSCs.
Collapse
|
3
|
Jahed FJ, Rahbarghazi R, Shafaei H, Rezabakhsh A, Karimipour M. Application of neurotrophic factor-secreting cells (astrocyte - Like cells) in the in-vitro Alzheimer's disease-like pathology on the human neuroblastoma cells. Brain Res Bull 2021; 172:180-189. [PMID: 33895268 DOI: 10.1016/j.brainresbull.2021.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 04/10/2021] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
This study investigated physical proximity and paracrine activity of neurotrophic factor-secreting cells (NTF-SCs) on beta-amyloid treated cells. Mesenchymal stem cells (MSCs) - to-NTF-SCs (Astrocyte -like cells) trans-differentiation was confirmed using immunofluorescence staining of GFAP. BDNF and NGF levels were measured by ELISA. To mimic AD-like condition, SH-SY5Y cells were exposed to 10 μM Aβ1-42. SH-SY5Y cells were allocated into Control; and Aβ1-42-treated cells. Treated cells were further classified into three subgroups including Aβ1-42 cells, Aβ1-42 cells + NTF-SCs (CM) and Aβ1-42 cells + NTF-SCs co-culture. Cell viability was measured by MTT assay. Anti-inflammatory and anti-tau hyperphosphorylation effects of NTF-SCs were assessed via monitoring TNF-α and hyperphosphorylated Tau protein expression level respectively. To explore the impact of NTF-SCs on synaptogenesis and synaptic functionality, real-time PCR assay was performed to measure the expression of synapsine 1, homer 1 and ZIF268. The level of synaptophysin was monitored via immunofluorescence staining. Data showed MSCs potential in trans-differentiating toward NTF-SCs indicated with enhanced GFAP expression (p < 0.05). ELISA assay confirmed the superiority of NTF-SCs in releasing NGF and BDNF compared to the MSCs (p < 0.05). Aβ significantly induced SH-SY5Y cells death while juxtacrine and paracrine activity of NTF-SCs significantly blunted these conditions (p < 0.05). Trans-differentiated cells had potential to reduce Tau hyperphosphorylation and TNF-α level after treatment with Aβ through juxtacrine and paracrine mechanisms (p < 0.05). Moreover, NTF-SCs significantly increased the expression rate of synapsin 1, homer 1 and zif 268 genes in Aβ-treated cells compared to matched-control group coincided with induction of synaptophysin at the protein level(p < 0.05). NTF-SCs reversed AD-like neuropathological alterations in SH-SY5Y cells via paracrine and juxtacrine mechanisms.
Collapse
Affiliation(s)
- Fatemeh Jafari Jahed
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Shafaei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimipour
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Wlodarek L, Cao F, Alibhai FJ, Fekete A, Noyan N, Tobin SW, Marvasti TB, Wu J, Li SH, Weisel RD, Wang LY, Jia Z, Li RK. Rectification of radiotherapy-induced cognitive impairments in aged mice by reconstituted Sca-1 + stem cells from young donors. J Neuroinflammation 2020; 17:51. [PMID: 32028989 PMCID: PMC7006105 DOI: 10.1186/s12974-019-1681-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 12/17/2019] [Indexed: 01/03/2023] Open
Abstract
Background Radiotherapy is widely used and effective for treating brain tumours, but inevitably impairs cognition as it arrests cellular processes important for learning and memory. This is particularly evident in the aged brain with limited regenerative capacity, where radiation produces irreparable neuronal damage and activation of neighbouring microglia. The latter is responsible for increased neuronal death and contributes to cognitive decline after treatment. To date, there are few effective means to prevent cognitive deficits after radiotherapy. Methods Here we implanted hematopoietic stem cells (HSCs) from young or old (2- or 18-month-old, respectively) donor mice expressing green fluorescent protein (GFP) into old recipients and assessed cognitive abilities 3 months post-reconstitution. Results Regardless of donor age, GFP+ cells homed to the brain of old recipients and expressed the macrophage/microglial marker, Iba1. However, only young cells attenuated deficits in novel object recognition and spatial memory and learning in old mice post-irradiation. Mechanistically, old recipients that received young HSCs, but not old, displayed significantly greater dendritic spine density and long-term potentiation (LTP) in CA1 neurons of the hippocampus. Lastly, we found that GFP+/Iba1+ cells from young and old donors were differentially polarized to an anti- and pro-inflammatory phenotype and produced neuroprotective factors and reactive nitrogen species in vivo, respectively. Conclusion Our results suggest aged peripherally derived microglia-like cells may exacerbate cognitive impairments after radiotherapy, whereas young microglia-like cells are polarized to a reparative phenotype in the irradiated brain, particularly in neural circuits associated with rewards, learning, and memory. These findings present a proof-of-principle for effectively reinstating central cognitive function of irradiated brains with peripheral stem cells from young donor bone marrow. Electronic supplementary material The online version of this article (10.1186/s12974-019-1681-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lukasz Wlodarek
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Feng Cao
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Program in Neurosciences & Mental Health, SickKids Research Institute, Floor 5, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Faisal J Alibhai
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Adam Fekete
- Program in Neurosciences & Mental Health, SickKids Research Institute, Floor 5, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Nima Noyan
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Stephanie W Tobin
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Tina B Marvasti
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada.,Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Jun Wu
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Shu-Hong Li
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Richard D Weisel
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada.,Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - Lu-Yang Wang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada. .,Program in Neurosciences & Mental Health, SickKids Research Institute, Floor 5, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.
| | - Zhengping Jia
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada. .,Program in Neurosciences & Mental Health, SickKids Research Institute, Floor 5, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.
| | - Ren-Ke Li
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada. .,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada. .,Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
5
|
Lee JY, Kim HS, Kim SH, Kim HS, Cho BP. Combination of Human Mesenchymal Stem Cells and Repetitive Transcranial Magnetic Stimulation Enhances Neurological Recovery of 6-Hydroxydopamine Model of Parkinsonian's Disease. Tissue Eng Regen Med 2020; 17:67-80. [PMID: 31970698 DOI: 10.1007/s13770-019-00233-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/23/2019] [Accepted: 11/28/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) has been in use for the treatment of various neurological diseases, including depression, anxiety, stroke and Parkinson's disease (PD), while its underlying mechanism is stills unclear. This study was undertaken to evaluate the potential synergism of rTMS treatment to the beneficial effect of human mesenchymal stem cells (hMSCs) administration for PD and to clarify the mechanism of action of this therapeutic approach. METHODS The neuroprotective effect in nigral dopamine neurons, neurotrophic/growth factors and anti-/pro-inflammatory cytokine regulation, and functional recovery were assessed in the rat 6-hydroxydopamine (6-OHDA) model of PD upon administration of hMSCs and rTMS. RESULTS Transplanted hMSCs were identified in the substantia nigra, and striatum. Enhancement of the survival of SN dopamine neurons and the expression of the tyrosine hydroxylase protein were observed in the hMSCs + rTMS compared to that of controls. Combination therapy significantly elevated the expression of several key neurotrophic factors, of which the highest expression was recorded in the rTMS + hMSC group. In addition, the combination therapy significantly upregulated IL-10 expression while decreased IFN-γ and TNF-α production in a synergistic manner. The treadmill locomotion test (TLT) revealed that motor function was improved in the rTMS + hMSC treatment with synergy. CONCLUSION Our findings demonstrate that rTMS treatment and hMSC transplantation could synergistically create a favorable microenvironment for cell survival within the PD rat brain, through alteration of soluble factors such as neurotrophic/growth factors and anti-/pro-inflammatory cytokines related to neuronal protection or repair, with preservation of DA neurons and improvement of motor functions.
Collapse
Affiliation(s)
- Ji Yong Lee
- Department of Anatomy, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, 26426, Republic of Korea
| | - Hyun Soo Kim
- FCB-Pharmicell Co. Ltd., 520 Sicox Tower, 484 Dunchon-daero, Jungwon-gu, Seongnam-si, Gyeonggi-do, 13229, Republic of Korea
| | - Sung Hoon Kim
- Department and Rehabilitation Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, 26426, Republic of Korea
| | - Han-Soo Kim
- Department of Biomedical Sciences, Catholic Kwandong University College of Medical Convergence, 24 Beomil-ro, 579 beon-gil, Gangneung-Si, Gangwon-do, 25601, Republic of Korea.
- Basic Research Division, Biomedical Institute of Mycological Resource, College of Medicine, Catholic Kwandong University, 24 Beomil-ro, 579 beon-gil, Gangneung-Si, Gangwon-do, 25601, Republic of Korea.
| | - Byung Pil Cho
- Department of Anatomy, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, 26426, Republic of Korea.
- Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, 26426, Republic of Korea.
| |
Collapse
|
6
|
Xue J, Liu Y, Darabi MA, Tu G, Huang L, Ying L, Xiao B, Wu Y, Xing M, Zhang L, Zhang L. An injectable conductive Gelatin-PANI hydrogel system serves as a promising carrier to deliver BMSCs for Parkinson's disease treatment. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 100:584-597. [DOI: 10.1016/j.msec.2019.03.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 12/27/2018] [Accepted: 03/07/2019] [Indexed: 12/17/2022]
|
7
|
Scheper V, Schwieger J, Hamm A, Lenarz T, Hoffmann A. BDNF-overexpressing human mesenchymal stem cells mediate increased neuronal protection in vitro. J Neurosci Res 2019; 97:1414-1429. [PMID: 31257632 PMCID: PMC6772136 DOI: 10.1002/jnr.24488] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/31/2019] [Accepted: 06/12/2019] [Indexed: 12/21/2022]
Abstract
The use of neurotrophic factors as therapeutic agents for neurodegenerative diseases is considered as an approach aimed at restoring and maintaining neuronal function in the peripheral and central nervous system. Since the neuroprotective effect is depending on chronic delivery of the neurotrophic factors a sustained application, e.g., via cell‐based delivery is necessary. Human mesenchymal stem cells (hMSCs) were lentivirally modified to overexpress brain‐derived neurotrophic factor (BDNF) and to express fluorescent marker genes for easy visualization. Since genetically modified cells should be site‐specifically retained (e.g., by encapsulation) in the patients to avoid adverse effects the cells were additionally differentiated to chondrocytes to hypothetically improve their vitality and survival in a delivery matrix. Different polycations for lentiviral transduction were investigated for their efficiency. The success of differentiation was determined by analysis of chondrocyte marker genes and the neuroprotective effect of BDNF‐overexpressing cells was exemplarily investigated on neurons of the peripheral auditory system. The genetically modified hMSCs overexpressed BDNF from under 1 to 125 ng ml−1 day−1 depending on the donor and transfection method. Using protamine sulfate the transfection efficacy was superior compared to the use of polybrene. The BDNF secreted by the MSCs was significantly neuroprotective in comparison to the relevant controls even though the produced mean concentrations were lower than the effective concentrations for recombinant industrially produced proteins described in literature. The presented system of BDNF‐overexpressing hMSCs is neuroprotective and is therefore considered as a promising method for sustained delivery of proteins in therapeutically relevant amounts to degenerating neuronal structures.
Collapse
Affiliation(s)
- Verena Scheper
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence Hearing4all, German Research Foundation, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Jana Schwieger
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Anika Hamm
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany.,Department of Orthopaedic Surgery, Hannover Medical School, Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence Hearing4all, German Research Foundation, Hannover, Germany.,Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Andrea Hoffmann
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany.,Department of Orthopaedic Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
8
|
Schulze J, Kaiser O, Paasche G, Lamm H, Pich A, Hoffmann A, Lenarz T, Warnecke A. Effect of hyperbaric oxygen on BDNF-release and neuroprotection: Investigations with human mesenchymal stem cells and genetically modified NIH3T3 fibroblasts as putative cell therapeutics. PLoS One 2017; 12:e0178182. [PMID: 28542481 PMCID: PMC5441643 DOI: 10.1371/journal.pone.0178182] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/09/2017] [Indexed: 12/30/2022] Open
Abstract
Hyperbaric oxygen therapy (HBOT) is a noninvasive widely applied treatment that increases the oxygen pressure in tissues. In cochlear implant (CI) research, intracochlear application of neurotrophic factors (NTFs) is able to improve survival of spiral ganglion neurons (SGN) after deafness. Cell-based delivery of NTFs such as brain-derived neurotrophic factor (BDNF) may be realized by cell-coating of the surface of the CI electrode. Human mesenchymal stem cells (MSC) secrete a variety of different neurotrophic factors and may be used for the development of a biohybrid electrode in order to release endogenously-derived neuroprotective factors for the protection of residual SGN and for a guided outgrowth of dendrites in the direction of the CI electrode. HBOT could be used to influence cell behaviour after transplantation to the inner ear. The aim of this study was to investigate the effect of HBOT on the proliferation, BDNF-release and secretion of neuroprotective factors. Thus, model cells (an immortalized fibroblast cell line (NIH3T3)–native and genetically modified) and MSCs were repeatedly (3 x – 10 x) exposed to 100% oxygen at different pressures. The effects of HBO on cell proliferation were investigated in relation to normoxic and normobaric conditions (NOR). Moreover, the neuroprotective and neuroregenerative effects of HBO-treated cells were analysed by cultivation of SGN in conditioned medium. Both, the genetically modified NIH3T3/BDNF and native NIH3T3 fibroblasts, showed a highly significant increased proliferation after five days of HBOT in comparison to normoxic controls. By contrast, the number of MSCs was decreased in MSCs treated with 2.0 bar of HBO. Treating SGN cultures with supernatants of fibroblasts and MSCs significantly increased the survival rate of SGN. HBO treatment did not influence (increase / reduce) this effect. Secretome analysis showed that HBO treatment altered the protein expression pattern in MSCs.
Collapse
Affiliation(s)
- Jennifer Schulze
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all”, Hannover, Germany
- * E-mail:
| | - Odett Kaiser
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all”, Hannover, Germany
| | - Gerrit Paasche
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all”, Hannover, Germany
| | - Hans Lamm
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Hannover, Germany
| | - Andreas Pich
- Core Facility Proteomics, Hannover Medical School, Hannover, Germany
| | - Andrea Hoffmann
- Department of Orthopaedic Surgery, Hannover Medical School, Hannover, Germany
| | - Thomas Lenarz
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all”, Hannover, Germany
| | - Athanasia Warnecke
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all”, Hannover, Germany
| |
Collapse
|
9
|
Blecker D, Elashry MI, Heimann M, Wenisch S, Arnhold S. New Insights into the Neural Differentiation Potential of Canine Adipose Tissue-Derived Mesenchymal Stem Cells. Anat Histol Embryol 2017; 46:304-315. [PMID: 28401575 DOI: 10.1111/ahe.12270] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/11/2017] [Indexed: 12/13/2022]
Abstract
Adipose tissue-derived stem cells (ASCs) can be obtained from different adipose tissue sources within the body. It is an abundant cell pool, easily accessible, suitable for cultivation and expansion in vitro and preparation for therapeutic approaches. Amongst these therapeutic approaches are tissue engineering and nervous system disorders such as spinal cord injuries. For such treatment, ASCs have to be reliably differentiated in to the neuronal direction. Therefore, we investigated the neural differentiation potential of ASCs using protocols with neurogenic inductors such as valproic acid and forskolin, while dog brain tissue served as control. Morphological changes could already be noticed 1 h after neuronal induction. Gene expression analysis revealed that the neuronal markers nestin and βIII-tubulin as well as MAP2 were expressed after induction of neuronal differentiation. Additionally, the expression of the neurotrophic factors NGF, BDNF and GDNF was determined. Some of the neuronal markers and neurotrophic factors were already expressed in undifferentiated cells. Our findings point out that ASCs can reliably be differentiated into the neuronal lineage; therefore, these cells are a suitable cell source for cell transplantation in disorders of the central nervous system. Follow-up studies would show the clinical benefit of these cells after transplantation.
Collapse
Affiliation(s)
- D Blecker
- Institute of Veterinary-Anatomy, -Histology and -Embryology, University of Giessen, Frankfurter Str. 98., 35392, Giessen, Germany
| | - M I Elashry
- Institute of Veterinary-Anatomy, -Histology and -Embryology, University of Giessen, Frankfurter Str. 98., 35392, Giessen, Germany.,Anatomy and Embryology Department, Faculty of Veterinary Medicine, University of Mansoura, 35516, Egypt
| | - M Heimann
- Institute of Veterinary-Anatomy, -Histology and -Embryology, University of Giessen, Frankfurter Str. 98., 35392, Giessen, Germany
| | - S Wenisch
- Department of Veterinary Clinical Sciences, Small Animal Clinic c/o Institute of Veterinary Anatomy, Histology and -Embryology, University of Giessen, Frankfurter Str. 98., 35392, Giessen, Germany
| | - S Arnhold
- Institute of Veterinary-Anatomy, -Histology and -Embryology, University of Giessen, Frankfurter Str. 98., 35392, Giessen, Germany
| |
Collapse
|
10
|
Darabi S, Tiraihi T, Delshad A, Sadeghizadeh M, Khalil W, Taheri T. In vitro non-viral murine pro-neurotrophin 3 gene transfer into rat bone marrow stromal cells. J Neurol Sci 2017; 375:137-145. [PMID: 28320116 DOI: 10.1016/j.jns.2017.01.058] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 12/12/2022]
Abstract
Neurotrophin 3 (NT-3) is an important factor for promoting prenatal neural development, as well as regeneration, axogenesis and plasticity in postnatal life. Therapy with NT-3 was reported to improve the condition of patients suffering from degenerative diseases and traumatic injuries, however, the disadvantage of NT-3 protein delivery is its short half-life, thus our alternative approach is the use of NT-3 gene therapy. In this study, the bone marrow stromal cells (BMSCs) were isolated from adult rats, cultured for 4 passages and transfected with either pEGFP-N1 or a constructed vector containing murine proNT-3 (pSecTag2/HygroB-murine proNT-3) using Lipofectamine 2000 followed by Hygromycin B (200mg/kg). The transfection efficiency of the transiently transfected BMSCs was evaluated using the green fluorescence protein containing vector (pEGFP-N1). A quantitative evaluation of the NT-3 expression of mRNA using real time qRT-PCR shows that there was double fold increase in NT-3 gene expression compared with non-transfected BMSCs, also, the culture supernatant yielded double fold increase in NT-3 using ELISA technique, the data were supported by immunoblotting technique. This suggests that the use of this transfection technique can be useful for gene therapy in different neurological disorders with neurodegenerative or traumatic origins.
Collapse
Affiliation(s)
- Shahram Darabi
- Cellular and Molecular Research Center, Qazvin University of Medical Science, Qazvin, Iran
| | - Taki Tiraihi
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | | | - Majid Sadeghizadeh
- Department of Genetics, Faculty of Basic Sciences, Tarbiat Modares University, Tehran, Iran
| | - Wisam Khalil
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Taher Taheri
- Shefa Neurosciences Research Center, Khatam Al-Anbia Hospital, Tehran, Iran
| |
Collapse
|
11
|
Shen Y, Huang J, Liu L, Xu X, Han C, Zhang G, Jiang H, Li J, Lin Z, Xiong N, Wang T. A Compendium of Preparation and Application of Stem Cells in Parkinson's Disease: Current Status and Future Prospects. Front Aging Neurosci 2016; 8:117. [PMID: 27303288 PMCID: PMC4885841 DOI: 10.3389/fnagi.2016.00117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Parkinson's Disease (PD) is a progressively neurodegenerative disorder, implicitly characterized by a stepwise loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and explicitly marked by bradykinesia, rigidity, resting tremor and postural instability. Currently, therapeutic approaches available are mainly palliative strategies, including L-3,4-dihydroxy-phenylalanine (L-DOPA) replacement therapy, DA receptor agonist and deep brain stimulation (DBS) procedures. As the disease proceeds, however, the pharmacotherapeutic efficacy is inevitably worn off, worse still, implicated by side effects of motor response oscillations as well as L-DOPA induced dyskinesia (LID). Therefore, the frustrating status above has propeled the shift to cell replacement therapy (CRT), a promising restorative therapy intending to secure a long-lasting relief of patients' symptoms. By far, stem cell lines of multifarious origins have been established, which can be further categorized into embryonic stem cells (ESCs), neural stem cells (NSCs), induced neural stem cells (iNSCs), mesenchymal stem cells (MSCs), and induced pluripotent stem cells (iPSCs). In this review, we intend to present a compendium of preparation and application of multifarious stem cells, especially in relation to PD research and therapy. In addition, the current status, potential challenges and future prospects for practical CRT in PD patients will be elaborated as well.
Collapse
Affiliation(s)
- Yan Shen
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Jinsha Huang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Ling Liu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Xiaoyun Xu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Chao Han
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Guoxin Zhang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Haiyang Jiang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Jie Li
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Zhicheng Lin
- Department of Psychiatry, Harvard Medical School, Division of Alcohol and Drug Abuse, and Mailman Neuroscience Research Center, McLean Hospital Belmont, MA, USA
| | - Nian Xiong
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Tao Wang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| |
Collapse
|
12
|
Transplantation of human mesenchymal stem cells into the cisterna magna and its neuroprotective effects in a parkinsonian animal model. Mol Cell Toxicol 2015. [DOI: 10.1007/s13273-015-0038-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
13
|
Chudickova M, Bruza P, Zajicova A, Trosan P, Svobodova L, Javorkova E, Kubinova S, Holan V. Targeted neural differentiation of murine mesenchymal stem cells by a protocol simulating the inflammatory site of neural injury. J Tissue Eng Regen Med 2015; 11:1588-1597. [DOI: 10.1002/term.2059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 01/19/2015] [Accepted: 04/29/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Milada Chudickova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
- Faculty of Science; Charles University; Prague Czech Republic
| | - Petr Bruza
- Faculty of Biomedical Engineering; Czech Technical University in Prague; Kladno Czech Republic
| | - Alena Zajicova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Peter Trosan
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
- Faculty of Science; Charles University; Prague Czech Republic
| | - Lucie Svobodova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Eliska Javorkova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
- Faculty of Science; Charles University; Prague Czech Republic
| | - Sarka Kubinova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Vladimir Holan
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
- Faculty of Science; Charles University; Prague Czech Republic
| |
Collapse
|
14
|
Gabr H, El-Kheir WA, Farghali HAMA, Ismail ZMK, Zickri MB, El Maadawi ZM, Kishk NA, Sabaawy HE. Intrathecal Transplantation of Autologous Adherent Bone Marrow Cells Induces Functional Neurological Recovery in a Canine Model of Spinal Cord Injury. Cell Transplant 2014; 24:1813-27. [PMID: 25199146 DOI: 10.3727/096368914x683025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) results in demyelination of surviving axons, loss of oligodendrocytes, and impairment of motor and sensory functions. We have developed a clinical strategy of cell therapy for SCI through the use of autologous bone marrow cells for transplantation to augment remyelination and enhance neurological repair. In a preclinical large mammalian model of SCI, experimental dogs were subjected to a clipping contusion of the spinal cord. Two weeks after the injury, GFP-labeled autologous minimally manipulated adherent bone marrow cells (ABMCs) were transplanted intrathecally to investigate the safety and efficacy of autologous ABMC therapy. The effects of ABMC transplantation in dogs with SCI were determined using functional neurological scoring, and the integration of ABMCs into the injured cords was determined using histopathological and immunohistochemical investigations and electron microscopic analyses of sections from control and transplanted spinal cords. Our data demonstrate the presence of GFP-labeled cells in the injured spinal cord for up to 16 weeks after transplantation in the subacute SCI stage. GFP-labeled cells homed to the site of injury and were detected around white matter tracts and surviving axons. ABMC therapy in the canine SCI model enhanced remyelination and augmented neural regeneration, resulting in improved neurological functions. Therefore, autologous ABMC therapy appears to be a safe and promising therapy for spinal cord injuries.
Collapse
Affiliation(s)
- Hala Gabr
- Department of Hematology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Glavaski-Joksimovic A, Bohn MC. Mesenchymal stem cells and neuroregeneration in Parkinson's disease. Exp Neurol 2013; 247:25-38. [DOI: 10.1016/j.expneurol.2013.03.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/14/2013] [Indexed: 02/06/2023]
|
16
|
Suhr F, Delhasse Y, Bungartz G, Schmidt A, Pfannkuche K, Bloch W. Cell biological effects of mechanical stimulations generated by focused extracorporeal shock wave applications on cultured human bone marrow stromal cells. Stem Cell Res 2013; 11:951-64. [PMID: 23880536 DOI: 10.1016/j.scr.2013.05.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 03/20/2013] [Accepted: 05/18/2013] [Indexed: 12/13/2022] Open
Abstract
Human bone marrow stromal cells (hBMSCs) bear tremendous clinical potential due to their immunomodulatory properties in transplantation settings and their contribution to tissue regeneration. In fact, they are among the most promising types of stem-like cells for therapeutic applications and are the subject of intense research. However, the clinical use of hBMSCs has been confounded by limitations in their availability; they are scarce cells cumbersome to isolate and purify. Additionally, they are difficult to target to the site of injury in regeneration experiments. In order to combat these limitations, focused extracorporeal shock waves (fESW, 0.2/0.3mJ∗mm(-2)) were applied to purified, cultured hBMSCs. fESW (0.2mJ∗mm(-2)) stimulations were found to increase hBMSCs' growth rate (p<0.05), proliferation (p<0.05), migration, cell tracking and wound healing (p<0.05, respectively), as well as to reduce the rate of apoptosis activation (p<0.05). The increase in hBMSC migration behavior was found to be mediated by active remodeling of the actin cytoskeleton as indicated by increased directed stress fiber formations (p<0.05). Furthermore, hBMSCs maintain their differentiation potentials after fESW treatment, whereas 0.2mJ∗mm(-2) is the most effective application. In conclusion, our results establish first-timely that hBMSCs' behavior can be modified and optimized in response to defined mechanical stimulation. These findings appear particularly promising as they suggest that mechanical stress preconditions hBMSCs for improved therapeutic performance without genetic manipulations and that mechanically preconditioned hBMSCs will be advantageous for hBMSC-based tissue regeneration. Therefore, this approach opens the door for exploiting the full potential of these cells in regenerative medicine.
Collapse
Affiliation(s)
- Frank Suhr
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Cologne, Germany.
| | | | | | | | | | | |
Collapse
|
17
|
Characteristics and neural-like differentiation of mesenchymal stem cells derived from foetal porcine bone marrow. Biosci Rep 2013; 33:e00032. [PMID: 23458182 PMCID: PMC3610297 DOI: 10.1042/bsr20120023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MSCs (mesenchymal stem cells) are a stem cell source that can be easily obtained from bone marrow. Despite the increasing importance of the pig as a large animal model, little is known about foetal pMSCs (porcine MSCs). In this study, we observed the gene expression of pluripotent markers in foetal pMSCs and the capacity of pMSCs to differentiate into adipocytes, osteocytes and neural-like cells using quantitative RT–PCR (reverse transcription–PCR), normal histological staining and immunohistochemistry. Foetal pMSCs have either a spindle or a flattened shape, and flow cytometry revealed the expression of the MSC-related proteins CD44 and CD105 (endoglin) but not CD34 and CD45. pMSCs express pluripotent markers such as Oct4 (octamer-binding transcription factor 4) and Nanog at the protein and mRNA levels. qRT-PCR (quantitative real-time PCR) analyses revealed that pMSCs expressed nestin [for NSCs (neural stem cells)]. Immunocytochemical and RT–PCR data showed that 29% and 23% of pMSCs expressed MAP2 (microtubule-associated protein 2) for neurons and β-tubulin III (Tuj1) for immature neurons, respectively, after induction of neural differentiation. These findings demonstrate the plasticity of pMSCs and their potential for use in cellular replacement therapy for neural diseases.
Collapse
|
18
|
El-Kheir WA, Gabr H, Awad MR, Ghannam O, Barakat Y, Farghali HAMA, El Maadawi ZM, Ewes I, Sabaawy HE. Autologous bone marrow-derived cell therapy combined with physical therapy induces functional improvement in chronic spinal cord injury patients. Cell Transplant 2013; 23:729-45. [PMID: 23452836 DOI: 10.3727/096368913x664540] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injuries (SCI) cause sensory loss and motor paralysis. They are normally treated with physical therapy, but most patients fail to recover due to limited neural regeneration. Here we describe a strategy in which treatment with autologous adherent bone marrow cells is combined with physical therapy to improve motor and sensory functions in early stage chronic SCI patients. In a phase I/II controlled single-blind clinical trial (clinicaltrials.gov identifier: NCT00816803), 70 chronic cervical and thoracic SCI patients with injury durations of at least 12 months were treated with either intrathecal injection(s) of autologous adherent bone marrow cells combined with physical therapy or with physical therapy alone. Patients were evaluated with clinical and neurological examinations using the American Spinal Injury Association (ASIA) Impairment Scale (AIS), electrophysiological somatosensory-evoked potential, magnetic resonance imaging (MRI), and functional independence measurements. Chronic cervical and thoracic SCI patients (15 AIS A and 35 AIS B) treated with autologous adherent bone marrow cells combined with physical therapy showed functional improvements over patients in the control group (10 AIS A and 10 AIS B) treated with physical therapy alone, and there were no long-term cell therapy-related side effects. At 18 months posttreatment, 23 of the 50 cell therapy-treated cases (46%) showed sustained functional improvement. Compared to those patients with cervical injuries, a higher rate of functional improvement was achieved in thoracic SCI patients with shorter durations of injury and smaller cord lesions. Therefore, when combined with physical therapy, autologous adherent bone marrow cell therapy appears to be a safe and promising therapy for patients with chronic SCI of traumatic origin. Randomized controlled multicenter trials are warranted.
Collapse
|
19
|
Shi H, Zhang T, Qiang L, Man L, Shen Y, Ding F. Mesenspheres of neural crest-derived cells enriched from bone marrow stromal cell subpopulation. Neurosci Lett 2012; 532:70-5. [PMID: 23127856 DOI: 10.1016/j.neulet.2012.10.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/16/2012] [Accepted: 10/18/2012] [Indexed: 10/27/2022]
Abstract
Neural crest-derived cells (NCCs) can be used for cell replacement therapy of neurodegenerative diseases and nerve injury, and it is of significance to open readily accessible tissue sources for NCCs due to their insufficient supply. In this study, we aimed to examine the possibility of enriching NCCs from bone marrow stromal cell (BMSC) subpopulation. The epidermal growth factor/fibroblast growth factor-2 (EGF/FGF2)-responsive BMSC subpopulation (BMSC-C2) was isolated from rat bone marrow by repetitive two-step condition culture. The BMSC-C2 subpopulation showed a long-term proliferative capacity and high cell growth rate, and possessed a significant sphere-forming ability. The mesenspheres derived from BMSC-C2 subpopulation were self-renewable and could express NCC markers, such as CD29, CD44, nestin, CD133 and p75(NTR). In particular, the mesenspheres could be induced to differentiate into neuron- and glia-like cells in vitro. Collectively, our results might provide a basis for in-depth studies of recruiting postmigratory NCCs from bone marrow and various neural crest-derived tissues.
Collapse
Affiliation(s)
- Haiyan Shi
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou JS 215123, China
| | | | | | | | | | | |
Collapse
|
20
|
Wen SR, Qi HP, Ren YJ, Liu GJ, Gong FC, Zhong H, Bi S. Expression of δNp73 in hippocampus of APP/PS1 transgenic mice following GFP-BMSCs transplantation. Neurol Res 2012; 33:1109-14. [PMID: 22196765 DOI: 10.1179/1743132811y.0000000051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To study the effect of hippocampal bone marrow stromal cells (GFP-BMSCs) transplantation on spatial memory and DeltaNp73 expression in APP/PS1 transgenic mice. METHODS Twelve APP/PS1 transgenic mice randomly received either 10 μl GFP-BMSCs suspension in medium (GFP-BMSCs transplantation group) or 10 μl complete medium (sham-operated group). Learning and memory function of mice in both groups were observed and tested in Morris water maze experiment at 2 weeks after surgery. Senile plaques and DeltaNp73 protein in hippocampuses were determined by immunohistochemistry and western blot at 3 weeks after surgery, respectively. RESULTS APP/PS1 mice treated with BMSCs performed significantly better on the water maze test than those in sham-operated group (P<0·05). Immunohistochemistry showed that GFP-BMSCs distributed uniformly and the number of Alzheimer's senile plaques reduced after transplantation. Western blot showed that quantified DeltaNp73 protein expression was significantly higher in BMSCs transplantation group when compared with sham-operated group (P<0·01). CONCLUSIONS Our results suggest that BMSCs transplatation could retard Alzheimer's disease (AD) like pathology and upregulate DeltaNp73 expression in hippocampuses of APP/PS1 transgenic mice. GFP-BMSCs transplantation will be a potential treatment for AD.
Collapse
Affiliation(s)
- Shi-Rong Wen
- The First Affiliated Hospital of Harbin Medical University, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Dupin E, Sommer L. Neural crest progenitors and stem cells: from early development to adulthood. Dev Biol 2012; 366:83-95. [PMID: 22425619 DOI: 10.1016/j.ydbio.2012.02.035] [Citation(s) in RCA: 176] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Accepted: 02/29/2012] [Indexed: 01/09/2023]
Abstract
In the vertebrate embryo, the neural crest forms transiently in the dorsal neural primordium to yield migratory cells that will invade nearly all tissues and later, will differentiate into bones and cartilages, neurons and glia, endocrine cells, vascular smooth muscle cells and melanocytes. Due to the amazingly diversified array of cell types it produces, the neural crest is an attractive model system in the stem cell field. We present here in vivo and in vitro studies of single cell fate, which led to the discovery and the characterization of stem cells in the neural crest of avian and mammalian embryos. Some of the key issues in neural crest cell diversification are discussed, such as the time of segregation of mesenchymal vs. neural/melanocytic lineages, and the origin and close relationships between the glial and melanocytic lineages. An overview is also provided of the diverse types of neural crest-like stem cells and progenitors, recently identified in a growing number of adult tissues in animals and humans. Current and future work, in which in vivo lineage studies and the use of injury models will complement the in vitro culture analysis, should help in unraveling the properties and function of neural crest-derived progenitors in development and disease.
Collapse
Affiliation(s)
- Elisabeth Dupin
- INSERM U894 Equipe Plasticité Gliale, Centre de Psychiatrie et de Neuroscience, 2 ter Rue d'Alésia 75014 Paris, France.
| | | |
Collapse
|
22
|
Isolation, culture and chondrogenic differentiation of canine adipose tissue- and bone marrow-derived mesenchymal stem cells--a comparative study. Vet Res Commun 2012; 36:139-48. [PMID: 22392598 DOI: 10.1007/s11259-012-9523-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2012] [Indexed: 12/29/2022]
Abstract
In the dog, mesenchymal stem cells (MSCs) have been shown to reside in the bone marrow (bone marrow-derived mesenchymal stem cells: BM-MSCs) as well as in the adipose tissue (adipose tissue-derived stem cells: ADSCs). Potential application fields for these multipotent MSCs in small animal practice are joint diseases as MSCs of both sources have shown to possess chondrogenic differentiation ability. However, it is not clear whether the chondrogenic differentiation potential of cells of these two distinct tissues is truly equal. Therefore, we compared MSCs of both origins in this study in terms of their chondrogenic differentiation ability and suitability for clinical application. BM-MSCs harvested from the femoral neck and ADSCs from intra-abdominal fat tissue were examined for their morphology, population doubling time (PDT) and CD90 surface antigen expression. RT-PCR served to assess expression of pluripotency marker Oct4 and early differentiation marker genes. Chondrogenic differentiation ability was compared and validated using histochemistry, transmission electron microscopy (TEM) and quantitative RT-PCR. Both cell populations presented a highly similar morphology and marker expression in an undifferentiated stage except that freshly isolated ADSCs demonstrated a significantly faster PDT than BM-MSCs. In contrast, BM-MSCs revealed a morphological superior cartilage formation by the production of a more abundant and structured hyaline matrix and higher expression of lineage specific genes under the applied standard differentiation protocol. However, further investigations are necessary in order to find out if chondrogenic differentiation can be improved in canine ADSCs using different protocols and/or supplements.
Collapse
|
23
|
Whone AL, Kemp K, Sun M, Wilkins A, Scolding NJ. Human bone marrow mesenchymal stem cells protect catecholaminergic and serotonergic neuronal perikarya and transporter function from oxidative stress by the secretion of glial-derived neurotrophic factor. Brain Res 2012; 1431:86-96. [DOI: 10.1016/j.brainres.2011.10.038] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 10/18/2011] [Accepted: 10/20/2011] [Indexed: 12/29/2022]
|
24
|
Lee ES, Yu SH, Jang YJ, Hwang DY, Jeon CJ. Transplantation of bone marrow-derived mesenchymal stem cells into the developing mouse eye. Acta Histochem Cytochem 2011; 44:213-21. [PMID: 22096261 PMCID: PMC3210426 DOI: 10.1267/ahc.11009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 07/06/2011] [Indexed: 12/23/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been studied widely for their potential to differentiate into various lineage cells including neural cells in vitro and in vivo. To investigate the influence of the developing host environment on the integration and morphological and molecular differentiation of MSCs, human bone marrow-derived mesenchymal stem cells (BM-MSCs) were transplanted into the developing mouse retina. Enhanced green fluorescent protein (GFP)-expressing BM-MSCs were transplanted by intraocular injections into mice, ranging in ages from 1 day postnatal (PN) to 10 days PN. The survival dates ranged from 7 days post-transplantation (DPT) to 28DPT, at which time an immunohistochemical analysis was performed on the eyes. The transplanted BM-MSCs survived and showed morphological differentiation into neural cells and some processes within the host retina. Some transplanted cells expressed microtubule associated protein 2 (MAP2ab, marker for mature neural cells) or glial fibrillary acid protein (GFAP, marker for glial cells) at 5PN 7DPT. In addition, some transplanted cells integrated into the developing retina. The morphological and molecular differentiation and integration within the 5PN 7DPT eye was greater than those of other-aged host eye. The present findings suggest that the age of the host environment can strongly influence the differentiation and integration of BM-MSCs.
Collapse
Affiliation(s)
- Eun-Shil Lee
- Department of Biology, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University
| | - Song-Hee Yu
- Department of Biology, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University
| | - Yu-Jin Jang
- Department of Biology, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University
| | - Dong-Youn Hwang
- Department of Biomedical Science, College of Life Science, CHA University
| | - Chang-Jin Jeon
- Department of Biology, College of Natural Sciences, and Brain Science and Engineering Institute, Kyungpook National University
| |
Collapse
|
25
|
Grigoriadis N, Lourbopoulos A, Lagoudaki R, Frischer JM, Polyzoidou E, Touloumi O, Simeonidou C, Deretzi G, Kountouras J, Spandou E, Kotta K, Karkavelas G, Tascos N, Lassmann H. Variable behavior and complications of autologous bone marrow mesenchymal stem cells transplanted in experimental autoimmune encephalomyelitis. Exp Neurol 2011; 230:78-89. [PMID: 21440544 DOI: 10.1016/j.expneurol.2011.02.021] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 12/17/2010] [Accepted: 02/15/2011] [Indexed: 12/13/2022]
Abstract
Autologous bone marrow stromal cells (BMSCs) offer significant practical advantages for potential clinical applications in multiple sclerosis (MS). Based on recent experimental data, a number of clinical trials have been designed for the intravenous (IV) and/or intrathecal (ITH) administration of BMSCs in MS patients. Delivery of BMSCs in the cerebrospinal fluid via intracerebroventricular (ICV) transplantation is a useful tool to identify mechanisms underlying the migration and function of these cells. In the current study, BMSCs were ICV administered in severe and mild EAE, as well as naive animals; neural precursor cells (NPCs) served as cellular controls. Our data indicated that ICV-transplanted BMSCs significantly ameliorated mild though not severe EAE. Moreover, BMSCs exerted significant anti-inflammatory effect on spinal cord with concomitant reduced axonopathy only in the mild EAE model. BMSCs migrated into the brain parenchyma and, depending on their cellular density, within brain parenchyma formed cellular masses characterized by focal inflammation, demyelination, axonal loss and increased collagen-fibronectin deposition. These masses were present in 64% of ICV BMASC-transplanted severe EAE animals whereas neither BMSCs transplanted in mild EAE cases nor the NPCs exhibited similar behavior. BMSCs possibly exerted their fibrogenic effect via both paracrine and autocrine manner, at least partly due to up-regulation of connective tissue growth factor (CTGF) under the trigger of TGFb1. Our findings are of substantial relevance for clinical trials in MS, particularly regarding the possibility that ICV transplanted BMSCs entering the inflamed central nervous system may exhibit - under conditions - a local pathology of yet unknown consequences.
Collapse
Affiliation(s)
- Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Macedonia, Greece.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Skouras E, Ozsoy U, Sarikcioglu L, Angelov DN. Intrinsic and therapeutic factors determining the recovery of motor function after peripheral nerve transection. Ann Anat 2011; 193:286-303. [PMID: 21458252 DOI: 10.1016/j.aanat.2011.02.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 02/03/2011] [Accepted: 02/04/2011] [Indexed: 01/01/2023]
Abstract
Insufficient recovery after peripheral nerve injury has been attributed to (i) poor pathfinding of regrowing axons, (ii) excessive collateral axonal branching at the lesion site and (iii) polyneuronal innervation of the neuromuscular junctions (NMJ). The facial nerve transection model has been used initially to measure restoration of function after varying therapies and to examine the mechanisms underlying their effects. Since it is very difficult to control the navigation of several thousand axons, efforts concentrated on collateral branching and NMJ-polyinnervation. Treatment with antibodies against trophic factors to combat branching improved the precision of reinnervation, but had no positive effects on functional recovery. This suggested that polyneuronal reinnervation--rather than collateral branching--may be the critical limiting factor. The former could be reduced by pharmacological agents known to perturb microtubule assembly and was followed by recovery of function. Because muscle polyinnervation is activity-dependent and can be manipulated, attempts to design a clinically feasible therapy were performed by electrical stimulation or by soft tissue massage. Electrical stimulation applied to the transected facial nerve or to paralysed facial muscles did not improve vibrissal motor performance and failed to diminish polyinnervation. In contrast, gentle stroking of the paralysed muscles (vibrissal, orbicularis oculi, tongue musculature) resulted in full recovery of function. This manual stimulation was also effective after hypoglossal-facial nerve suture and after interpositional nerve grafting, but not after surgical reconstruction of the median nerve. All these findings raise hopes that clinically feasible and effective therapies could be soon designed and tested.
Collapse
Affiliation(s)
- Emmanouil Skouras
- Department of Orthopedics and Traumatology, University of Cologne, Joseph-Stelzmann-Strasse 9, Cologne, Germany
| | | | | | | |
Collapse
|
27
|
Arnhold S, Glüer S, Hartmann K, Raabe O, Addicks K, Wenisch S, Hoopmann M. Amniotic-Fluid Stem Cells: Growth Dynamics and Differentiation Potential after a CD-117-Based Selection Procedure. Stem Cells Int 2011; 2011:715341. [PMID: 21437196 PMCID: PMC3062157 DOI: 10.4061/2011/715341] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 10/25/2010] [Accepted: 01/09/2011] [Indexed: 12/16/2022] Open
Abstract
Amniotic fluid (AF) has become an interesting source of fetal stem cells. However, AF contains heterogeneous and multiple, partially differentiated cell types. After isolation from the amniotic fluid, cells were characterized regarding their morphology and growth dynamics. They were sorted by magnetic associated cell sorting using the surface marker CD 117. In order to show stem cell characteristics such as pluripotency and to evaluate a possible therapeutic application of these cells, AF fluid-derived stem cells were differentiated along the adipogenic, osteogenic, and chondrogenic as well as the neuronal lineage under hypoxic conditions. Our findings reveal that magnetic associated cell sorting (MACS) does not markedly influence growth characteristics as demonstrated by the generation doubling time. There was, however, an effect regarding an altered adipogenic, osteogenic, and chondrogenic differentiation capacity in the selected cell fraction. In contrast, in the unselected cell population neuronal differentiation is enhanced.
Collapse
Affiliation(s)
- S Arnhold
- Department of Veterinary Anatomy, Justus-Liebig-University Giessen, Frankfurter Straße 98, 35392 Giessen, Germany
| | | | | | | | | | | | | |
Collapse
|
28
|
Inflammatory cytokine induced regulation of superoxide dismutase 3 expression by human mesenchymal stem cells. Stem Cell Rev Rep 2011; 6:548-59. [PMID: 20683679 DOI: 10.1007/s12015-010-9178-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Increasing evidence suggests that bone marrow derived-mesenchymal stem cells (MSCs) have neuroprotective properties and a major mechanism of action is through their capacity to secrete a diverse range of potentially neurotrophic or anti-oxidant factors. The recent discovery that MSCs secrete superoxide dismutase 3 (SOD3) may help explain studies in which MSCs have a direct anti-oxidant activity that is conducive to neuroprotection in both in vivo and in vitro. SOD3 attenuates tissue damage and reduces inflammation and may confer neuroprotective effects against nitric oxide-mediated stress to cerebellar neurons; but, its role in relation to central nervous system inflammation and neurodegeneration has not been extensively investigated. Here we have performed a series of experiments showing that SOD3 secretion by human bone marrow-derived MSCs is regulated synergistically by the inflammatory cytokines TNF-alpha and IFN-gamma, rather than through direct exposure to reactive oxygen species. Furthermore, we have shown SOD3 secretion by MSCs is increased by activated microglial cells. We have also shown that MSCs and recombinant SOD are able to increase both neuronal and axonal survival in vitro against nitric oxide or microglial induced damage, with an increased MSC-induced neuroprotective effect evident in the presence of inflammatory cytokines TNF-alpha and IFN-gamma. We have shown MSCs are able to convey these neuroprotective effects through secretion of soluble factors alone and furthermore demonstrated that SOD3 secretion by MSCs is, at least, partially responsible for this phenomenon. SOD3 secretion by MSCs maybe of relevance to treatment strategies for inflammatory disease of the central nervous system.
Collapse
|
29
|
Witherick J, Wilkins A, Scolding N, Kemp K. Mechanisms of oxidative damage in multiple sclerosis and a cell therapy approach to treatment. Autoimmune Dis 2010; 2011:164608. [PMID: 21197107 PMCID: PMC3010615 DOI: 10.4061/2011/164608] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 11/10/2010] [Indexed: 12/14/2022] Open
Abstract
Although significant advances have recently been made in the understanding and treatment of multiple sclerosis, reduction of long-term disability remains a key goal. Evidence suggests that inflammation and oxidative stress within the central nervous system are major causes of ongoing tissue damage in the disease. Invading inflammatory cells, as well as resident central nervous system cells, release a number of reactive oxygen and nitrogen species which cause demyelination and axonal destruction, the pathological hallmarks of multiple sclerosis. Reduction in oxidative damage is an important therapeutic strategy to slow or halt disease processes. Many drugs in clinical practice or currently in trial target this mechanism. Cell-based therapies offer an alternative source of antioxidant capability. Classically thought of as being important for myelin or cell replacement in multiple sclerosis, stem cells may, however, have a more important role as providers of supporting factors or direct attenuators of the disease. In this paper we focus on the antioxidant properties of mesenchymal stem cells and discuss their potential importance as a cell-based therapy for multiple sclerosis.
Collapse
Affiliation(s)
- Jonathan Witherick
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| | - Alastair Wilkins
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| | - Neil Scolding
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| | - Kevin Kemp
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol BS16 1LE, UK
| |
Collapse
|
30
|
Kemp K, Hares K, Mallam E, Heesom KJ, Scolding N, Wilkins A. Mesenchymal stem cell-secreted superoxide dismutase promotes cerebellar neuronal survival. J Neurochem 2010; 114:1569-80. [PMID: 20028455 DOI: 10.1111/j.1471-4159.2009.06553.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It has been postulated that bone marrow-derived mesenchymal stem cells (MSCs) might be effective treatments for neurodegenerative disorders either by replacement of lost cells by differentiation into functional neural tissue; modulation of the immune system to prevent further neurodegeneration; and/or provision of trophic support for the diseased nervous system. Here we have performed a series of experiments showing that human bone marrow-derived MSCs are able to protect cultured rodent cerebellar neurons, and specifically cells expressing Purkinje cell markers, against either nitric oxide exposure or withdrawal of trophic support via cell-cell contact and/or secretion of soluble factors, or through secretion of soluble factors alone. We have demonstrated that MSCs protect cerebellar neurons against toxic insults via modulation of both the phosphatidylinositol 3-kinase/Akt and MAPK pathways and defined superoxide dismutase 3 as a secreted active antioxidant biomolecule by which MSCs modulate, at least in part, their neuroprotective effect on cerebellar cells in vitro. Together, the results demonstrate new and specific mechanisms by which MSCs promote cerebellar neuronal survival and add further evidence to the concept that MSCs may be potential therapeutic agents for neurological disorders involving the cerebellum.
Collapse
Affiliation(s)
- Kevin Kemp
- Multiple Sclerosis and Stem Cell Group, Institute of Clinical Neurosciences, Clinical Sciences North Bristol, University of Bristol, Bristol, UK.
| | | | | | | | | | | |
Collapse
|
31
|
Baumgartner L, Arnhold S, Brixius K, Addicks K, Bloch W. Human mesenchymal stem cells: Influence of oxygen pressure on proliferation and chondrogenic differentiation in fibrin glue in vitro. J Biomed Mater Res A 2010; 93:930-40. [PMID: 19708077 DOI: 10.1002/jbm.a.32577] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Tissue engineering using biomaterials is a promising solution for cartilage replacement. The purpose of this study was to investigate whether the fibrin sealant Tissucol(R) provides a suitable scaffold for re-implanting stem cells during chondrogenic replacement therapy. Pluripotent stem cells were isolated from adult human bone marrow (hMSCs), cultured and characterized by FACS (CD105+/CD106+, CD45-/CD14-/CD34-). A large-holed porous hMSC-containing fibrin matrix was built that allowed hMSCs to survive throughout the period of culture (42 days) in either proliferation or chondrogenic differentiation medium under normoxic (21% O2) or hypoxic (3% O2) conditions. Morphology (as determined by electron microscopy) and proliferation (Ki67 staining) of the embedded hMSCs did not markedly vary under normoxic and hypoxic culture even after 42 days in culture. The stem cell marker Oct-4 was expressed during the whole culture period. Under chondrogenic differentiation conditions, especially under hypoxic conditions, we observed rounded chondrocyte-like cell types and a chondral phenotype assessed by mRNA expression of collagen II and Alcian blue staining. hMSCs seeded into large-holed porous preparations of Tissucol survive, proliferate and keep their stem cell character. Furthermore, culturing the cells in a corresponding medium induces chondrogenic differentiation, which could be remarkably and significantly enhanced under hypoxic conditions.
Collapse
Affiliation(s)
- Laura Baumgartner
- Department of Molecular and Cellular Sport Medicine, Institute for Circulation Research and Sport Medicine, Cologne, Germany.
| | | | | | | | | |
Collapse
|
32
|
Qi X, Shao M, Peng H, Bi Z, Su Z, Li H. In vitro differentiation of bone marrow stromal cells into neurons and glial cells and differential protein expression in a two-compartment bone marrow stromal cell/neuron co-culture system. J Clin Neurosci 2010; 17:908-13. [DOI: 10.1016/j.jocn.2009.10.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 10/13/2009] [Indexed: 12/14/2022]
|
33
|
Intranigral transplantation of epigenetically induced BDNF-secreting human mesenchymal stem cells: implications for cell-based therapies in Parkinson's disease. Biol Blood Marrow Transplant 2010; 16:1530-40. [PMID: 20542127 DOI: 10.1016/j.bbmt.2010.06.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 06/01/2010] [Indexed: 12/12/2022]
Abstract
It is thought that the ability of human mesenchymal stem cells (hMSC) to deliver neurotrophic factors might be potentially useful for the treatment of neurodegenerative disorders. The aim of the present study was to characterize signals and/or molecules that regulate brain-derived neurotrophic factor (BDNF) protein expression/delivery in hMSC cultures and evaluate the effect of epigenetically generated BDNF-secreting hMSC on the intact and lesioned substantia nigra (SN). We tested 4 different culture media and found that the presence of fetal bovine serum (FBS) decreased the expression of BDNF, whereas exogenous addition of epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) to serum-free medium was required to induce BDNF release (125 ± 12 pg/day/10⁶ cells). These cells were called hM(N)SC. Although the induction medium inhibited the expression of alpha smooth muscle actin (ASMA), an hMSC marker, and increased the nestin-positive subpopulation of hMSC cultures, the ability to express BDNF was restricted to the nestin-negative subpopulation. One week after transplantation into the SN, the human cells integrated into the surrounding tissue, and some showed a dopaminergic phenotype. We also observed the activation of Trk receptors for neurotrophic factors around the implant site, including the BDNF receptor TrkB. When we transplanted these cells into the unilateral lesioned SN induced by striatal injection of 6-hydroxydopamine (6-OHDA), a significant hypertrophy of nigral tyrosine hydroxylase (TH)(+) cells, an increase of striatal TH-staining and stabilization of amphetamine-induced motor symptoms were observed. Therefore, hMSC cultures exposed to the described induction medium might be highly useful as a vehicle for neurotrophic delivery to the brain and specifically are strong candidates for future therapeutic application in Parkinson's disease.
Collapse
|
34
|
Amelioration of diabetic retinopathy by engrafted human adipose-derived mesenchymal stem cells in streptozotocin diabetic rats. Graefes Arch Clin Exp Ophthalmol 2010; 248:1415-22. [PMID: 20437245 DOI: 10.1007/s00417-010-1384-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 03/18/2010] [Accepted: 04/02/2010] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Diabetic retinopathy is a common complication of diabetes, which is caused by injury to retinal microvasculature and neurons. Mesenchymal stem cells (MSCs), which proved to have multi-linkage differentiation capacity, including endothelial cells and neurons, might be a promising cell therapy resource. The current pilot study was performed using the streptozotocin (STZ) rat model of diabetic retinopathy injected intravenously with human adipose-derived mesenchymal stem cells (AMSCs) in an effort to investigate the potency and possible therapeutic effects of AMSCs. METHODS Four experimental groups of Wistar rats were included in the current study: an untreated control group of STZ diabetic rats (n = 10), a normal non-diabetic control group (n = 20), an AMSC therapy group of STZ diabetic rats (n = 50), and a sham group of STZ diabetic rats (n = 50). Blood glucose levels were monitored closely. Immunofluorescence was used to study AMSC distribution and differentiation. The integrity of the blood-retinal barrier (BRB) was evaluated by Evans blue dye infusion to evaluate the therapeutic effects. RESULTS After 1 week of transplantation, a significant reduction in blood glucose levels was observed in the AMSC therapy group relative to the sham group. BRB integrity was also improved, as less Evans blue dye leakage was observed. Donor cells were observed in the retinas of therapy group rats, and they expressed rhodopsin and glial fibrillary acidic protein (GFAP), specific markers for photoreceptors and astrocytes, respectively. CONCLUSIONS Taken together, the results of the current study suggest that AMSCs may improve the integrity of the BRB in diabetic rats by differentiation into photoreceptor and glial-like cells in the retina and by reducing the blood glucose levels. Furthermore, the data presented herein provide evidence that AMSCs may serve as a promising therapeutic approach for diabetic retinopathy.
Collapse
|
35
|
Kemp K, Mallam E, Scolding N, Wilkins A. Stem cells in genetic myelin disorders. Regen Med 2010; 5:425-39. [DOI: 10.2217/rme.10.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The genetic myelin disorders are a range of diseases that manifest with severe neurological problems, often from infancy. It has been postulated for some time that stem cells might be an effective treatment for these disorders, primarily as agents to restore dysfunctional or lost myelin. Stem cells, however, may offer a wider range of therapeutic potential, for instance as vehicles to replace abnormal enzymes or genes, or to provide trophic support for residual CNS tissue. This article will review several of the more common genetic myelin disorders and currently available therapies, including bone marrow transplantation for adrenoleukodystrophy. Specific stem cell subtypes and their relevance to potential therapeutic use will be discussed and stem cell transplantation in animal model studies will also be reviewed.
Collapse
Affiliation(s)
- Kevin Kemp
- MS & Stem Cell Laboratories, Burden Centre, Frenchay Hospital, Bristol, UK
- Department of Neurology, Frenchay Hospital, Bristol, UK
| | - Elizabeth Mallam
- MS & Stem Cell Laboratories, Burden Centre, Frenchay Hospital, Bristol, UK
- Department of Neurology, Frenchay Hospital, Bristol, UK
| | - Neil Scolding
- MS & Stem Cell Laboratories, Burden Centre, Frenchay Hospital, Bristol, UK
- Department of Neurology, Frenchay Hospital, Bristol, UK
| | | |
Collapse
|
36
|
Tate CC, Fonck C, McGrogan M, Case CC. Human mesenchymal stromal cells and their derivative, SB623 cells, rescue neural cells via trophic support following in vitro ischemia. Cell Transplant 2010; 19:973-84. [PMID: 20350349 DOI: 10.3727/096368910x494885] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cell transplantation is a promising treatment strategy for many neurological disorders, including stroke, which can target multiple therapeutic mechanisms in a sustained fashion. We investigated the ability of human mesenchymal stromal cells (MSCs) and MSC-derived SB623 cells to rescue neural cells via trophic support following an in vitro stroke model. Following oxygen glucose deprivation, cortical neurons or hippocampal slices were cocultured with either MSCs or SB623 cells separated by a semiporous membrane (prohibits cell-cell contact) or with MSC- or SB623 cell-conditioned medium. MSCs, SB623 cells, MSC-conditioned media, and SB623 cell-conditioned media all significantly reduced neural cell damage/death compared to untreated conditions, and the rescue effect of the conditioned media was dose dependent. We identified 11 neurotrophic factors secreted by MSCs and/or SB623 cells. This study emphasizes the importance of trophic support provided by marrow-derived cells, which likely contributes to the efficacy of cell therapy for brain injury.
Collapse
|
37
|
Kamishina H, Cheeseman JA, Farese JP, Milner RJ, Clemmons RM. Migration and differentiation of canine bone marrow stromal cells transplanted into the developing mouse brain. J Vet Med Sci 2009; 72:353-6. [PMID: 19952514 DOI: 10.1292/jvms.09-0353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To evaluate whether canine bone marrow stromal cells (BMSCs) can migrate and adopt neural phenotypes in the developing mouse brain we transplanted fluorescently labeled BMSCs into the lateral ventricle of immunocompromised neonatal mice. Most fibroblasts, used as a control, and BMSCs isolated from adult dogs remained around the injection site and exhibited a spindle-shaped appearance. A small number of BMSCs from young dogs were found in the subventricular zone, rostral migratory stream, and olfactory bulbs, and retained expression of neuron marker. Our findings suggest that BMSCs isolated from adult dogs have limited ability of migration and differentiation toward neural cells in the developing brain. Bone marrow of young dogs may contain a primitive stem cell population with neural differentiation capacity.
Collapse
Affiliation(s)
- Hiroaki Kamishina
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610-0126, USA.
| | | | | | | | | |
Collapse
|
38
|
Sadan O, Bahat-Stromza M, Barhum Y, Levy YS, Pisnevsky A, Peretz H, Ilan AB, Bulvik S, Shemesh N, Krepel D, Cohen Y, Melamed E, Offen D. Protective effects of neurotrophic factor-secreting cells in a 6-OHDA rat model of Parkinson disease. Stem Cells Dev 2009; 18:1179-90. [PMID: 19243240 DOI: 10.1089/scd.2008.0411] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stem cell-based therapy is a promising treatment for neurodegenerative diseases. In our laboratory, a novel protocol has been developed to induce bone marrow-derived mesenchymal stem cells (MSC) into neurotrophic factors- secreting cells (NTF-SC), thus combining stem cell-based therapy with the NTF-based neuroprotection. These cells produce and secrete factors such as brain-derived neurotrophic factor (BDNF) and glial cell-derived neurotrophic factor. Conditioned medium of the NTF-SC that was applied to a neuroblastoma cell line (SH-SY5Y) 1 h before exposure to the neurotoxin 6-hydroxydopamine (6-OHDA) demonstrated marked protection. An efficacy study was conducted on the 6-OHDA-induced lesion, a rat model of Parkinson's disease. The cells, either MSC or NTF-SC, were transplanted on the day of 6-OHDA administration and amphetamine-induced rotations were measured as a primary behavior index. We demonstrated that when transplanted posterior to the 6-OHDA lesion, the NTF-SC ameliorated amphetamine-induced rotations by 45%. HPLC analysis demonstrated that 6-OHDA induced dopamine depletion to a level of 21% compared to the untreated striatum. NTF-SC inhibited dopamine depletion to a level of 72% of the contralateral striatum. Moreover, an MRI study conducted with iron-labeled cells, followed by histological verification, revealed that the engrafted cells migrated toward the lesion. In a histological assessment, we found that the cells induced regeneration in the damaged striatal dopaminergic nerve terminal network. We therefore conclude that the induced MSC have a therapeutic potential for neurodegenerative processes and diseases, both by the NTFs secretion and by the migratory trait toward the diseased tissue.
Collapse
Affiliation(s)
- Ofer Sadan
- Laboratory of Neurosciences, Felsenstein Medical Research Center, Department of Neurology, Rabin Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Boucherie C, Schäfer S, Lavand'homme P, Maloteaux JM, Hermans E. Chimerization of astroglial population in the lumbar spinal cord after mesenchymal stem cell transplantation prolongs survival in a rat model of amyotrophic lateral sclerosis. J Neurosci Res 2009; 87:2034-46. [PMID: 19267424 DOI: 10.1002/jnr.22038] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adult mesenchymal stem cells (MSCs) exhibit neuroprotective properties when introduced into the degenerating central nervous system through different putative mechanisms including secretion of growth factors and transdifferentiation. In the present study, we injected MSCs into the cerebrospinal fluid of symptomatic hSOD1(G93A) rats, a transgenic animal model of familial amyotrophic lateral sclerosis (ALS) expressing a mutated form of the human superoxide dismutase. MSCs were found to infiltrate the nervous parenchyma and migrate substantially into the ventral gray matter, where motor neurons degenerate. Even though overall astrogliosis was not modified, MSCs differentiated massively into astrocytes at the site of degeneration. The intrathecal delivery of MSCs and the subsequent generation of healthy astrocytes at symptomatic stage decreased motor neuron loss in the lumbar spinal cord, preserving motor functions and extending the survival of hSOD1(G93A) rats. This neuroprotection was correlated with decreased inflammation, as shown by the lower proliferation of microglial cells and the reduced expressiontion of COX-2 and NOX-2. Together, these data highlight the protective capacity of adult MSC-derived astrocytes when grafted into the central nervous system and illustrate an attractive strategy to target excessive inflammation in ALS.
Collapse
Affiliation(s)
- Cédric Boucherie
- Institute of Neuroscience (INES), Université catholique de Louvain, Brussels, Belgium
| | | | | | | | | |
Collapse
|
40
|
Wilkins A, Kemp K, Ginty M, Hares K, Mallam E, Scolding N. Human bone marrow-derived mesenchymal stem cells secrete brain-derived neurotrophic factor which promotes neuronal survival in vitro. Stem Cell Res 2009; 3:63-70. [DOI: 10.1016/j.scr.2009.02.006] [Citation(s) in RCA: 211] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 12/03/2008] [Accepted: 02/25/2009] [Indexed: 12/19/2022] Open
|
41
|
Lee PH, Park HJ. Bone marrow-derived mesenchymal stem cell therapy as a candidate disease-modifying strategy in Parkinson's disease and multiple system atrophy. J Clin Neurol 2009; 5:1-10. [PMID: 19513327 PMCID: PMC2686892 DOI: 10.3988/jcn.2009.5.1.1] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 02/13/2009] [Accepted: 02/16/2009] [Indexed: 12/17/2022] Open
Abstract
Parkinson's disease (PD) and multiple system atrophy (MSA) are neurodegenerative diseases representative of α-synucleinopathies characterized pathologically by α-synuclein-abundant Lewy bodies and glial cytoplasmic inclusions, respectively. Embryonic stem cells, fetal mesencephalic neurons, and neural stem cells have been introduced as restorative strategies in PD animals and patients, but ethical and immunological problems as well as the serious side effects of tumorigenesis and disabling dyskinesia have limited clinical application of these stem cells. Meanwhile, cell therapy using mesenchymal stem cells (MSCs) is attractive clinically because these cells are free from ethical and immunological problems. MSCs are present in adult bone marrow and represent <0.01% of all nucleated bone marrow cells. MSCs are themselves capable of multipotency, differentiating under appropriate conditions into chondrocytes, skeletal myocytes, and neurons. According to recent studies, the neuroprotective effect of MSCs is mediated by their ability to produce various trophic factors that contribute to functional recovery, neuronal cell survival, and stimulation of endogenous regeneration and by immunoregulatory properties that not only inhibit nearly all cells participating in the immune response cell-cell-contact-dependent mechanism, but also release various soluble factors associated with immunosuppressive activity. However, the use of MSCs as neuroprotectives in PD and MSA has seldom been studied. Here we comprehensively review recent advances in the therapeutic roles of MSCs in PD and MSA, especially focusing on their neuroprotective properties and use in disease-modifying therapeutic strategies.
Collapse
Affiliation(s)
- Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | | |
Collapse
|
42
|
The effects of canine bone marrow stromal cells on neuritogenesis from dorsal root ganglion neurons in vitro. Vet Res Commun 2009; 33:645-57. [DOI: 10.1007/s11259-009-9213-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Accepted: 02/03/2009] [Indexed: 01/01/2023]
|
43
|
Torrente Y, Polli E. Mesenchymal stem cell transplantation for neurodegenerative diseases. Cell Transplant 2009; 17:1103-13. [PMID: 19181205 DOI: 10.3727/096368908787236576] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative diseases are characterized by a progressive degeneration of selective neural populations. The lack of effective treatment and the characteristic of their pathology make these diseases appropriate candidates for cell therapy. Mesenchymal stem cells (MSCs) are multipotent stem-like cells that are capable of differentiating into mesenchymal and nonmesenchymal lineages. Their regenerative capacity after in vivo transplantation into animal models of neurodegenerative diseases has suggested that they could be useful against human diseases. Human bone marrow-derived MSCs (hMSCs) can be easily amplified in vitro and their transdifferentiation has been claimed in vitro and in vivo in neural cells. There are some doubts concerning the exact mechanisms responsible for the beneficial outcome observed after MSC transplantation into neurodegenerating tissues. Possible interpretations include cell replacement, trophic factor delivery, and immunomodulation. This review mainly concerns hMSCs transplantation in neurodegenerative diseases, because it has proven to be feasible, safe, and potentially effective. Although they have been used in hundreds of clinical trials, mixed results and no functional and long-lasting integration have so far been observed. hMSCs transplantations therefore still have their "dark side." However, the challenge in well-planned clinical trials merits discussion.
Collapse
Affiliation(s)
- Yvan Torrente
- Stem Cell Laboratory, Department of Neurological Science, Fondazione IRCCS Ospedale Maggiore Policlinico, Centro Dino Ferrari, University of Milan, Milan, Italy
| | | |
Collapse
|
44
|
Bahat-Stroomza M, Barhum Y, Levy YS, Karpov O, Bulvik S, Melamed E, Offen D. Induction of adult human bone marrow mesenchymal stromal cells into functional astrocyte-like cells: potential for restorative treatment in Parkinson's disease. J Mol Neurosci 2009; 39:199-210. [PMID: 19127447 DOI: 10.1007/s12031-008-9166-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Accepted: 11/25/2008] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder with its motor phenomena due mostly to loss of dopamine-producing neurons in the substantia nigra. Pharmacological treatments aimed to increase the deficient dopaminergic neurotransmission are effective in ameliorating the cardinal symptoms, but none of these therapies is curative. It has been suggested that treatment with neurotrophic factors (NTFs) might protect and prevent death of the surviving dopaminergic neurons and induce proliferation of their axonal nerve terminals with reinnervations of the deafferented striatum. However, long-term delivery of such proteins into the CNS is problematic. We therefore aimed to differentiate ex vivo human bone marrow-derived mesenchymal stromal cells into astrocyte-like cells, capable of generating NTFs for future transplantation into basal ganglia of PD patients. Indeed, mesenchymal stromal cells treated with our novel astrocyte differentiation medium, present astrocyte-like morphology and express the astrocyte markers S100beta, glutamine synthetase and glial fibrillary acidic protein. Moreover, these astrocyte-like cells produce and secrete significant amounts of glial cell line-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and brain-derived neurotrophic factor as indicated by messenger RNA, real-time polymerase chain reaction, ELISA, and Western blot analyses. Such NTF-producing cells transplanted into the striatum of 6-hydroxydopamine-lesioned rats, a model of PD, produced a progressive reduction in the apomorphine-induced contralateral rotations as well as behavioral improvement in rotor-rod and the "sunflower seeds" eating motor tests. Histological assessments revealed that the engrafted cells survived and expressed astrocyte and human markers and acted to regenerate the damaged dopaminergic nerve terminal system. Findings indicate that our novel procedure to induce NTF-producing astrocyte-like cells derived from human bone marrow stromal cells might become a promising and feasible autologous transplantation strategy for PD.
Collapse
Affiliation(s)
- Merav Bahat-Stroomza
- Laboratory of Neurosciences, Felsenstein Medical Research, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The characteristic CNS responses to injury including increased cell production and attempts at regenerative repair - implicitly predicted where not directly demonstrated by Cajal, but only now more fully confirmed - have important implications for regenerative therapies. Spontaneous CNS cell replacement compares poorly with the regenerative functional repair seen elsewhere, but harnessing, stimulating or supplementing this process represents a new and attractive therapeutic concept.Stem cells, traditionally defined as clone-forming, self-renewing, pluripotent progenitor cells, have already proved themselves to be an invaluable source of transplantation material in several clinical settings, most notably haematological malignancy, and attention is now turning to a wider variety of diseases in which there may be potential for therapeutic intervention with stem cell transplantation. Neurological diseases, with their reputation for relentless progression and incurability are particularly tantalising targets. The optimal source of stem cells remains to be determined but bone marrow stem cells may themselves be included amongst the contenders.Any development of therapies using stem cells must depend on an underlying knowledge of their basic biology. The haemopoietic system has long been known to maintain circulating populations of cells with short life spans, and this system has greatly informed our knowledge of stem cell biology. In particular, it has helped yield the traditional stem cell model - a hierarchical paradigm of progressive lineage restriction. As cells differentiate, their fate choices become progressively more limited, and their capacity for proliferation reduced, until fully differentiated, mitotically quiescent cells are generated. Even this, however, is now under challenge.
Collapse
Affiliation(s)
- C M Rice
- University of Bristol Institute of Clinical Neurosciences, Frenchay Hospital, Bristol, UK
| | | |
Collapse
|
46
|
Transdifferentiation of stem cells: a critical view. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2009; 114:73-106. [PMID: 19343303 DOI: 10.1007/10_2008_49] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recently a large amount of new data on the plasticity of stem cells of various lineages have emerged, providing new perspectives especially for the therapeutic application of adult stem cells. Previously unknown possibilities of cell differentiation beyond the known commitment of a given stem cell have been described using keywords such as "blood to liver," or "bone to brain." Controversies on the likelihood, as well as the biological significance, of these conversions almost immediately arose within this young field of stem cell biology. This chapter will concentrate on these controversies and focus on selected examples demonstrating the technical aspects of stem cell transdifferentiation and the evaluation of the tools used to analyze these events.
Collapse
|
47
|
El Seady R, Huisman MA, Löwik CWGM, Frijns JHM. Uncomplicated differentiation of stem cells into bipolar neurons and myelinating glia. Biochem Biophys Res Commun 2008; 376:358-62. [PMID: 18789899 DOI: 10.1016/j.bbrc.2008.08.166] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Accepted: 08/28/2008] [Indexed: 02/07/2023]
Abstract
Epidermal neural crest stem cells (EPI-NCSCs), derived from the bulge of hair follicles, appear to be promising donor stem cell candidates. In the current work, EPI-NCSCs were harvested from rodents and humans. Isolation procedures revealed high levels of nestin-positive neural stem cells and the percentage of human neural stem cells (95+/-0.6%) is even higher than the percentage found in cultures of hair follicles from rodents (90+/-0.9%). Furthermore, differentiation of EPI-NCSCs into bipolar neurons, myelinating Schwann cells and oligodendrocytes occurred by applying a simple and straightforward method. Bipolar neurons could be obtained by culturing on a collagen matrix and are of great interest for auditory neuron regeneration since auditory neurons are bipolar. We propose that this type of stem cells, would make an excellent model for autologous transplantation and offers great potential for neural regeneration in diseases, such as Parkinson's and Alzheimer's disease.
Collapse
Affiliation(s)
- Ragad El Seady
- Department of Otorhinolaryngology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | | | | | | |
Collapse
|
48
|
Migration of Neurotrophic Factors-Secreting Mesenchymal Stem Cells Toward a Quinolinic Acid Lesion as Viewed by Magnetic Resonance Imaging. Stem Cells 2008; 26:2542-51. [DOI: 10.1634/stemcells.2008-0240] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
49
|
Abstract
Typically patients with multiple sclerosis (MS) experience acute episodes of neurological dysfunction, which recover followed, at a later stage, by slow and insidious accumulation of disability (disease progression). Disease progression reflects axon damage and loss within the central nervous system. However, the precise mechanism of axon injury in MS is not clear. Inflammation occurring during acute relapses undoubtedly causes some degree of acute axon damage, but epidemiological data and treatment studies have suggested that inflammation alone is not the sole cause of axonopathy. Indeed, there appears to be dissociation between inflammation and disease progression once a certain level of clinical disability has been reached because immune suppression in patients who have established disease progression does not halt the slow decrease of function. The slow and insidious loss of neurological function that occurs during the progressive phase of the disease implies a degenerative process. Whether axon drop-out occurs at these later stages because of previous inflammatory damage to axons; because of low grade inflammation causing damage to already vulnerable demyelinated axons; because of loss of trophic environment for axons to survive; or as part of a completely independent neurodegenerative process is not clear. Understanding disease mechanisms involved in the axonopathy of MS allows for the development of rational therapies for disease progression.
Collapse
Affiliation(s)
- A Wilkins
- Department of Neurology, Institute of Clinical Neurosciences, University of Bristol, Frenchay Hospital, Bristol, UK.
| | | |
Collapse
|
50
|
Grosheva M, Guntinas-Lichius O, Arnhold S, Skouras E, Kuerten S, Streppel M, Angelova SK, Wewetzer K, Radtke C, Dunlop SA, Angelov DN. Bone marrow-derived mesenchymal stem cell transplantation does not improve quality of muscle reinnervation or recovery of motor function after facial nerve transection in rats. Biol Chem 2008; 389:873-88. [DOI: 10.1515/bc.2008.100] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AbstractRecently, we devised and validated a novel strategy in rats to improve the outcome of facial nerve reconstruction by daily manual stimulation of the target muscles. The treatment resulted in full recovery of facial movements (whisking), which was achieved by reducing the proportion of pathologically polyinnervated motor endplates. Here, we posed whether manual stimulation could also be beneficial after a surgical procedure potentially useful for treatment of large peripheral nerve defects, i.e., entubulation of the transected facial nerve in a conduit filled with suspension of isogeneic bone marrow-derived mesenchymal stem cells (BM-MSCs) in collagen. Compared to control treatment with collagen only, entubulation with BM-MSCs failed to decrease the extent of collateral axonal branching at the lesion site and did not improve functional recovery. Post-operative manual stimulation of vibrissal muscles also failed to promote a better recovery following entubulation with BM-MSCs. We suggest that BM-MSCs promote excessive trophic support for regenerating axons which, in turn, results in excessive collateral branching at the lesion site and extensive polyinnervation of the motor endplates. Furthermore, such deleterious effects cannot be overridden by manual stimulation. We conclude that entubulation with BM-MSCs is not beneficial for facial nerve repair.
Collapse
|