51
|
Generation of neurospheres from human adipose-derived stem cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:743714. [PMID: 25815334 PMCID: PMC4357140 DOI: 10.1155/2015/743714] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/24/2015] [Accepted: 02/10/2015] [Indexed: 01/29/2023]
Abstract
Transplantation of neural stem cells (NSCs) to treat neurodegenerative disease shows promise; however, the clinical application of NSCs is limited by the invasive procurement and ethical concerns. Adipose-derived stem cells (ADSCs) are a source of multipotent stem cells that can self-renew and differentiate into various kinds of cells; this study intends to generate neurospheres from human ADSCs by culturing ADSCs on uncoated culture flasks in serum-free neurobasal medium supplemented with B27, basic fibroblast growth factor (bFGF), and epidermal growth factor (EGF); the ADSCs-derived neurospheres were terminally differentiated after growth factor withdrawal. Expression of Nestin, NeuN, MAP2, and GFAP in ADSCs and terminally differentiated neurospheres was shown by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blotting, and immunocytochemistry; cell proliferation in neurospheres was evaluated by cell cycle analyses, immunostaining, and flow cytometry. These data strongly support the conclusion that human ADSCs can successfully differentiate into neurospheres efficiently on uncoated culture flasks, which present similar molecular marker pattern and proliferative ability with NSCs derived from embryonic and adult brain tissues. Therefore, human ADSCs may be an ideal alternative source of stem cells for the treatment of neurodegenerative diseases.
Collapse
|
52
|
Razavi S, Nazem G, Mardani M, Esfandiari E, Salehi H, Esfahani SHZ. Neurotrophic factors and their effects in the treatment of multiple sclerosis. Adv Biomed Res 2015; 4:53. [PMID: 25802822 PMCID: PMC4361963 DOI: 10.4103/2277-9175.151570] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 04/24/2014] [Indexed: 12/24/2022] Open
Abstract
Neurotrophins are small molecules of polypeptides, which include nerve growth factor (NGF) family, glial cell line–derived neurotrophic factor (GDNF) family ligands, and neuropoietic cytokines. These factors have an important role in neural regeneration, remyelination, and regulating the development of the peripheral and central nervous systems (PNS and CNS, respectively) by intracellular signaling through specific receptors. It has been suggested that the pathogenesis of human neurodegenerative disorders may be due to an alteration in the neurotrophic factors and their receptors. The use of neurotrophic factors as therapeutic agents is a novel strategy for restoring and maintaining neuronal function during neurodegenerative disorders such as multiple sclerosis. Innate and adaptive immune responses contribute to pathology of neurodegenerative disorders. Furthermore, autoimmune and mesenchymal stem cells, by the release of neurotrophic factors, have the ability to protect neuronal population and can efficiently suppress the formation of new lesions. So, these cells may be an alternative source for delivering neurotrophic factors into the CNS.
Collapse
Affiliation(s)
- Shahnaz Razavi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ghasemi Nazem
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Mardani
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ebrahim Esfandiari
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
53
|
Novozhilova E, Englund-Johansson U, Kale A, Jiao Y, Olivius P. Effects of ROCK inhibitor Y27632 and EGFR inhibitor PD168393 on human neural precursors co-cultured with rat auditory brainstem explant. Neuroscience 2014; 287:43-54. [PMID: 25514049 DOI: 10.1016/j.neuroscience.2014.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 12/05/2014] [Accepted: 12/05/2014] [Indexed: 12/24/2022]
Abstract
Hearing function lost by degeneration of inner ear spiral ganglion neurons (SGNs) in the auditory nervous system could potentially be compensated by cellular replacement using suitable donor cells. Donor cell-derived neuronal development with functional synaptic formation with auditory neurons of the cochlear nucleus (CN) in the brainstem is a prerequisite for a successful transplantation. Here a rat auditory brainstem explant culture system was used as a screening platform for donor cells. The explants were co-cultured with human neural precursor cells (HNPCs) to determine HNPCs developmental potential in the presence of environmental cues characteristic for the auditory brainstem region in vitro. We explored effects of pharmacological inhibition of GTPase Rho with its effector Rho-associated kinase (ROCK) and epidermal growth factor receptor (EGFR) signaling on the co-cultures. Pharmacological agents ROCK inhibitor Y27632 and EGFR blocker PD168393 were tested. Effect of the treatment on explant penetration by green fluorescent protein (GFP)-labeled HNPCs was evaluated based on the following criteria: number of GFP-HNPCs located within the explant; distance migrated by the GFP-HNPCs deep into the explant; length of the GFP+/neuronal class III β-tubulin (TUJ1)+ processes developed and phenotypes displayed. In a short 2-week co-culture both inhibitors had growth-promoting effects on HNPCs, prominent in neurite extension elongation. Significant enhancement of migration and in-growth of HNPCs into the brain slice tissue was only observed in Y27632-treated co-cultures. Difference between Y27632- and PD168393-treated HNPCs acquiring neuronal fate was significant, though not different from the fates acquired in control co-culture. Our data suggest the presence of inhibitory mechanisms in the graft-host environment of the auditory brainstem slice co-culture system with neurite growth arresting properties which can be modulated by administration of signaling pathways antagonists. Therefore the co-culture system can be utilized for screens of donor cells and compounds regulating neuronal fate determination.
Collapse
Affiliation(s)
- E Novozhilova
- Center for Hearing and Communication Research and the Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital, Stockholm 17176, Sweden.
| | - U Englund-Johansson
- Department of Ophthalmology, Institution of Clinical Sciences in Lund, Lund University, 221 84 Lund, Sweden
| | - A Kale
- Center for Hearing and Communication Research and the Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital, Stockholm 17176, Sweden
| | - Y Jiao
- Center for Hearing and Communication Research and the Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital, Stockholm 17176, Sweden; Department of Otolaryngology, Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, 100730 Beijing, China
| | - P Olivius
- Center for Hearing and Communication Research and the Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institute, Karolinska University Hospital, Stockholm 17176, Sweden; Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, SE-58183, Sweden.
| |
Collapse
|
54
|
Czepiel M, Boddeke E, Copray S. Human oligodendrocytes in remyelination research. Glia 2014; 63:513-30. [DOI: 10.1002/glia.22769] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/05/2014] [Indexed: 01/04/2023]
Affiliation(s)
- Marcin Czepiel
- Department of Neuroscience; University Medical Center Groningen; A.Deusinglaan 1, 9713AV Groningen The Netherlands
| | - Erik Boddeke
- Department of Neuroscience; University Medical Center Groningen; A.Deusinglaan 1, 9713AV Groningen The Netherlands
| | - Sjef Copray
- Department of Neuroscience; University Medical Center Groningen; A.Deusinglaan 1, 9713AV Groningen The Netherlands
| |
Collapse
|
55
|
Wu L, Ran C, Liu S, Liao L, Chen Y, Guo H, Wu W, Yan C. Jiaweisinisan facilitates neurogenesis in the hippocampus after stress damage. Neural Regen Res 2014; 8:1091-102. [PMID: 25206403 PMCID: PMC4145897 DOI: 10.3969/j.issn.1673-5374.2013.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 03/06/2013] [Indexed: 11/18/2022] Open
Abstract
The traditional Chinese medicine Jiaweisinisan has antidepressant effects, and can inhibit hypothalamus-pituitary-adrenal gland axis hyperactivity in stress-induced depression. In this study, rat hippocampal neural precursor cells were cultured in serum-free medium in vitro and a stress damage model was established with 120 μM corticosterone. Cells were treated with 10% (v/v) Jiaweisinisan drug-containing serum and the corticosterone antagonist RU38486. Results of the 3-(4,5-dimethylthiazol-2-yl)-3,5-di-phenytetrazoliumromide assay showed that both Jiaweisinisan drug-containing serum and RU38486 promoted the proliferation of neural precursor cells after corticosterone exposure. Immunofluorescence detection showed that after Jiaweisinisan drug-containing serum and RU38486 treatment, the 5-bromo-2-deoxyuridine/terminal deoxynucleotidyl transferase dUTP nick end labeling ratio in hippocampal neural precursor cells significantly increased, and the apoptotic rates of glial cells reduced, and neuron-like cell differentiation from neural precursor cells significantly increased. Our experimental findings indicate that Jiaweisinisan promotes hippocampal neurogenesis after stress damage.
Collapse
Affiliation(s)
- Lili Wu
- Department of Basic Theory of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510060, Guangdong Province, China
| | - Chuanlian Ran
- Department of Traditional Chinese Medicine, Luzhou Medicine College, Luzhou 646000, Sichuang Province, China
| | - Shukao Liu
- Institute of Pharmaceutical Research, Yangtze River Pharmaceutical Group Company Limited, Taizhou 225321, Jiangsu Province, China
| | - Lizhen Liao
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Yanling Chen
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Hualei Guo
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Weikang Wu
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Can Yan
- Department of Basic Theory of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
56
|
BDNF increases survival and neuronal differentiation of human neural precursor cells cotransplanted with a nanofiber gel to the auditory nerve in a rat model of neuronal damage. BIOMED RESEARCH INTERNATIONAL 2014; 2014:356415. [PMID: 25243135 PMCID: PMC4160623 DOI: 10.1155/2014/356415] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 05/02/2014] [Accepted: 07/29/2014] [Indexed: 12/03/2022]
Abstract
Objectives. To study possible nerve regeneration of a damaged auditory nerve by the use of stem cell transplantation. Methods. We transplanted HNPCs to the rat AN trunk by the internal auditory meatus (IAM). Furthermore, we studied if addition of BDNF affects survival and phenotypic differentiation of the grafted HNPCs. A bioactive nanofiber gel (PA gel), in selected groups mixed with BDNF, was applied close to the implanted cells. Before transplantation, all rats had been deafened by a round window niche application of β-bungarotoxin. This neurotoxin causes a selective toxic destruction of the AN while keeping the hair cells intact. Results. Overall, HNPCs survived well for up to six weeks in all groups. However, transplants receiving the BDNF-containing PA gel demonstrated significantly higher numbers of HNPCs and neuronal differentiation. At six weeks, a majority of the HNPCs had migrated into the brain stem and differentiated. Differentiated human cells as well as neurites were observed in the vicinity of the cochlear nucleus. Conclusion. Our results indicate that human neural precursor cells (HNPC) integration with host tissue benefits from additional brain derived neurotrophic factor (BDNF) treatment and that these cells appear to be good candidates for further regenerative studies on the auditory nerve (AN).
Collapse
|
57
|
Mortazavi MM, Harmon OA, Adeeb N, Deep A, Tubbs RS. Treatment of spinal cord injury: a review of engineering using neural and mesenchymal stem cells. Clin Anat 2014; 28:37-44. [PMID: 25156268 DOI: 10.1002/ca.22443] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/02/2014] [Indexed: 12/16/2022]
Abstract
Over time, various treatment modalities for spinal cord injury have been trialed, including pharmacological and nonpharmacological methods. Among these, replacement of the injured neural and paraneural tissues via cellular transplantation of neural and mesenchymal stem cells has been the most attractive. Extensive experimental studies have been done to identify the safety and effectiveness of this transplantation in animal and human models. Herein, we review the literature for studies conducted, with a focus on the human-related studies, recruitment, isolation, and transplantation, of these multipotent stem cells, and associated outcomes.
Collapse
Affiliation(s)
- Martin M Mortazavi
- Department of Neurosurgery, University of Washington, Seattle, Washington
| | | | | | | | | |
Collapse
|
58
|
Takahashi M, Hirakawa H, Yajima H, Izumi-Nakajima N, Okayasu R, Fujimori A. Carbon ion beam is more effective to induce cell death in sphere-type A172 human glioblastoma cells compared with X-rays. Int J Radiat Biol 2014; 90:1125-32. [PMID: 24882391 DOI: 10.3109/09553002.2014.927933] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE To obtain human glioblastoma cells A172 expressing stem cell-related protein and comparison of radiosensitivity in these cells with X-rays and carbon beam. METHODS Human monolayer-type A172 glioblastoma cells were maintained in normal medium with 10% bovine serum. In order to obtain sphere-type A172 cells the medium was replaced with serum-free medium supplemented with growth factors. Both types of A172 cells were irradiated with either X-rays or carbon ion beams and their radiosensitivity was evaluated. RESULTS Serum-free medium induced expression of stem cell-related proteins in A172 cells along with the neurosphere-like appearance. These sphere-type cells were found resistant to both X-rays and carbon ion beams. Phosphorylation of histone H2A family member X persisted for a longer period in the cells exposed to carbon ion beams than in those exposed to X-rays and it disappeared quicker in the sphere type than in the monolayer type. Relative radioresistance of the sphere type cells was smaller for carbon ion beams than for X-rays. CONCLUSIONS We demonstrated that glioblastoma A172 cells with induced stem cell-related proteins turned resistant to irradiation. Accelerated heavy ion particles may have advantage over X-rays in overcoming the tumor resistance due to cell stemness.
Collapse
Affiliation(s)
- Momoko Takahashi
- Translational Research Center, Fukushima Medical University , Fukushima
| | | | | | | | | | | |
Collapse
|
59
|
Translation: screening for novel therapeutics with disease-relevant cell types derived from human stem cell models. Biol Psychiatry 2014; 75:952-60. [PMID: 23876186 PMCID: PMC3815991 DOI: 10.1016/j.biopsych.2013.05.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 05/02/2013] [Accepted: 05/29/2013] [Indexed: 12/23/2022]
Abstract
The advent of somatic cell reprogramming technologies-which enables the generation of patient-specific, induced pluripotent stem cell and other trans-differentiated human neuronal cell models-provides new means of gaining insight into the molecular mechanisms and neural substrates of psychiatric disorders. By allowing a more precise understanding of genotype-phenotype relationship in disease-relevant human cell types, the use of reprogramming technologies in tandem with emerging genome engineering approaches provides a previously "missing link" between basic research and translational efforts. In this review, we summarize advances in applying human pluripotent stem cell and reprogramming technologies to generate specific neural subtypes with a focus on the use of these in vitro systems for the discovery of small molecule-probes and novel therapeutics. Examples are given where human cell models of psychiatric disorders have begun to reveal new mechanistic insight into pathophysiology and simultaneously have provided the foundation for developing disease-relevant, phenotypic assays suitable for both functional genomic and chemical screens. A number of areas for future research are discussed, including the need to develop robust methodology for the reproducible, large-scale production of disease-relevant neural cell types in formats compatible with high-throughput screening modalities, including high-content imaging, multidimensional, signature-based screening, and in vitro network with multielectrode arrays. Limitations, including the challenges in recapitulating neurocircuits and non-cell autonomous phenotypes are discussed. Although these technologies are still in active development, we conclude that, as our understanding of how to efficiently generate and probe the plasticity of patient-specific stem models improves, their utility is likely to advance rapidly.
Collapse
|
60
|
Shelley BC, Gowing G, Svendsen CN. A cGMP-applicable expansion method for aggregates of human neural stem and progenitor cells derived from pluripotent stem cells or fetal brain tissue. J Vis Exp 2014. [PMID: 24962813 DOI: 10.3791/51219] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A cell expansion technique to amass large numbers of cells from a single specimen for research experiments and clinical trials would greatly benefit the stem cell community. Many current expansion methods are laborious and costly, and those involving complete dissociation may cause several stem and progenitor cell types to undergo differentiation or early senescence. To overcome these problems, we have developed an automated mechanical passaging method referred to as "chopping" that is simple and inexpensive. This technique avoids chemical or enzymatic dissociation into single cells and instead allows for the large-scale expansion of suspended, spheroid cultures that maintain constant cell/cell contact. The chopping method has primarily been used for fetal brain-derived neural progenitor cells or neurospheres, and has recently been published for use with neural stem cells derived from embryonic and induced pluripotent stem cells. The procedure involves seeding neurospheres onto a tissue culture Petri dish and subsequently passing a sharp, sterile blade through the cells effectively automating the tedious process of manually mechanically dissociating each sphere. Suspending cells in culture provides a favorable surface area-to-volume ratio; as over 500,000 cells can be grown within a single neurosphere of less than 0.5 mm in diameter. In one T175 flask, over 50 million cells can grow in suspension cultures compared to only 15 million in adherent cultures. Importantly, the chopping procedure has been used under current good manufacturing practice (cGMP), permitting mass quantity production of clinical-grade cell products.
Collapse
|
61
|
Perígolo-Vicente R, Ritt K, Gonçalves-de-Albuquerque CF, Castro-Faria-Neto HC, Paes-de-Carvalho R, Giestal-de-Araujo E. IL-6, A1 and A2aR: a crosstalk that modulates BDNF and induces neuroprotection. Biochem Biophys Res Commun 2014; 449:477-82. [PMID: 24845382 DOI: 10.1016/j.bbrc.2014.05.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 05/08/2014] [Indexed: 11/15/2022]
Abstract
Several diseases are related to retinal ganglion cell death, such as glaucoma, diabetes and other retinopathies. Many studies have attempted to identify factors that could increase neuroprotection after axotomy of these cells. Interleukin-6 has been shown to be able to increase the survival and regeneration of retinal ganglion cells (RGC) in mixed culture as well as in vivo. In this work we show that the trophic effect of IL-6 is mediated by adenosine receptor (A2aR) activation and also by the presence of extracellular BDNF. We also show that there is a complex cross-talk between IL-6, BDNF, the Adenosine A1 and A2a receptors that results in neuroprotection of retinal ganglion cells.
Collapse
Affiliation(s)
- Rafael Perígolo-Vicente
- Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio de Janeiro CEP: 24020-140, Brazil; Blizard Institute - Queen Mary, University of London, 4 Newark St, London, City of London, Greater London E1 2AT, United Kingdom.
| | - Karen Ritt
- Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio de Janeiro CEP: 24020-140, Brazil
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Departamento de Fisiologia e Farmacodinâmica, Av. Brasil, n° 4365, Manguinhos, CEP: 21045-900 Rio de Janeiro, RJ, Brazil
| | - Hugo Caire Castro-Faria-Neto
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Departamento de Fisiologia e Farmacodinâmica, Av. Brasil, n° 4365, Manguinhos, CEP: 21045-900 Rio de Janeiro, RJ, Brazil
| | - Roberto Paes-de-Carvalho
- Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio de Janeiro CEP: 24020-140, Brazil
| | - Elizabeth Giestal-de-Araujo
- Departamento de Neurobiologia, Instituto de Biologia, Universidade Federal Fluminense, Outeiro de São João Batista s/n, Niterói, Rio de Janeiro CEP: 24020-140, Brazil
| |
Collapse
|
62
|
Compagnucci C, Nizzardo M, Corti S, Zanni G, Bertini E. In vitro neurogenesis: development and functional implications of iPSC technology. Cell Mol Life Sci 2014; 71:1623-39. [PMID: 24252976 PMCID: PMC11113522 DOI: 10.1007/s00018-013-1511-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 10/28/2013] [Accepted: 10/30/2013] [Indexed: 12/21/2022]
Abstract
Neurogenesis is the developmental process regulating cell proliferation of neural stem cells, determining their differentiation into glial and neuronal cells, and orchestrating their organization into finely regulated functional networks. Can this complex process be recapitulated in vitro using induced pluripotent stem cell (iPSC) technology? Can neurodevelopmental and neurodegenerative diseases be modeled using iPSCs? What is the potential of iPSC technology in neurobiology? What are the recent advances in the field of neurological diseases? Since the applications of iPSCs in neurobiology are based on the capacity to regulate in vitro differentiation of human iPSCs into different neuronal subtypes and glial cells, and the possibility of obtaining iPSC-derived neurons and glial cells is based on and hindered by our poor understanding of human embryonic development, we reviewed current knowledge on in vitro neural differentiation from a developmental and cellular biology perspective. We highlight the importance to further advance our understanding on the mechanisms controlling in vivo neurogenesis in order to efficiently guide neurogenesis in vitro for cell modeling and therapeutical applications of iPSCs technology.
Collapse
Affiliation(s)
- Claudia Compagnucci
- Unit of Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Department of Neurosciences, Bambino Gesù Children's Research Hospital, IRCCS, 0165, Rome, Italy,
| | | | | | | | | |
Collapse
|
63
|
Wakeman DR, Redmond DE, Dodiya HB, Sladek JR, Leranth C, Teng YD, Samulski RJ, Snyder EY. Human neural stem cells survive long term in the midbrain of dopamine-depleted monkeys after GDNF overexpression and project neurites toward an appropriate target. Stem Cells Transl Med 2014; 3:692-701. [PMID: 24744393 DOI: 10.5966/sctm.2013-0208] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Transplanted multipotent human fetal neural stem cells (hfNSCs) significantly improved the function of parkinsonian monkeys in a prior study primarily by neuroprotection, with only 3%-5% of cells expressing a dopamine (DA) phenotype. In this paper, we sought to determine whether further manipulation of the neural microenvironment by overexpression of a developmentally critical molecule, glial cell-derived neurotrophic factor (GDNF), in the host striatum could enhance DA differentiation of hfNSCs injected into the substantia nigra and elicit growth of their axons to the GDNF-expressing target. hfNSCs were transplanted into the midbrain of 10 green monkeys exposed to 1-methyl-4-phenyl-1,2,3,6-tetrahydro-pyridine. GDNF was delivered concomitantly to the striatum via an adeno-associated virus serotype 5 vector, and the fate of grafted cells was assessed after 11 months. Donor cells remained predominantly within the midbrain at the injection site and sprouted numerous neurofilament-immunoreactive fibers that appeared to course rostrally toward the striatum in parallel with tyrosine hydroxylase-immunoreactive fibers from the host substantia nigra but did not mature into DA neurons. This work suggests that hfNSCs can generate neurons that project long fibers in the adult primate brain. However, in the absence of region-specific signals and despite GDNF overexpression, hfNSCs did not differentiate into mature DA neurons in large numbers. It is encouraging, however, that the adult primate brain appeared to retain axonal guidance cues. We believe that transplantation of stem cells, specifically instructed ex vivo to yield DA neurons, could lead to reconstruction of some portion of the nigrostriatal pathway and prove beneficial for the parkinsonian condition.
Collapse
Affiliation(s)
- Dustin R Wakeman
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - D Eugene Redmond
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hemraj B Dodiya
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - John R Sladek
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Csaba Leranth
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yang D Teng
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - R Jude Samulski
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Evan Y Snyder
- Graduate Program in Biomedical Sciences, University of California at San Diego, La Jolla, California, USA; Program in Stem Cell and Regenerative Biology, Sanford-Burnham Medical Research Institute, La Jolla, California, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA; Departments of Psychiatry, Neurosurgery, and Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA; Department of Neurology, University of Colorado School of Medicine, Denver, Colorado, USA; Department of Neurosurgery and Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA; Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
64
|
Baumann J, Barenys M, Gassmann K, Fritsche E. Comparative human and rat "neurosphere assay" for developmental neurotoxicity testing. ACTA ACUST UNITED AC 2014; 59:12.21.1-24. [PMID: 24898107 DOI: 10.1002/0471140856.tx1221s59] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The developing nervous system is highly vulnerable to the adverse effects of chemical agents. Currently, there is an increasing need for testing and regulating chemical compounds in general use and, due to the lack of available data, to identify those which are developmental neurotoxicants. In this context, alternative testing strategies are needed in order to allow fast and cost-efficient screening and to reduce the number of animal experiments usually required. In this unit we present an in vitro three-dimensional model for developmental neurotoxicity screening based on human and rat neural progenitor cells. This model enables the detection of disturbances in basic processes of brain development, such as proliferation, migration, differentiation and apoptosis, and allows the distinction of these specific disturbances from general cytotoxicity. Furthermore, the comparison of human and rat data provides useful insights into species differences for toxicodynamics of compounds contributing to human risk assessment of developmental neurotoxicants.
Collapse
Affiliation(s)
- Jenny Baumann
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany; Both authors contributed equally to this unit
| | | | | | | |
Collapse
|
65
|
Talaverón R, Matarredona ER, de la Cruz RR, Macías D, Gálvez V, Pastor AM. Implanted neural progenitor cells regulate glial reaction to brain injury and establish gap junctions with host glial cells. Glia 2014; 62:623-38. [PMID: 24481572 DOI: 10.1002/glia.22630] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/20/2013] [Accepted: 12/23/2013] [Indexed: 01/19/2023]
Abstract
Transplantation of neural stem/progenitor cells (NPCs) in the lesioned brain is able to restore morphological and physiological alterations induced by different injuries. The local microenvironment created at the site of grafting and the communication between grafted and host cells are crucial in the beneficial effects attributed to the NPC implants. We have previously described that NPC transplantation in an animal model of central axotomy restores firing properties and synaptic coverage of lesioned neurons and modulates their trophic factor content. In this study, we aim to explore anatomical relationships between implanted NPCs and host glia that might account for the implant-induced neuroprotective effects. Postnatal rat subventricular zone NPCs were isolated and grafted in adult rats after transection of the medial longitudinal fascicle. Brains were removed and analyzed eight weeks later. Immunohistochemistry for different glial markers revealed that NPC-grafted animals displayed significantly greater microglial activation than animals that received only vehicle injections. Implanted NPCs were located in close apposition to activated microglia and reactive astrocytes. The gap junction protein connexin43 was present in NPCs and glial cells at the lesion site and was often found interposed within adjacent implanted and glial cells. Gap junctions were identified between implanted NPCs and host astrocytes and less frequently between NPCs and microglia. Our results show that implanted NPCs modulate the glial reaction to lesion and establish the possibility of communication through gap junctions between grafted and host glial cells which might be involved in the restorative effects of NPC implants.
Collapse
Affiliation(s)
- Rocío Talaverón
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | | | | | | | | | | |
Collapse
|
66
|
Rinaldi F, Hartfield EM, Crompton LA, Badger JL, Glover CP, Kelly CM, Rosser AE, Uney JB, Caldwell MA. Cross-regulation of Connexin43 and β-catenin influences differentiation of human neural progenitor cells. Cell Death Dis 2014; 5:e1017. [PMID: 24457961 PMCID: PMC4040652 DOI: 10.1038/cddis.2013.546] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 10/30/2013] [Accepted: 11/04/2013] [Indexed: 12/03/2022]
Abstract
Connexin43 (Cx43) is the most widely and abundantly expressed gap junction (GJ) protein and it is strongly associated with the regulation of cell cycle progression. Emerging roles for Cx43 in cell adhesion and migration during neural differentiation have also been recently recognized, and this has emphasized the involvement of Cx43 in different physiological process beyond its role as a GJ protein. In this study, we explore the function of Cx43 in the differentiation of human neural progenitor cells (hNPCs) using viral vectors that mediate the overexpression or knockdown of the protein. Results showed that in the absence of this protein fetal cortex-derived hNPCs differentiated toward a neuronal phenotype at expenses of a glial phenotype. Furthermore, the silencing of Cx43 did not affect hNPC proliferation rate or numbers of apoptotic cells. The increase in the number of neurons was not recapitulated when GJ intercellular communications were pharmacologically blocked, and this suggested that Cx43 was influencing hNPCs differentiation with a GJ-independent effect. In addition, Cx43 knockdown significantly increased β-catenin signaling, which has been shown to regulate the transcription of pro-neuronal genes during embryonic neural development. Our results add further support to the hypothesis that Cx43 protein itself regulates key signaling pathways during development and neurogenesis beyond its role as GJ protein.
Collapse
Affiliation(s)
- F Rinaldi
- Stem Cells and Neuroregeneration Research Group, School of Clinical Sciences, Medical Sciences Building, University Walk, University of Bristol, Bristol, UK
| | - E M Hartfield
- Stem Cells and Neuroregeneration Research Group, School of Clinical Sciences, Medical Sciences Building, University Walk, University of Bristol, Bristol, UK
| | - L A Crompton
- Stem Cells and Neuroregeneration Research Group, School of Clinical Sciences, Medical Sciences Building, University Walk, University of Bristol, Bristol, UK
| | - J L Badger
- Stem Cells and Neuroregeneration Research Group, School of Clinical Sciences, Medical Sciences Building, University Walk, University of Bristol, Bristol, UK
| | - C P Glover
- Stem Cells and Neuroregeneration Research Group, School of Clinical Sciences, Medical Sciences Building, University Walk, University of Bristol, Bristol, UK
| | - C M Kelly
- Brain Repair Group, School of Biosciences, Life Science Building, University of Cardiff, Cardiff, UK
| | - A E Rosser
- Brain Repair Group, School of Biosciences, Life Science Building, University of Cardiff, Cardiff, UK
- Department of Neurology, School of Medicine, University of Cardiff, Cardiff, UK
| | - J B Uney
- Stem Cells and Neuroregeneration Research Group, School of Clinical Sciences, Medical Sciences Building, University Walk, University of Bristol, Bristol, UK
| | - M A Caldwell
- Stem Cells and Neuroregeneration Research Group, School of Clinical Sciences, Medical Sciences Building, University Walk, University of Bristol, Bristol, UK
| |
Collapse
|
67
|
Abstract
The aim of stem cell therapy for Parkinson's disease is to reconstruct nigro-striatal neuronal pathways using endogenous neural stem/precursor cells or grafted dopaminergic neurons. As an alternative, transplantation of stem cell-derived dopaminergic neurons into the striatum has been attempted, with the aim of stimulating local synapse formation and/or release of dopamine and cytokines from grafted cells. Candidate stem cells include neural stem/precursor cells, embryonic stem cells and other stem/precursor cells. Among these, embryonic stem cells are pluripotent cells that proliferate extensively, making them a good potential donor source for transplantation. However, tumor formation and ethical issues present major problems for embryonic stem cell therapy. This review describes the current status of stem cell therapy for Parkinson's disease, as well as future research approaches from a clinical perspective.
Collapse
Affiliation(s)
- Jun Takahashi
- Kyoto University, Department of Biological Repair, Institute for Frontier Medical Sciences, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
68
|
Li YC, Tsai LK, Wang JH, Young TH. A neural stem/precursor cell monolayer for neural tissue engineering. Biomaterials 2014; 35:1192-204. [DOI: 10.1016/j.biomaterials.2013.10.066] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 10/20/2013] [Indexed: 11/30/2022]
|
69
|
Human Stem/Progenitor Cell-Based Assays for Neurodevelopmental Toxicity Testing. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2014. [DOI: 10.1007/978-1-4939-0521-8_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
70
|
Crompton LA, Byrne ML, Taylor H, Kerrigan TL, Bru-Mercier G, Badger JL, Barbuti PA, Jo J, Tyler SJ, Allen SJ, Kunath T, Cho K, Caldwell MA. Stepwise, non-adherent differentiation of human pluripotent stem cells to generate basal forebrain cholinergic neurons via hedgehog signaling. Stem Cell Res 2013; 11:1206-21. [PMID: 24013066 DOI: 10.1016/j.scr.2013.08.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 07/12/2013] [Accepted: 08/02/2013] [Indexed: 10/26/2022] Open
Abstract
Basal forebrain cholinergic neurons (bfCNs) which provide innervation to the hippocampus and cortex, are required for memory and learning, and are primarily affected in Alzheimer's Disease (AD), resulting in related cognitive decline. Therefore generation of a source of bfCNs from human pluripotent stem cells (hPSCs) is crucial for in vitro disease modeling and development of novel AD therapies. In addition, for the advancement of regenerative approaches there is a requirement for an accurate developmental model to study the neurogenesis and survival of this population. Here we demonstrate the efficient production of bfCNs, using a novel embryoid body (EB) based non-adherent differentiation (NAdD) protocol. We establish a specific basal forebrain neural stem cell (NSC) phenotype via expression of the basal forebrain transcription factors NKX2.1 and LHX8, as well as the general forebrain marker FOXG1. We present evidence that this lineage is achieved via recapitulation of embryonic events, with induction of intrinsic hedgehog signaling, through the use of a 3D non-adherent differentiation system. This is the first example of hPSC-derived basal forebrain-like NSCs, which are scalable via self-renewal in prolonged culture. Furthermore upon terminal differentiation these basal forebrain-like NSCs generate high numbers of cholinergic neurons expressing the specific markers ChAT, VACht and ISL1. These hPSC-derived bfCNs possess characteristics that are crucial in a model to study AD related cholinergic neuronal loss in the basal forebrain. Examples are expression of the therapeutic target p75(NTR), the release of acetylcholine, and demonstration of a mature, and functional electrophysiological profile. In conclusion, this work provides a renewable source of human functional bfCNs applicable for studying AD specifically in the cholinergic system, and also provides a model of the key embryonic events in human bfCN development.
Collapse
Affiliation(s)
- Lucy A Crompton
- Henry Wellcome Laboratory for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Stem cells expanded from the human embryonic hindbrain stably retain regional specification and high neurogenic potency. J Neurosci 2013; 33:12407-22. [PMID: 23884946 DOI: 10.1523/jneurosci.0130-13.2013] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Stem cell lines that faithfully maintain the regional identity and developmental potency of progenitors in the human brain would create new opportunities in developmental neurobiology and provide a resource for generating specialized human neurons. However, to date, neural progenitor cultures derived from the human brain have either been short-lived or exhibit restricted, predominantly glial, differentiation capacity. Pluripotent stem cells are an alternative source, but to ascertain definitively the identity and fidelity of cell types generated solely in vitro is problematic. Here, we show that hindbrain neuroepithelial stem (hbNES) cells can be derived and massively expanded from early human embryos (week 5-7, Carnegie stage 15-17). These cell lines are propagated in adherent culture in the presence of EGF and FGF2 and retain progenitor characteristics, including SOX1 expression, formation of rosette-like structures, and high neurogenic capacity. They generate GABAergic, glutamatergic and, at lower frequency, serotonergic neurons. Importantly, hbNES cells stably maintain hindbrain specification and generate upper rhombic lip derivatives on exposure to bone morphogenetic protein (BMP). When grafted into neonatal rat brain, they show potential for integration into cerebellar development and produce cerebellar granule-like cells, albeit at low frequency. hbNES cells offer a new system to study human cerebellar specification and development and to model diseases of the hindbrain. They also provide a benchmark for the production of similar long-term neuroepithelial-like stem cells (lt-NES) from pluripotent cell lines. To our knowledge, hbNES cells are the first demonstration of highly expandable neuroepithelial stem cells derived from the human embryo without genetic immortalization.
Collapse
|
72
|
Tsukamoto A, Uchida N, Capela A, Gorba T, Huhn S. Clinical translation of human neural stem cells. Stem Cell Res Ther 2013; 4:102. [PMID: 23987648 PMCID: PMC3854682 DOI: 10.1186/scrt313] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Human neural stem cell transplants have potential as therapeutic candidates to treat a vast number of disorders of the central nervous system (CNS). StemCells, Inc. has purified human neural stem cells and developed culture conditions for expansion and banking that preserve their unique biological properties. The biological activity of these human central nervous system stem cells (HuCNS-SC®) has been analyzed extensively in vitro and in vivo. When formulated for transplantation, the expanded and cryopreserved banked cells maintain their stem cell phenotype, self-renew and generate mature oligodendrocytes, neurons and astrocytes, cells normally found in the CNS. In this overview, the rationale and supporting data for pursuing neuroprotective strategies and clinical translation in the three components of the CNS (brain, spinal cord and eye) are described. A phase I trial for a rare myelin disorder and phase I/II trial for spinal cord injury are providing intriguing data relevant to the biological properties of neural stem cells, and the early clinical outcomes compel further development.
Collapse
|
73
|
Dooley D, Vidal P, Hendrix S. Immunopharmacological intervention for successful neural stem cell therapy: New perspectives in CNS neurogenesis and repair. Pharmacol Ther 2013; 141:21-31. [PMID: 23954656 DOI: 10.1016/j.pharmthera.2013.08.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 12/11/2022]
Abstract
The pharmacological support and stimulation of endogenous and transplanted neural stem cells (NSCs) is a major challenge in brain repair. Trauma to the central nervous system (CNS) results in a distinct inflammatory response caused by local and infiltrating immune cells. This makes NSC-supported regeneration difficult due to the presence of inhibitory immune factors which are upregulated around the lesion site. The continual and dual role of the neuroinflammatory response leaves it difficult to decipher upon a single modulatory strategy. Therefore, understanding the influence of cytokines upon regulation of NSC self-renewal, proliferation and differentiation is crucial when designing therapies for CNS repair. There is a plethora of partially conflicting data in vitro and in vivo on the role of cytokines in modulating the stem cell niche and the milieu around NSC transplants. This is mainly due to the pleiotropic role of many factors. In order for cell-based therapy to thrive, treatment must be phase-specific to the injury and also be personalized for each patient, i.e. taking age, sex, neuroimmune and endocrine status as well as other key parameters into consideration. In this review, we will summarize the most relevant information concerning interleukin (IL)-1, IL-4, IL-10, IL-15, IFN-γ, the neuropoietic cytokine family and TNF-α in order to extract promising therapeutic approaches for further research. We will focus on the consequences of neuroinflammation on endogenous brain stem cells and the transplantation environment, the effects of the above cytokines on NSCs, as well as immunopharmacological manipulation of the microenvironment for potential therapeutic use.
Collapse
Affiliation(s)
- Dearbhaile Dooley
- Dep. of Morphology & Biomedical Research Institute, Hasselt University, Belgium
| | - Pia Vidal
- Dep. of Morphology & Biomedical Research Institute, Hasselt University, Belgium
| | - Sven Hendrix
- Dep. of Morphology & Biomedical Research Institute, Hasselt University, Belgium.
| |
Collapse
|
74
|
Review of transplantation of neural stem/progenitor cells for spinal cord injury. Int J Dev Neurosci 2013; 31:701-13. [DOI: 10.1016/j.ijdevneu.2013.07.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 07/02/2013] [Accepted: 07/26/2013] [Indexed: 11/17/2022] Open
|
75
|
Konagaya S, Kato K, Nakaji-Hirabayashi T, Iwata H. Selective and rapid expansion of human neural progenitor cells on substrates with terminally anchored growth factors. Biomaterials 2013; 34:6008-14. [DOI: 10.1016/j.biomaterials.2013.04.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 04/23/2013] [Indexed: 12/12/2022]
|
76
|
Gil-Perotín S, Duran-Moreno M, Cebrián-Silla A, Ramírez M, García-Belda P, García-Verdugo JM. Adult neural stem cells from the subventricular zone: a review of the neurosphere assay. Anat Rec (Hoboken) 2013; 296:1435-52. [PMID: 23904071 DOI: 10.1002/ar.22746] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 06/18/2013] [Indexed: 01/17/2023]
Abstract
The possibility of obtaining large numbers of cells with potential to become functional neurons implies a great advance in regenerative medicine. A source of cells for therapy is the subventricular zone (SVZ) where adult neural stem cells (NSCs) retain the ability to proliferate, self-renew, and differentiate into several mature cell types. The neurosphere assay, a method to isolate, maintain, and expand these cells has been extensively utilized by research groups to analyze the biological properties of aNSCs and to graft into injured brains from animal models. In this review we briefly describe the neurosphere assay and its limitations, the methods to optimize culture conditions, the identity and the morphology of aNSC-derived neurospheres (including new ultrastructural data). The controversy regarding the identity and "stemness" of cells within the neurosphere is revised. The fine morphology of neurospheres, described thoroughly, allows for phenotypical characterization of cells in the neurospheres and may reveal slight changes that indirectly inform about cell integrity, cell damage, or oncogenic transformation. Along this review we largely highlight the critical points that researchers have to keep in mind before extrapolating results or translating experimental transplantation of neurosphere-derived cells to the clinical setting.
Collapse
Affiliation(s)
- Sara Gil-Perotín
- Laboratory of Comparative Neurobiology, Instituto Cavanilles de Biodiversidad y Biología Evolutiva, University of Valencia, C/Catedratico Jose Beltran no 2, Paterna, Valencia, CIBERNED, Spain
| | | | | | | | | | | |
Collapse
|
77
|
Mousa A, Bakhiet M. Role of cytokine signaling during nervous system development. Int J Mol Sci 2013; 14:13931-57. [PMID: 23880850 PMCID: PMC3742226 DOI: 10.3390/ijms140713931] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 06/19/2013] [Accepted: 06/25/2013] [Indexed: 01/24/2023] Open
Abstract
Cytokines are signaling proteins that were first characterized as components of the immune response, but have been found to have pleiotropic effects in diverse aspects of body function in health and disease. They are secreted by numerous cells and are used extensively in intercellular communications to produce different activities, including intricate processes engaged in the ontogenetic development of the brain. This review discusses factors involved in brain growth regulation and recent findings exploring cytokine signaling pathways during development of the central nervous system. In view of existing data suggesting roles for neurotropic cytokines in promoting brain growth and repair, these molecules and their signaling pathways might become targets for therapeutic intervention in neurodegenerative processes due to diseases, toxicity, or trauma.
Collapse
Affiliation(s)
- Alyaa Mousa
- Department of Anatomy, Faculty of Medicine, Health Sciences Centre, Kuwait University, Safat 13060, Kuwait; E-Mail:
| | - Moiz Bakhiet
- Department of Molecular Medicine, Princess Al-Jawhara Center for Genetics and Inherited Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, P.O. Box 26671 Manama, Bahrain
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +973-1723-7300
| |
Collapse
|
78
|
Tsymbalyuk VI, Vasil’eva IG, Oleksenko NP, Chopik NG, Tsyubko OI, Galanta OS. Stimulation of cholinogenesis in human fetal nerve cell cultures. CYTOL GENET+ 2013. [DOI: 10.3103/s0095452713030109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
79
|
Jha RM, Chrenek R, Magnotti LM, Cardozo DL. The isolation, differentiation, and survival in vivo of multipotent cells from the postnatal rat filum terminale. PLoS One 2013; 8:e65974. [PMID: 23762453 PMCID: PMC3675200 DOI: 10.1371/journal.pone.0065974] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 05/03/2013] [Indexed: 01/25/2023] Open
Abstract
Neural stem cells (NSCs) are undifferentiated cells in the central nervous system (CNS) that are capable of self-renewal and can be induced to differentiate into neurons and glia. Current sources of mammalian NSCs are confined to regions of the CNS that are critical to normal function and surgically difficult to access, which limits their therapeutic potential in human disease. We have found that the filum terminale (FT), a previously unexplored, expendable, and easily accessible tissue at the caudal end of the spinal cord, is a source of multipotent cells in postnatal rats and humans. In this study, we used a rat model to isolate and characterize the potential of these cells. Neurospheres derived from the rat FT are amenable to in vitro expansion in the presence of a combination of growth factors. These proliferating, FT-derived cells formed neurospheres that could be induced to differentiate into neural progenitor cells, neurons, astrocytes, and oligodendrocytes by exposure to serum and/or adhesive substrates. Through directed differentiation using sonic hedgehog and retinoic acid in combination with various neurotrophic factors, FT-derived neurospheres generated motor neurons that were capable of forming neuromuscular junctions in vitro. In addition, FT-derived progenitors that were injected into chick embryos survived and could differentiate into both neurons and glia in vivo.
Collapse
Affiliation(s)
- Ruchira M. Jha
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ryan Chrenek
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Laura M. Magnotti
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| | - David L. Cardozo
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
80
|
Selden NR, Al-Uzri A, Huhn SL, Koch TK, Sikora DM, Nguyen-Driver MD, Guillaume DJ, Koh JL, Gultekin SH, Anderson JC, Vogel H, Sutcliffe TL, Jacobs Y, Steiner RD. Central nervous system stem cell transplantation for children with neuronal ceroid lipofuscinosis. J Neurosurg Pediatr 2013; 11:643-52. [PMID: 23581634 DOI: 10.3171/2013.3.peds12397] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECT Infantile and late-infantile neuronal ceroid lipofuscinoses (NCLs) are invariably fatal lysosomal storage diseases associated with defects in lysosomal enzyme palmitoyl-protein thioesterase 1 (PPT-1) or tripeptidyl peptidase 1 (TPP1) activity. Previous preclinical studies have demonstrated that human CNS stem cells (HuCNS-SCs) produce both PPT-1 and TPP1 and result in donor cell engraftment and reduced accumulation of storage material in the brain when tested in an NCL mouse model. METHODS HuCNS-SC transplantation was tested in an open-label dose-escalation Phase I clinical trial as a potential treatment for infantile and late-infantile NCL. Study design included direct neurosurgical transplantation of allogeneic HuCNS-SCs into the cerebral hemispheres and lateral ventricles accompanied by 12 months of immunosuppression. RESULTS Six children with either the infantile or late-infantile forms of NCL underwent low- (3 patients) and high- (3 patients) dose transplantation of HuCNS-SCs followed by immunosuppression. The surgery, immunosuppression, and cell transplantation were well tolerated. Adverse events following transplantation were consistent with the underlying disease, and none were directly attributed to the donor cells. Observations regarding efficacy of the intervention were limited by the enrollment criteria requiring that patients be in advanced stages of disease. CONCLUSIONS This study represents the first-in-human clinical trial involving transplantation of a purified population of human neural stem cells for a neurodegenerative disorder. The feasibility of this approach and absence of transplantation-related serious adverse events support further exploration of HuCNS-SC transplantation as a potential treatment for select subtypes of NCL, and possibly for other neurodegenerative disorders.
Collapse
Affiliation(s)
- Nathan R Selden
- Department of Neurological Surgery, Oregon Health & Science University, 3303 SW Bond Avenue, Portland, OR 97239, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
van Gorp S, Leerink M, Kakinohana O, Platoshyn O, Santucci C, Galik J, Joosten EA, Hruska-Plochan M, Goldberg D, Marsala S, Johe K, Ciacci JD, Marsala M. Amelioration of motor/sensory dysfunction and spasticity in a rat model of acute lumbar spinal cord injury by human neural stem cell transplantation. Stem Cell Res Ther 2013; 4:57. [PMID: 23710605 PMCID: PMC3706882 DOI: 10.1186/scrt209] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 04/25/2013] [Indexed: 12/15/2022] Open
Abstract
Introduction Intraspinal grafting of human neural stem cells represents a promising approach to promote recovery of function after spinal trauma. Such a treatment may serve to: I) provide trophic support to improve survival of host neurons; II) improve the structural integrity of the spinal parenchyma by reducing syringomyelia and scarring in trauma-injured regions; and III) provide neuronal populations to potentially form relays with host axons, segmental interneurons, and/or α-motoneurons. Here we characterized the effect of intraspinal grafting of clinical grade human fetal spinal cord-derived neural stem cells (HSSC) on the recovery of neurological function in a rat model of acute lumbar (L3) compression injury. Methods Three-month-old female Sprague–Dawley rats received L3 spinal compression injury. Three days post-injury, animals were randomized and received intraspinal injections of either HSSC, media-only, or no injections. All animals were immunosuppressed with tacrolimus, mycophenolate mofetil, and methylprednisolone acetate from the day of cell grafting and survived for eight weeks. Motor and sensory dysfunction were periodically assessed using open field locomotion scoring, thermal/tactile pain/escape thresholds and myogenic motor evoked potentials. The presence of spasticity was measured by gastrocnemius muscle resistance and electromyography response during computer-controlled ankle rotation. At the end-point, gait (CatWalk), ladder climbing, and single frame analyses were also assessed. Syrinx size, spinal cord dimensions, and extent of scarring were measured by magnetic resonance imaging. Differentiation and integration of grafted cells in the host tissue were validated with immunofluorescence staining using human-specific antibodies. Results Intraspinal grafting of HSSC led to a progressive and significant improvement in lower extremity paw placement, amelioration of spasticity, and normalization in thermal and tactile pain/escape thresholds at eight weeks post-grafting. No significant differences were detected in other CatWalk parameters, motor evoked potentials, open field locomotor (Basso, Beattie, and Bresnahan locomotion score (BBB)) score or ladder climbing test. Magnetic resonance imaging volume reconstruction and immunofluorescence analysis of grafted cell survival showed near complete injury-cavity-filling by grafted cells and development of putative GABA-ergic synapses between grafted and host neurons. Conclusions Peri-acute intraspinal grafting of HSSC can represent an effective therapy which ameliorates motor and sensory deficits after traumatic spinal cord injury.
Collapse
|
82
|
Hou Y, Ouyang X, Wan R, Cheng H, Mattson MP, Cheng A. Mitochondrial superoxide production negatively regulates neural progenitor proliferation and cerebral cortical development. Stem Cells 2013; 30:2535-47. [PMID: 22949407 DOI: 10.1002/stem.1213] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although high amounts of reactive oxygen species (ROS) can damage cells, ROS can also play roles as second messengers, regulating diverse cellular processes. Here, we report that embryonic mouse cerebral cortical neural progenitor cells (NPCs) exhibit intermittent spontaneous bursts of mitochondrial superoxide (SO) generation (mitochondrial SO flashes) that require transient opening of membrane permeability transition pores (mPTP). This quantal SO production negatively regulates NPC self-renewal. Mitochondrial SO scavengers and mPTP inhibitors reduce SO flash frequency and enhance NPC proliferation, whereas prolonged mPTP opening and SO generation increase SO flash incidence and decrease NPC proliferation. The inhibition of NPC proliferation by mitochondrial SO involves suppression of extracellular signal-regulated kinases. Moreover, mice lacking SOD2 (SOD2-/- mice) exhibit significantly fewer proliferative NPCs and differentiated neurons in the embryonic cerebral cortex at midgestation compared with wild-type littermates. Cultured SOD2-/- NPCs exhibit a significant increase in SO flash frequency and reduced NPC proliferation. Taken together, our findings suggest that mitochondrial SO flashes negatively regulate NPC self-renewal in the developing cerebral cortex.
Collapse
Affiliation(s)
- Yan Hou
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224, USA
| | | | | | | | | | | |
Collapse
|
83
|
Majumder A, Banerjee S, Harrill JA, Machacek DW, Mohamad O, Bacanamwo M, Mundy WR, Wei L, Dhara SK, Stice SL. Neurotrophic effects of leukemia inhibitory factor on neural cells derived from human embryonic stem cells. Stem Cells 2013; 30:2387-99. [PMID: 22899336 DOI: 10.1002/stem.1201] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Various growth factor cocktails have been used to proliferate and then differentiate human neural progenitor (NP) cells derived from embryonic stem cells (ESC) for in vitro and in vivo studies. However, the cytokine leukemia inhibitory factor (LIF) has been largely overlooked. Here, we demonstrate that LIF significantly enhanced in vitro survival and promoted differentiation of human ESC-derived NP cells. In NP cells, as well as NP-derived neurons, LIF reduced caspase-mediated apoptosis and reduced both spontaneous and H2O2-induced reactive oxygen species in culture. In vitro, NP cell proliferation and the yield of differentiated neurons were significantly higher in the presence of LIF. In NP cells, LIF enhanced cMyc phosphorylation, commonly associated with self-renewal/proliferation. Also, in differentiating NP cells LIF activated the phosphoinositide 3-kinase and signal transducer and activator of transcription 3 pathways, associated with cell survival and reduced apoptosis. When differentiated in LIF+ media, neurite outgrowth and ERK1/2 phosphorylation were potentiated together with increased expression of gp130, a component of the LIF receptor complex. NP cells, pretreated in vitro with LIF, were effective in reducing infarct volume in a model of focal ischemic stroke but LIF did not lead to significantly improved initial NP cell survival over nontreated NP cells. Our results show that LIF signaling significantly promotes human NP cell proliferation, survival, and differentiation in vitro. Activated LIF signaling should be considered in cell culture expansion systems for future human NP cell-based therapeutic transplant studies.
Collapse
Affiliation(s)
- Anirban Majumder
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia 30602, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Gupta N, Henry RG, Strober J, Kang SM, Lim DA, Bucci M, Caverzasi E, Gaetano L, Mandelli ML, Ryan T, Perry R, Farrell J, Jeremy RJ, Ulman M, Huhn SL, Barkovich AJ, Rowitch DH. Neural stem cell engraftment and myelination in the human brain. Sci Transl Med 2013; 4:155ra137. [PMID: 23052294 DOI: 10.1126/scitranslmed.3004373] [Citation(s) in RCA: 215] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pelizaeus-Merzbacher disease (PMD) is a rare leukodystrophy caused by mutation of the proteolipid protein 1 gene. Defective oligodendrocytes in PMD fail to myelinate axons, causing global neurological dysfunction. Human central nervous system stem cells (HuCNS-SCs) can develop into oligodendrocytes and confer structurally normal myelin when transplanted into a hypomyelinating mouse model. A 1-year, open-label phase-1 study was undertaken to evaluate safety and to detect evidence of myelin formation after HuCNS-SC transplantation. Allogeneic HuCNS-SCs were surgically implanted into the frontal lobe white matter in four male subjects with an early-onset severe form of PMD. Immunosuppression was administered for 9 months. Serial neurological evaluations, developmental assessments, and cranial magnetic resonance imaging (MRI) and MR spectroscopy, including high-angular resolution diffusion tensor imaging (DTI), were performed at baseline and after transplantation. The neurosurgical procedure, immunosuppression regimen, and HuCNS-SC transplantation were well tolerated. Modest gains in neurological function were observed in three of the four subjects. No clinical or radiological adverse effects were directly attributed to the donor cells. Reduced T1 and T2 relaxation times were observed in the regions of transplantation 9 months after the procedure in the three subjects. Normalized DTI showed increasing fractional anisotropy and reduced radial diffusivity, consistent with myelination, in the region of transplantation compared to control white matter regions remote to the transplant sites. These phase 1 findings indicate a favorable safety profile for HuCNS-SCs in subjects with PMD. The MRI results suggest durable cell engraftment and donor-derived myelin in the transplanted host white matter.
Collapse
Affiliation(s)
- Nalin Gupta
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Gold- and silver nanoparticles affect the growth characteristics of human embryonic neural precursor cells. PLoS One 2013; 8:e58211. [PMID: 23505470 PMCID: PMC3594300 DOI: 10.1371/journal.pone.0058211] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 02/01/2013] [Indexed: 01/09/2023] Open
Abstract
Rapid development of nanotechnologies and their applications in clinical research have raised concerns about the adverse effects of nanoparticles (NPs) on human health and environment. NPs can be directly taken up by organs exposed, but also translocated to secondary organs, such as the central nervous system (CNS) after systemic- or subcutaneous administration, or via the olfactory system. The CNS is particularly vulnerable during development and recent reports describe transport of NPs across the placenta and even into brain tissue using in vitro and in vivo experimental systems. Here, we investigated whether well-characterized commercial 20 and 80 nm Au- and AgNPs have an effect on human embryonic neural precursor cell (HNPC) growth. After two weeks of NP exposure, uptake of NPs, morphological features and the amount of viable and dead cells, proliferative cells (Ki67 immunostaining) and apoptotic cells (TUNEL assay), respectively, were studied. We demonstrate uptake of both 20 and 80 nm Au- and AgNPs respectively, by HNPCs during proliferation. A significant effect on the sphere size- and morphology was found for all cultures exposed to Au- and AgNPs. AgNPs of both sizes caused a significant increase in numbers of proliferating and apoptotic HNPCs. In contrast, only the highest dose of 20 nm AuNPs significantly affected proliferation, whereas no effect was seen on apoptotic cell death. Our data demonstrates that both Au- and AgNPs interfere with the growth profile of HNPCs, indicating the need of further detailed studies on the adverse effects of NPs on the developing CNS.
Collapse
|
86
|
Novozhilova E, Olivius P, Siratirakun P, Lundberg C, Englund-Johansson U. Neuronal differentiation and extensive migration of human neural precursor cells following co-culture with rat auditory brainstem slices. PLoS One 2013; 8:e57301. [PMID: 23505423 PMCID: PMC3591396 DOI: 10.1371/journal.pone.0057301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 01/21/2013] [Indexed: 11/18/2022] Open
Abstract
Congenital or acquired hearing loss is often associated with a progressive degeneration of the auditory nerve (AN) in the inner ear. The AN is composed of processes and axons of the bipolar spiral ganglion neurons (SGN), forming the connection between the hair cells in the inner ear cochlea and the cochlear nuclei (CN) in the brainstem (BS). Therefore, replacement of SGNs for restoring the AN to improve hearing function in patients who receive a cochlear implantation or have severe AN malfunctions is an attractive idea. A human neural precursor cell (HNPC) is an appropriate donor cell to investigate, as it can be isolated and expanded in vitro with maintained potential to form neurons and glia. We recently developed a post-natal rodent in vitro auditory BS slice culture model including the CN and the central part of the AN for initial studies of candidate cells. Here we characterized the survival, distribution, phenotypic differentiation, and integration capacity of HNPCs into the auditory circuitry in vitro. HNPC aggregates (spheres) were deposited adjacent to or on top of the BS slices or as a monoculture (control). The results demonstrate that co-cultured HNPCs compared to monocultures (1) survive better, (2) distribute over a larger area, (3) to a larger extent and in a shorter time-frame form mature neuronal and glial phenotypes. HNPC showed the ability to extend neurites into host tissue. Our findings suggest that the HNPC-BS slice co-culture is appropriate for further investigations on the integration capacity of HNPCs into the auditory circuitry.
Collapse
Affiliation(s)
- Ekaterina Novozhilova
- Division of Oto-Rhino-Laryngology and Head and Neck Surgery, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- Center for Hearing and Communication Research, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Section of Otorhinolaryngology, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Petri Olivius
- Department of ENT—Head and Neck Surgery, UHL, County Council of Östergötland, Linköping, Sweden
- Division of Oto-Rhino-Laryngology and Head and Neck Surgery, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- Center for Hearing and Communication Research, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Section of Otorhinolaryngology, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
- * E-mail:
| | - Piyaporn Siratirakun
- Center for Hearing and Communication Research, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Section of Otorhinolaryngology, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Cecilia Lundberg
- CNS Gene Therapy Unit, Dept. of Experimental Medical Science, Lund University, Lund, Sweden
| | - Ulrica Englund-Johansson
- Department of Ophthalmology, Institution of Clinical Sciences in Lund, Lund University, Lund, Sweden
| |
Collapse
|
87
|
IL-6 treatment increases the survival of retinal ganglion cells in vitro: The role of adenosine A1 receptor. Biochem Biophys Res Commun 2013; 430:512-8. [DOI: 10.1016/j.bbrc.2012.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 12/03/2012] [Indexed: 11/23/2022]
|
88
|
Inducible regulation of GDNF expression in human neural stem cells. SCIENCE CHINA-LIFE SCIENCES 2012; 56:32-9. [PMID: 23269553 DOI: 10.1007/s11427-012-4424-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 11/02/2012] [Indexed: 10/27/2022]
|
89
|
Joo KM, Kang BG, Yeon JY, Cho YJ, An JY, Song HS, Won JH, Kim SJ, Hong SC, Nam DH. Experimental and clinical factors influencing long-term stable in vitro expansion of multipotent neural cells from human adult temporal lobes. Exp Neurol 2012. [PMID: 23201097 DOI: 10.1016/j.expneurol.2012.11.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Autologous adult human neural stem cells may be used for regenerative cell therapies bypass potential ethical problems. However, stable in vitro expansion protocols and experimental/clinical factors influencing primary cultures need to be further elucidated for clinically applicable techniques. To address these issues, we obtained biopsy specimens from 23 temporal lobe epilepsy patients and adult human multipotent neural cells (ahMNCs) were primarily cultured in a defined attachment culture condition. When the success of primary cultures was defined as stable expansion of cells (>ten in vitro passages) and expression of NSC markers, success rate of the primary culture was 39% (nine of 23 temporal lobes). During the long-term expansion, expressions of NSC markers and differentiation potentials into astrocytes and neurons were maintained. After the 18th sub-culture, spontaneous senescence and differentiation were observed, and the cultivated ahMNCs ceased their proliferation. The culture results were not affected by seizure characteristics; however, an older age (>40 years) and a smaller sample volume (<2 ml) were found to exert negative influences on the primary culture results. Furthermore therapeutic effects of ahMNCs against stroke were analyzed in an animal model. Transplantation of ahMNCs cells reduced infarction volumes and enhanced motor activity, significantly. The results here would provide promising experimental and clinical strategy of using patient-specific autologous ahMNCs in regenerative medicine in the future.
Collapse
Affiliation(s)
- Kyeung Min Joo
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, #300 Cheoncheon-dong, Jangan-gu, Suwon, Gyeonggi-do, 440-746, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Mothe AJ, Tator CH. Advances in stem cell therapy for spinal cord injury. J Clin Invest 2012; 122:3824-34. [PMID: 23114605 DOI: 10.1172/jci64124] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating condition producing great personal and societal costs and for which there is no effective treatment. Stem cell transplantation is a promising therapeutic strategy, though much preclinical and clinical research work remains. Here, we briefly describe SCI epidemiology, pathophysiology, and experimental and clinical stem cell strategies. Research in stem cell biology and cell reprogramming is rapidly advancing, with the hope of moving stem cell therapy closer to helping people with SCI. We examine issues important for clinical translation and provide a commentary on recent developments, including termination of the first human embryonic stem cell transplantation trial in human SCI.
Collapse
Affiliation(s)
- Andrea J Mothe
- Toronto Western Research Institute and Krembil Neuroscience Centre, Toronto Western Hospital, Toronto, Ontario, Canada
| | | |
Collapse
|
91
|
Hibaoui Y, Feki A. Human pluripotent stem cells: applications and challenges in neurological diseases. Front Physiol 2012; 3:267. [PMID: 22934023 PMCID: PMC3429043 DOI: 10.3389/fphys.2012.00267] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 06/25/2012] [Indexed: 12/16/2022] Open
Abstract
The ability to generate human pluripotent stem cells (hPSCs) holds great promise for the understanding and the treatment of human neurological diseases in modern medicine. The hPSCs are considered for their in vitro use as research tools to provide relevant cellular model for human diseases, drug discovery, and toxicity assays and for their in vivo use in regenerative medicine applications. In this review, we highlight recent progress, promises, and challenges of hPSC applications in human neurological disease modeling and therapies.
Collapse
Affiliation(s)
- Youssef Hibaoui
- Stem Cell Research Laboratory, Department of Obstetrics and Gynecology, Geneva University Hospitals Geneva, Switzerland
| | | |
Collapse
|
92
|
Subburaju S, Benes FM. Induction of the GABA cell phenotype: an in vitro model for studying neurodevelopmental disorders. PLoS One 2012; 7:e33352. [PMID: 22457755 PMCID: PMC3310062 DOI: 10.1371/journal.pone.0033352] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 02/10/2012] [Indexed: 12/30/2022] Open
Abstract
Recent studies of the hippocampus have suggested that a network of genes is associated with the regulation of the GAD₆₇ (GAD1) expression and may play a role in γ-amino butyric acid (GABA) dysfunction in schizophrenia (SZ) and bipolar disorder (BD). To obtain a more detailed understanding of how GAD₆₇ regulation may result in GABAergic dysfunction, we have developed an in vitro model in which GABA cells are differentiated from the hippocampal precursor cell line, HiB5. Growth factors, such as PDGF, and BDNF, regulate the GABA phenotype by inducing the expression of GAD₆₇ and stimulating the growth of cellular processes, many with growth cones that form appositions with the cell bodies and processes of other GAD₆₇-positive cells. These changes are associated with increased expression of acetylated tubulin, microtubule-associated protein 2 (MAP2) and the post-synaptic density protein 95 (PSD95). The addition of BDNF, together with PDGF, increases the levels of mRNA and protein for GAD₆₇, as well as the high affinity GABA uptake protein, GAT1. These changes are associated with increased concentrations of GABA in the cytoplasm of "differentiated" HiB5 neurons. In the presence of Ca²⁺ and K⁺, newly synthesized GABA is released extracellularly. When the HiB5 cells appear to be fully differentiated, they also express GAD₆₅, parvalbumin and calbindin, and GluR subtypes as well as HDAC1, DAXX, PAX5, Runx2, associated with GAD₆₇ regulation. Overall, these results suggest that the HiB5 cells can differentiate into functionally mature GABA neurons in the presence of gene products that are associated with GAD₆₇ regulation in the adult hippocampus.
Collapse
Affiliation(s)
- Sivan Subburaju
- Program in Structural and Molecular Neuroscience, McLean Hospital, Belmont, Massachusetts, United States of America
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Francine M. Benes
- Program in Structural and Molecular Neuroscience, McLean Hospital, Belmont, Massachusetts, United States of America
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
93
|
Lundberg J, Södersten E, Sundström E, Le Blanc K, Andersson T, Hermanson O, Holmin S. Targeted Intra-arterial Transplantation of Stem Cells to the Injured CNS is more Effective than Intravenous Administration: Engraftment is Dependent on Cell Type and Adhesion Molecule Expression. Cell Transplant 2012; 21:333-43. [DOI: 10.3727/096368911x576036] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Stem cell transplantation procedures using intraparenchymal injections cause tissue injury in addition to associated surgical risks. Intravenous cell administration give engraftment in parenchymal lesions although the method has low efficacy and specificity. In pathological conditions with inflammation, such as traumatic brain injury, there is a transient up-regulation of ICAM-1 and VCAM-1 which might provide environmental cues for migration of stem cells from blood to parenchyma. The aim of this study was to i) analyze the effect of intra-arterial administration on cellular engraftment, ii) compare engraftment and side effects between three different stem cell systems, and iii) analyze gene expression in these three systems. We performed specific intra-arterial transplantations with human mesenchymal stem cells (hMSCs), human neural progenitor cells (hNPCs), and rat neural progenitor cells (rNPCs) in a rat model of traumatic brain injury. These results were compared to the intravenous route for each cell type, respectively. Analysis of engraftment and recipient characterization was performed by immunohistochemistry. We further characterized the different types of cells by microarray and RT-qPCR analysis. Specific intra-arterial transplantations produced significantly higher engraftment compared to intravenous transplantation with hMSCs and rNPCs. No engraftment was detected after intra-arterial or intravenous administration of hNPCs. Characterization of integrin expression indicated that CD49dVCAM-1 and possibly ICAM-1 interactions through CD18 and CD11a, respectively, are important for engraftment after intravascular cell administration. No side effects, such as thromboembolic complications, were detected. When translating stem cell therapies to clinical practice, the route of transplantation and the properties of the cell lines (homing, diapedesis, and migration) become important. This study supports the use of selective intra-arterial transplantation for improving engraftment after traumatic brain injury. In addition, we conclude that careful analysis of cells intended for local, intra-arterial transplantation with respect to integrin expression is important.
Collapse
Affiliation(s)
- Johan Lundberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Erik Södersten
- DBRM, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Erik Sundström
- Division of Neurodegeneration, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stiftelsen Stockholms Sjukhem, Stockholm, Sweden
| | - Katarina Le Blanc
- Department of Laboratory Medicine, Division of Clinical Immunology, Karolinska Institutet, Stockholm, Sweden
- Hematology Center, Karolinska University Hospital, Stockholm, Sweden
| | - Tommy Andersson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Ola Hermanson
- DBRM, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Staffan Holmin
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
94
|
Shi B, Deng L, Shi X, Dai S, Zhang H, Wang Y, Bi J, Guo M. The enhancement of neural stem cell survival and growth by coculturing with expanded Sertoli cells in vitro. Biotechnol Prog 2011; 28:196-205. [PMID: 22109810 DOI: 10.1002/btpr.720] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 08/20/2011] [Indexed: 12/19/2022]
Abstract
Sertoli cells (SCs) have been described as the "nurse cells" of testis to provide essential growth factors and to create a proper environment for the development of other cells (e.g., germinal and neural stem cell). However, the physiological functions of the SCs obtained from different culture conditions are different in a coculturing system, and thus the optimal SC culturing condition should be investigated in vitro. In this paper, primary Sertoli cells were isolated from a 12-day-old mouse and expanded in two different culture conditions: a two dimensional (2D) plastic tissue disc and a three dimensional (3D) microcarrier culture system. They were then cocultured with neural stem cells (NSCs) isolated from 14-day-old mouse embryos. The metabolic activities of SCs(2D) (SCs in 2D) and SCs(3D) (SCs in 3D) and the amount of proteins secreted from two culturing systems were compared. The results show that the metabolic activity and the amount of secreted proteins from SCs(3D) were higher than both from SCs(2D). Three coculturing groups: NSCs+SC(2D), NSCs+SC(3D), and NSCs +SC-conditioned medium (SCCM, control group) were also compared regarding cell morphology and the numbers of neurons, neural outgrowths and neurospheres. The quantity of neurons, neural outgrowths and neurospheres were the highest in the NSCs+SC(3D) group. SCs cultured in the 3D system had a strong trophic effect on NSCs and enhanced their survival and growth. Besides, the mRNA of trophic and nutritive factors such as Glial-cell-line-derived neurotrophic factor (GDNF) and Interleukin-1 α (IL-1 α) secreted by the SCs from both 2D and 3D culture system were analyzed by real time-PCR and gel assay. The mRNA transcription of GDNF and IL-1α is more apparent in the 3D culture system than that from the 2D one. The coculturing system of NSCs+SC(3D) is a promising candidate for future neural stem cell transplantation.
Collapse
Affiliation(s)
- Bingyang Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Bakhru S, Nain AS, Highley C, Wang J, Campbell P, Amon C, Zappe S. Direct and cell signaling-based, geometry-induced neuronal differentiation of neural stem cells. Integr Biol (Camb) 2011; 3:1207-14. [PMID: 22057544 DOI: 10.1039/c1ib00098e] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neural Stem Cells (NSCs) are multipotent precursors inhabiting the subventricular and hippocampal subgranular regions of the adult mammalian brain, able to self-renew and differentiate into neurons, astrocytes, and oligodendrocytes, the three primary neural cell types of the adult brain. NSC fate is influenced by the physical and chemical microenvironment experienced by the cell, both in vitro and in vivo. Towards characterizing the influence of topographical, geometric cues on NSC fate, we fabricated highly aligned, single- and double-layer polystyrene nanofiber meshes. Seeding of NSCs on laminin-coated fibers induces polarized NSC morphology and cellular elongation in the directions of fiber alignment, with cells extending membranous processes over hundreds of microns along the fiber surfaces. Additionally, these aligned fiber substrates promote neuronal lineage specification of NSCs with an efficiency of 82.3 ± 11.1% within days of seeding. Moreover, not only do cells on fibers yield neurons, but also neighboring cells in close proximity to those differentiating on aligned fibers, with an efficiency of 72.8 ± 9.7%. This neighboring, cell-induced differentiation occurs without cell-cell contact over millimetres away from the fibers, suggesting a paracrine signaling effect not previously reported for NSCs undergoing neurogenesis. In contrast, NSCs farther away from these fiber substrates nearly uniformly yield glia.
Collapse
Affiliation(s)
- Sasha Bakhru
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
96
|
Neural stem/progenitor cells from the adult human spinal cord are multipotent and self-renewing and differentiate after transplantation. PLoS One 2011; 6:e27079. [PMID: 22073257 PMCID: PMC3206885 DOI: 10.1371/journal.pone.0027079] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 10/10/2011] [Indexed: 12/18/2022] Open
Abstract
Neural stem/progenitor cell (NSPC) transplantation is a promising therapy for spinal cord injury (SCI). However, little is known about NSPC from the adult human spinal cord as a donor source. We demonstrate for the first time that multipotent and self-renewing NSPC can be cultured, passaged and transplanted from the adult human spinal cord of organ transplant donors. Adult human spinal cord NSPC require an adherent substrate for selection and expansion in EGF (epidermal growth factor) and FGF2 (fibroblast growth factor) enriched medium. NSPC as an adherent monolayer can be passaged for at least 9 months and form neurospheres when plated in suspension culture. In EGF/FGF2 culture, NSPC proliferate and primarily express nestin and Sox2, and low levels of markers for differentiating cells. Leukemia inhibitory factor (LIF) promotes NSPC proliferation and significantly enhances GFAP expression in hypoxia. In differentiating conditions in the presence of serum, these NSPC show multipotentiality, expressing markers of neurons, astrocytes, and oligodendrocytes. Dibutyryl cyclic AMP (dbcAMP) significantly enhances neuronal differentiation. We transplanted the multipotent NSPC into SCI rats and show that the xenografts survive, are post-mitotic, and retain the capacity to differentiate into neurons and glia. Together, these findings reveal that multipotent self-renewing NSPC cultured and passaged from adult human spinal cords of organ transplant donors, respond to exogenous factors that promote selective differentiation, and survive and differentiate after transplantation into the injured spinal cord.
Collapse
|
97
|
Wallman L, Åkesson E, Ceric D, Andersson PH, Day K, Hovatta O, Falci S, Laurell T, Sundström E. Biogrid--a microfluidic device for large-scale enzyme-free dissociation of stem cell aggregates. LAB ON A CHIP 2011; 11:3241-3248. [PMID: 21850297 DOI: 10.1039/c1lc20316a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Culturing stem cells as free-floating aggregates in suspension facilitates large-scale production of cells in closed systems, for clinical use. To comply with GMP standards, the use of substances such as proteolytic enzymes should be avoided. Instead of enzymatic dissociation, the growing cell aggregates may be mechanically cut at passage, but available methods are not compatible with large-scale cell production and hence translation into the clinic becomes a severe bottle-neck. We have developed the Biogrid device, which consists of an array of micrometerscale knife edges, micro-fabricated in silicon, and a manifold in which the microgrid is placed across the central fluid channel. By connecting one side of the Biogrid to a syringe or a pump and the other side to the cell culture, the culture medium with suspended cell aggregates can be aspirated, forcing the aggregates through the microgrid, and ejected back to the cell culture container. Large aggregates are thereby dissociated into smaller fragments while small aggregates pass through the microgrid unaffected. As proof-of-concept, we demonstrate that the Biogrid device can be successfully used for repeated passage of human neural stem/progenitor cells cultured as so-called neurospheres, as well as for passage of suspension cultures of human embryonic stem cells. We also show that human neural stem/progenitor cells tolerate transient pressure changes far exceeding those that will occur in a fluidic system incorporating the Biogrid microgrids. Thus, by using the Biogrid device it is possible to mechanically passage large quantities of cells in suspension cultures in closed fluidic systems, without the use of proteolytic enzymes.
Collapse
Affiliation(s)
- Lars Wallman
- Division of Nanobiotechnology, Department of Measurement Technology and Industrial Electrical Engineering, Lund University, P.O. Box 118, S-22100 Lund, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Abstract
Over the past 20 years, the conception of brain development has radically changed from a fixed and limited hierarchical process to a more plastic and continuous one. Most surprising, the field has learned that postnatal neurogenesis is not just a seasonal phenomenon in songbirds but a process that occurs across species and seasons. Astrocytes, whose primary role in the central nervous system was thought to be strictly supportive, have emerged as a heterogeneous population, a subset of which is the neural stem cell. Postnatal neurogenesis persists in specialized niches within the rostral subventricular zone and hippocampal dentate gyrus and, for a limited period, within the white matter tracts and external granular layer of the cerebellum. These specialized microenvironments are influenced by factors in the blood, cerebrospinal fluid, and local extracellular matrix. This article reviews the current understanding of adult neurogenesis, which is conserved across many vertebrate species, underscoring the value of animal models in past and present studies of human neurogenesis and neurogenic disease.
Collapse
Affiliation(s)
- R. M. Walton
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
99
|
Serre A, Snyder EY, Mallet J, Buchet D. Overexpression of basic helix-loop-helix transcription factors enhances neuronal differentiation of fetal human neural progenitor cells in various ways. Stem Cells Dev 2011; 21:539-53. [PMID: 21561385 DOI: 10.1089/scd.2011.0079] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In a perspective of regenerative medicine, multipotent human neural progenitor cells (hNPCs) offer a therapeutic advantage over pluripotent stem cells in that they are already invariantly "neurally committed" and lack tumorigenicity. However, some of their intrinsic properties, such as slow differentiation and uncontrolled multipotency, remain among the obstacles to their routine use for transplantation. Although rodent NPCs have been genetically modified in vitro to overcome some of these limitations, the translation of this strategy to human cells remains in its early stages. In the present study, we compare the actions of 4 basic helix-loop-helix transcription factors on the proliferation, specification, and terminal differentiation of hNPCs isolated from the fetal dorsal telencephalon. Consistent with their proneural activity, Ngn1, Ngn2, Ngn3, and Mash1 prompted rapid commitment of the cells. The Ngns induced a decrease in proliferation, whereas Mash1 maintained committed progenitors in a proliferative state. As opposed to Ngn1 and Ngn3, which had no effect on glial differentiation, Ngn2 induced an increase in astrocytes in addition to neurons, whereas Mash1 led to both neuronal and oligodendroglial specification. GABAergic, cholinergic, and motor neuron differentiations were considerably increased by overexpression of Ngn2 and, to a lesser extent, of Ngn3 and Mash1. Thus, we provide evidence that hNPCs can be efficiently, rapidly, and safely expanded in vitro as well as rapidly differentiated toward mature neural (typically neuronal) lineages by the overexpression of select proneural genes.
Collapse
Affiliation(s)
- Angéline Serre
- Centre de Recherche de l'Institut du Cerveau et de la Moelle Epinière, Université Pierre et Marie Curie-Paris 6, UMR-S975, Paris, France
| | | | | | | |
Collapse
|
100
|
Tunici P, Pellegatta S, Finocchiaro G. The potential of stem cells for the treatment of brain tumors and globoid cell leukodystrophy. Cytotechnology 2011; 41:93-101. [PMID: 19002946 DOI: 10.1023/a:1024818621377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Stem cells of different origin are under careful scrutiny as potential new tools for the treatment of several neurological diseases. The major focus of these reaserches have been neurodegenerative disorders, such as Huntington Chorea or Parkinson Disease (Shihabuddin et al., 1999). More recently attention has been devoted to their use for brain repair after stroke (Savitz et al., 2002). In this review we will focus on the potential of stem cell treatments for glioblastoma multiforme (Holland, 2000), the most aggressive primary brain tumor, and globoid cell leukodystrophy (Krabbe disease), a metabolic disorder of the white matter (Berger et al., 2001). These two diseases may offer a paradigm of what the stem cell approach may offer in term of treatment, alone or in combination with other therapeutic approaches. Two kinds of stem cells will be consideredhere: neural stem cells and hematopoietic stem cells, both obtained after birth. The review will focus on experimental models, with an eye on clinical perspectives.
Collapse
Affiliation(s)
- Patrizia Tunici
- Istituto Nazionale Neurologico Besta, Unit of Neuro-Oncology and Gene Therapy, Milan, Italy (Author for correspondence)
| | | | | |
Collapse
|