1
|
Mustapha O, Grochow T, Olopade J, Fietz SA. Neocortex neurogenesis and maturation in the African greater cane rat. Neural Dev 2023; 18:7. [PMID: 37833718 PMCID: PMC10571270 DOI: 10.1186/s13064-023-00175-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Neocortex development has been extensively studied in altricial rodents such as mouse and rat. Identification of alternative animal models along the "altricial-precocial" spectrum in order to better model and understand neocortex development is warranted. The Greater cane rat (GCR, Thyronomys swinderianus) is an indigenous precocial African rodent. Although basic aspects of brain development in the GCR have been documented, detailed information on neocortex development including the occurrence and abundance of the distinct types of neural progenitor cells (NPCs) in the GCR are lacking. METHODS GCR embryos and fetuses were obtained from timed pregnant dams between gestation days 50-140 and their neocortex was analyzed by immunofluorescence staining using characteristic marker proteins for NPCs, neurons and glia cells. Data were compared with existing data on closely related precocial and altricial species, i.e. guinea pig and dwarf rabbit. RESULTS The primary sequence of neuro- and gliogenesis, and neuronal maturation is preserved in the prenatal GCR neocortex. We show that the GCR exhibits a relatively long period of cortical neurogenesis of 70 days. The subventricular zone becomes the major NPC pool during mid-end stages of neurogenesis with Pax6 + NPCs constituting the major basal progenitor subtype in the GCR neocortex. Whereas dendrite formation in the GCR cortical plate appears to initiate immediately after the onset of neurogenesis, major aspects of axon formation and maturation, and astrogenesis do not begin until mid-neurogenesis. Similar to the guinea pig, the GCR neocortex exhibits a high maturation status, containing neurons with well-developed dendrites and myelinated axons and astrocytes at birth, thus providing further evidence for the notion that a great proportion of neocortex growth and maturation in precocial mammals occurs before birth. CONCLUSIONS Together, this work has deepened our understanding of neocortex development of the GCR, of the timing and the cellular differences that regulate brain growth and development within the altricial-precocial spectrum and its suitability as a research model for neurodevelopmental studies. The timelines of brain development provided by this study may serve as empirical reference data and foundation in future studies in order to model and better understand neurodevelopment and associated alterations.
Collapse
Affiliation(s)
- Oluwaseun Mustapha
- Neuroscience Unit, Department of Veterinary Anatomy, College of Veterinary Medicine, Federal University of Agriculture Abeokuta, Abeokuta, Ogun State, Nigeria
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Thomas Grochow
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - James Olopade
- Neuroscience Unit, Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Simone A Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
2
|
Lee S, Nam H, Joo KM, Lee SH. Advances in Neural Stem Cell Therapy for Spinal Cord Injury: Safety, Efficacy, and Future Perspectives. Neurospine 2022; 19:946-960. [PMID: 36351442 PMCID: PMC9816608 DOI: 10.14245/ns.2244658.329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating central nervous system injury that leads to severe disabilities in motor and sensory functions, causing significant deterioration in patients' quality of life. Owing to the complexity of SCI pathophysiology, there has been no effective treatment for reversing neural tissue damage and recovering neurological functions. Several novel therapies targeting different stages of pathophysiological mechanisms of SCI have been developed. Among these, treatments using stem cells have great potential for the regeneration of damaged neural tissues. In this review, we have summarized recent preclinical and clinical studies focusing on neural stem cells (NSCs). NSCs are multipotent cells with specific differentiation capabilities for neural lineage. Several preclinical studies have demonstrated the regenerative effects of transplanted NSCs in SCI animal models through both paracrine effects and direct neuronal differentiation, restoring synaptic connectivity and neural networks. Based on the positive results of several preclinical studies, phase I and II clinical trials using NSCs have been performed. Despite several hurdles and issues that need to be addressed in the clinical use of NSCs in patients with SCI, gradual progress in the technical development and therapeutic efficacy of NSCs treatments has enhanced the prospects for cell-based treatments in SCI.
Collapse
Affiliation(s)
- Sungjoon Lee
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Nam
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea,Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea,Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Kyeung-Min Joo
- Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Korea,Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea,Corresponding Author Kyeung-Min Joo Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Korea
| | - Sun-Ho Lee
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea,Stem Cell and Regenerative Medicine Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea,Co-corresponding Author Sun-Ho Lee Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
| |
Collapse
|
3
|
Ha GH, Kim EJ, Park JS, Kim JE, Nam H, Yeon JY, Lee SH, Lee K, Kim CK, Joo KM. JAK2/STAT3 pathway mediates neuroprotective and pro-angiogenic treatment effects of adult human neural stem cells in middle cerebral artery occlusion stroke animal models. Aging (Albany NY) 2022; 14:8944-8969. [PMID: 36446389 PMCID: PMC9740376 DOI: 10.18632/aging.204410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022]
Abstract
Mismatches between pre-clinical and clinical results of stem cell therapeutics for ischemic stroke limit their clinical applicability. To overcome these discrepancies, precise planning of pre-clinical experiments that can be translated to clinical trials and the scientific elucidation of treatment mechanisms is important. In this study, adult human neural stem cells (ahNSCs) derived from temporal lobe surgical samples were used (to avoid ethical and safety issues), and their therapeutic effects on ischemic stroke were examined using middle cerebral artery occlusion animal models. 5 × 105 ahNSCs was directly injected into the lateral ventricle of contralateral brain hemispheres of immune suppressed rat stroke models at the subacute phase of stroke. Compared with the mock-treated group, ahNSCs reduced brain tissue atrophy and neurological sensorimotor and memory functional loss. Tissue analysis demonstrated that the significant therapeutic effects were mediated by the neuroprotective and pro-angiogenic activities of ahNSCs, which preserved neurons in ischemic brain areas and decreased reactive astrogliosis and microglial activation. The neuroprotective and pro-angiogenic effects of ahNSCs were validated in in vitro stroke models and were induced by paracrine factors excreted by ahNSCs. When the JAK2/STAT3 signaling pathway was inhibited by a specific inhibitor, AG490, the paracrine neuroprotective and pro-angiogenic effects of ahNSCs were reversed. This pre-clinical study that closely simulated clinical settings and provided treatment mechanisms of ahNSCs for ischemic stroke may aid the development of protocols for subsequent clinical trials of ahNSCs and the realization of clinically available stem cell therapeutics for ischemic stroke.
Collapse
Affiliation(s)
- Geun-Hyoung Ha
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea
| | - Eun Ji Kim
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea
| | - Jee Soo Park
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea
| | - Ji Eun Kim
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea
| | - Hyun Nam
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea,Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, South Korea,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Je Young Yeon
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Sun-Ho Lee
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, South Korea
| | - Kyunghoon Lee
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, South Korea,Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea
| | - Chung Kwon Kim
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea,Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea
| | - Kyeung Min Joo
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea,Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea,Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, South Korea,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, South Korea,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, South Korea,Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea
| |
Collapse
|
4
|
Park TIH, Smyth LCD, Aalderink M, Woolf ZR, Rustenhoven J, Lee K, Jansson D, Smith A, Feng S, Correia J, Heppner P, Schweder P, Mee E, Dragunow M. Routine culture and study of adult human brain cells from neurosurgical specimens. Nat Protoc 2022; 17:190-221. [PMID: 35022619 DOI: 10.1038/s41596-021-00637-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 09/21/2021] [Indexed: 12/22/2022]
Abstract
When modeling disease in the laboratory, it is important to use clinically relevant models. Patient-derived human brain cells grown in vitro to study and test potential treatments provide such a model. Here, we present simple, highly reproducible coordinated procedures that can be used to routinely culture most cell types found in the human brain from single neurosurgically excised brain specimens. The cell types that can be cultured include dissociated cultures of neurons, astrocytes, microglia, pericytes and brain endothelial and neural precursor cells, as well as explant cultures of the leptomeninges, cortical slice cultures and brain tumor cells. The initial setup of cultures takes ~2 h, and the cells are ready for further experiments within days to weeks. The resulting cells can be studied as purified or mixed population cultures, slice cultures and explant-derived cultures. This protocol therefore enables the investigation of human brain cells to facilitate translation of neuroscience research to the clinic.
Collapse
Affiliation(s)
- Thomas I-H Park
- Hugh Green Biobank & Department of Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Neurosurgical Research Unit, Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Leon C D Smyth
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Miranda Aalderink
- Hugh Green Biobank & Department of Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Zoe R Woolf
- Hugh Green Biobank & Department of Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Justin Rustenhoven
- Center for Brain Immunology and Glia (BIG), Washington University, St. Louis, MO, USA
| | - Kevin Lee
- Department of Physiology, Faculty of Medical Science and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Deidre Jansson
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine VISN 20 Mental Illness Research, Education and Clinical Centre (MIRECC), VA Puget Sound Health Care System, Seattle, WA, USA
| | - Amy Smith
- Hugh Green Biobank & Department of Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Sheryl Feng
- Hugh Green Biobank & Department of Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Jason Correia
- Neurosurgical Research Unit, Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Peter Heppner
- Neurosurgical Research Unit, Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Patrick Schweder
- Neurosurgical Research Unit, Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Edward Mee
- Neurosurgical Research Unit, Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Mike Dragunow
- Hugh Green Biobank & Department of Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
- Neurosurgical Research Unit, Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
5
|
Park TIH, Waldvogel HJ, Montgomery JM, Mee EW, Bergin PS, Faull RLM, Dragunow M, Curtis MA. Identifying Neural Progenitor Cells in the Adult Human Brain. Methods Mol Biol 2022; 2389:125-154. [PMID: 34558008 DOI: 10.1007/978-1-0716-1783-0_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discovery, in 1998, that the adult human brain contains at least two populations of progenitor cells and that progenitor cells are upregulated in response to a range of degenerative brain diseases has raised hopes for their use in replacing dying brain cells. Since these early findings, the race has been on to understand the biology of progenitor cells in the human brain, and they have now been isolated and studied in many major neurodegenerative diseases. Before these cells can be exploited for cell replacement purposes, it is important to understand how to (1) locate them, (2) label them, (3) determine what receptors they express, (4) isolate them, and (5) examine their electrophysiological properties when differentiated. In this chapter we have described the methods we use for studying progenitor cells in the adult human brain and in particular the tissue processing, immunohistochemistry, autoradiography, progenitor cell culture, and electrophysiology on brain cells. The Neurological Foundation of New Zealand Human Brain Bank has been receiving human tissue for approximately 25 years during which time we have developed a number of unique ways to examine and isolate progenitor cells from resected surgical specimens as well as from postmortem brain tissue. There are ethical and technical considerations that are unique to working with human brain tissue, and these, as well as the processing of this tissue and the culturing of it for the purpose of studying progenitor cells, are the topic of this chapter.
Collapse
Affiliation(s)
- Thomas I H Park
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.,Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Edward W Mee
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.,Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Peter S Bergin
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.,Department of Neurology, Auckland City Hospital, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.,Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.,Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand. .,Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
6
|
Abdolahi S, Aligholi H, Khodakaram-Tafti A, Khaleghi Ghadiri M, Stummer W, Gorji A. Improvement of Rat Spinal Cord Injury Following Lentiviral Vector-Transduced Neural Stem/Progenitor Cells Derived from Human Epileptic Brain Tissue Transplantation with a Self-assembling Peptide Scaffold. Mol Neurobiol 2021; 58:2481-2493. [PMID: 33443682 PMCID: PMC8128971 DOI: 10.1007/s12035-020-02279-5] [Citation(s) in RCA: 11] [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: 07/02/2020] [Accepted: 12/30/2020] [Indexed: 12/29/2022]
Abstract
Spinal cord injury (SCI) is a disabling neurological disorder that causes neural circuit dysfunction. Although various therapies have been applied to improve the neurological outcomes of SCI, little clinical progress has been achieved. Stem cell-based therapy aimed at restoring the lost cells and supporting micromilieu at the site of the injury has become a conceptually attractive option for tissue repair following SCI. Adult human neural stem/progenitor cells (hNS/PCs) were obtained from the epileptic human brain specimens. Induction of SCI was followed by the application of lentiviral vector-mediated green fluorescent protein-labeled hNS/PCs seeded in PuraMatrix peptide hydrogel (PM). The co-application of hNS/PCs and PM at the SCI injury site significantly enhanced cell survival and differentiation, reduced the lesion volume, and improved neurological functions compared to the control groups. Besides, the transplanted hNS/PCs seeded in PM revealed significantly higher migration abilities into the lesion site and the healthy host tissue as well as a greater differentiation into astrocytes and neurons in the vicinity of the lesion as well as in the host tissue. Our data suggest that the transplantation of hNS/PCs seeded in PM could be a promising approach to restore the damaged tissues and improve neurological functions after SCI.
Collapse
Affiliation(s)
- Sara Abdolahi
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Hadi Aligholi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Walter Stummer
- Department of Neurosurgery, Westfälische Wilhelms-Universität, Münster, Germany
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
- Epilepsy Research Center, Department of Neurology and Institute for Translational Neurology, Westfälische Wilhelms-Universität Münster, 48149, Münster, Germany.
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Kalusa M, Heinrich MD, Sauerland C, Morawski M, Fietz SA. Developmental Differences in Neocortex Neurogenesis and Maturation Between the Altricial Dwarf Rabbit and Precocial Guinea Pig. Front Neuroanat 2021; 15:678385. [PMID: 34135738 PMCID: PMC8200626 DOI: 10.3389/fnana.2021.678385] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/06/2021] [Indexed: 11/13/2022] Open
Abstract
Mammals are born on a precocial-altricial continuum. Altricial species produce helpless neonates with closed distant organs incapable of locomotion, whereas precocial species give birth to well-developed young that possess sophisticated sensory and locomotor capabilities. Previous studies suggest that distinct patterns of cortex development differ between precocial and altricial species. This study compares patterns of neocortex neurogenesis and maturation in the precocial guinea pig and altricial dwarf rabbit, both belonging to the taxon of Glires. We show that the principal order of neurodevelopmental events is preserved in the neocortex of both species. Moreover, we show that neurogenesis starts at a later postconceptional day and takes longer in absolute gestational days in the precocial than the altricial neocortex. Intriguingly, our data indicate that the dwarf rabbit neocortex contains a higher abundance of highly proliferative basal progenitors than the guinea pig, which might underlie its higher encephalization quotient, demonstrating that the amount of neuron production is determined by complex regulation of multiple factors. Furthermore, we show that the guinea pig neocortex exhibits a higher maturation status at birth, thus providing evidence for the notions that precocial species might have acquired the morphological machinery required to attain their high functional state at birth and that brain expansion in the precocial newborn is mainly due to prenatally initiating processes of gliogenesis and neuron differentiation instead of increased neurogenesis. Together, this study reveals important insights into the timing and cellular differences that regulate mammalian brain growth and maturation and provides a better understanding of the evolution of mammalian altriciality and presociality.
Collapse
Affiliation(s)
- Mirjam Kalusa
- Faculty of Veterinary Medicine, Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, Leipzig, Germany
| | - Maren D. Heinrich
- Faculty of Veterinary Medicine, Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, Leipzig, Germany
| | - Christine Sauerland
- Faculty of Veterinary Medicine, Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, Leipzig, Germany
| | - Markus Morawski
- Medical Faculty, Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Simone A. Fietz
- Faculty of Veterinary Medicine, Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
8
|
Harberts J, Kusch M, O’Sullivan J, Zierold R, Blick RH. A Temperature-Controlled Patch Clamp Platform Demonstrated on Jurkat T Lymphocytes and Human Induced Pluripotent Stem Cell-Derived Neurons. Bioengineering (Basel) 2020; 7:E46. [PMID: 32455868 PMCID: PMC7355542 DOI: 10.3390/bioengineering7020046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 01/07/2023] Open
Abstract
Though patch clamping at room temperature is a widely disseminated standard procedure in the electrophysiological community, it does not represent the biological system in mammals at around 37 °C. In order to better mimic the natural environment in electrophysiological studies, we present a custom-built, temperature-controlled patch clamp platform for upright microscopes, which can easily be adapted to any upright patch clamp setup independently, whether commercially available or home built. Our setup can both cool and heat the platform having only small temperature variations of less than 0.5 °C. We demonstrate our setup with patch clamp measurements at 36 °C on Jurkat T lymphocytes and human induced pluripotent stem cell-derived neurons. Passive membrane parameters and characteristic electrophysiological properties, such as the gating properties of voltage-gated ion channels and the firing of action potentials, are compared to measurements at room temperature. We observe that many processes that are not explicitly considered as temperature dependent show changes with temperature. Thus, we believe in the need of a temperature control in patch clamp measurements if improved physiological conditions are required. Furthermore, we advise researchers to only compare electrophysiological results directly that have been measured at similar temperatures since small variations in cellular properties might be caused by temperature alterations.
Collapse
Affiliation(s)
- Jann Harberts
- Center for Hybrid Nanostructures, Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany; (J.H.); (M.K.); (R.H.B.)
| | - Max Kusch
- Center for Hybrid Nanostructures, Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany; (J.H.); (M.K.); (R.H.B.)
| | - John O’Sullivan
- Center for Hybrid Nanostructures, Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany; (J.H.); (M.K.); (R.H.B.)
- Department of Physics and Astronomy, University College London, London WC1E 6BT , UK
| | - Robert Zierold
- Center for Hybrid Nanostructures, Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany; (J.H.); (M.K.); (R.H.B.)
| | - Robert H. Blick
- Center for Hybrid Nanostructures, Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany; (J.H.); (M.K.); (R.H.B.)
- Material Science and Engineering, College of Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
9
|
Neurotoxicity of silver nanoparticles stabilized with different coating agents: In vitro response of neuronal precursor cells. Food Chem Toxicol 2020; 136:110935. [DOI: 10.1016/j.fct.2019.110935] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/18/2019] [Accepted: 10/29/2019] [Indexed: 12/31/2022]
|
10
|
Harberts J, Haferkamp U, Haugg S, Fendler C, Lam D, Zierold R, Pless O, Blick RH. Interfacing human induced pluripotent stem cell-derived neurons with designed nanowire arrays as a future platform for medical applications. Biomater Sci 2020; 8:2434-2446. [DOI: 10.1039/d0bm00182a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanostructured substrates such as nanowire arrays form a powerful tool for building next-generation medical devices.
Collapse
Affiliation(s)
- Jann Harberts
- Center for Hybrid Nanostructures
- Universität Hamburg
- 22761 Hamburg
- Germany
| | | | - Stefanie Haugg
- Center for Hybrid Nanostructures
- Universität Hamburg
- 22761 Hamburg
- Germany
| | - Cornelius Fendler
- Center for Hybrid Nanostructures
- Universität Hamburg
- 22761 Hamburg
- Germany
| | - Dennis Lam
- Fraunhofer IME ScreeningPort
- 22525 Hamburg
- Germany
| | - Robert Zierold
- Center for Hybrid Nanostructures
- Universität Hamburg
- 22761 Hamburg
- Germany
| | - Ole Pless
- Fraunhofer IME ScreeningPort
- 22525 Hamburg
- Germany
| | - Robert H. Blick
- Center for Hybrid Nanostructures
- Universität Hamburg
- 22761 Hamburg
- Germany
- Material Science and Engineering
| |
Collapse
|
11
|
Culturing primary neurons from rat hippocampus and cortex. Neuronal Signal 2019; 3:NS20180207. [PMID: 32714598 PMCID: PMC7363313 DOI: 10.1042/ns20180207] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 01/21/2023] Open
Abstract
Primary neurons from rodent brain hippocampus and cortex have served as important tools in biomedical research over the years. However, protocols for the preparation of primary neurons vary, which often lead to conflicting results. This report provides a robust and reliable protocol for the production of primary neuronal cultures from the cortex and hippocampus with minimal contribution of non-neuronal cells. The neurons were grown in serum-free media and maintained for several weeks without any additional feeder cells. The neuronal cultures maintained according to this protocol differentiate and by 3 weeks develop extensive axonal and dendritic branching. The cultures produced by this method show excellent reproducibility and can be used for histological, molecular and biochemical methods.
Collapse
|
12
|
Harbom LJ, Rudisill TL, Michel N, Litwa KA, Beenhakker MP, McConnell MJ. The effect of rho kinase inhibition on morphological and electrophysiological maturity in iPSC-derived neurons. Cell Tissue Res 2018; 375:641-654. [PMID: 30406823 DOI: 10.1007/s00441-018-2942-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023]
Abstract
Induced pluripotent stem cell (iPSC)-derived neurons permit the study of neurogenesis and neurological disease in a human setting. However, the electrophysiological properties of iPSC-derived neurons are consistent with those observed in immature cortical neurons, including a high membrane resistance depolarized resting membrane potential and immature firing properties, limiting their use in modeling neuronal activity in adult cells. Based on the proven association between inhibiting rho kinase (ROCK) and increased neurite complexity, we seek to determine if short-term ROCK inhibition during the first 1-2 weeks of differentiation would increase morphological complexity and electrophysiological maturity after several weeks of differentiation. While inhibiting ROCK resulted in increased neurite formation after 24 h, this effect did not persist at 3 and 6 weeks of age. Additionally, there was no effect of ROCK inhibition on electrophysiological properties at 2-3, 6, or 12 weeks of age, despite an increase in evoked and spontaneous firing and a more hyperpolarized resting membrane potential over time. These results indicate that while there is a clear effect of time on electrophysiological maturity, ROCK inhibition did not accelerate maturity.
Collapse
Affiliation(s)
- Lise J Harbom
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
- Department of Biochemistry and Molecular Genetics and Neuroscience, Centers for Brain Immunology and Glia, Public Health Genomics, and Children's Health Research, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Taylor L Rudisill
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Nadine Michel
- Department of Biochemistry and Molecular Genetics and Neuroscience, Centers for Brain Immunology and Glia, Public Health Genomics, and Children's Health Research, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Karen A Litwa
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Mark P Beenhakker
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA.
| | - Michael J McConnell
- Department of Biochemistry and Molecular Genetics and Neuroscience, Centers for Brain Immunology and Glia, Public Health Genomics, and Children's Health Research, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA.
| |
Collapse
|
13
|
Yeon JY, Hwang JY, Lee HW, Pyeon HJ, Won JS, Noh YJ, Nam H, Joo KM. Optimized Clump Culture Methods for Adult Human Multipotent Neural Cells. Int J Mol Sci 2018; 19:ijms19113380. [PMID: 30380605 PMCID: PMC6274905 DOI: 10.3390/ijms19113380] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/30/2022] Open
Abstract
Adult human multipotent neural cell (ahMNC) is a candidate for regeneration therapy for neurodegenerative diseases. Here, we developed a primary clump culture method for ahMNCs to increase the efficiency of isolation and in vitro expansion. The same amount of human temporal lobe (1 g) was partially digested and then filtered through strainers with various pore sizes, resulting in four types of clumps: Clump I > 100 µm, 70 µm < Clump II < 100 µm, 40 µm < Clump III < 70 µm, and Clump IV < 40 µm. At 3 and 6 days after culture, Clump II showed significantly higher number of colonies than the other Clumps. Moreover, ahMNCs derived from Clump II (ahMNCs-Clump II) showed stable proliferation, and shortened the time to first passage from 19 to 15 days, and the time to 1 × 109 cells from 42 to 34 days compared with the previous single-cell method. ahMNCs-Clump II had neural differentiation and pro-angiogenic potentials, which are the characteristics of ahMNCs. In conclusion, the novel clump culture method for ahMNCs has significantly higher efficiency than previous techniques. Considering the small amount of available human brain tissue, the clump culture method would promote further clinical applications of ahMNCs.
Collapse
Affiliation(s)
- Je Young Yeon
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
| | - Ji-Yoon Hwang
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
| | - Hye Won Lee
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
| | - Hee-Jang Pyeon
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
| | - Jeong-Seob Won
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea.
| | - Yoo-Jung Noh
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
| | - Hyun Nam
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
| | - Kyeung Min Joo
- Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
- Department of Anatomy & Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea.
| |
Collapse
|
14
|
Cortical and spinal conditioned media modify the inward ion currents and excitability and promote differentiation of human striatal primordium. J Chem Neuroanat 2018; 90:87-97. [DOI: 10.1016/j.jchemneu.2017.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 11/18/2022]
|
15
|
Jahanbazi Jahan-Abad A, Sahab Negah S, Hosseini Ravandi H, Ghasemi S, Borhani-Haghighi M, Stummer W, Gorji A, Khaleghi Ghadiri M. Human Neural Stem/Progenitor Cells Derived From Epileptic Human Brain in a Self-Assembling Peptide Nanoscaffold Improve Traumatic Brain Injury in Rats. Mol Neurobiol 2018; 55:9122-9138. [PMID: 29651746 DOI: 10.1007/s12035-018-1050-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 03/27/2018] [Indexed: 01/09/2023]
Abstract
Traumatic brain injury (TBI) is a disruption in the brain functions following a head trauma. Cell therapy may provide a promising treatment for TBI. Among different cell types, human neural stem cells cultured in self-assembling peptide scaffolds have been suggested as a potential novel method for cell replacement treatment after TBI. In the present study, we accessed the effects of human neural stem/progenitor cells (hNS/PCs) derived from epileptic human brain and human adipose-derived stromal/stem cells (hADSCs) seeded in PuraMatrix hydrogel (PM) on brain function after TBI in an animal model of brain injury. hNS/PCs were isolated from patients with medically intractable epilepsy undergone epilepsy surgery. hNS/PCs and hADSCs have the potential for proliferation and differentiation into both neuronal and glial lineages. Assessment of the growth characteristics of hNS/PCs and hADSCs revealed that the hNS/PCs doubling time was significantly longer and the growth rate was lower than hADSCs. Transplantation of hNS/PCs and hADSCs seeded in PM improved functional recovery, decreased lesion volume, inhibited neuroinflammation, and reduced the reactive gliosis at the injury site. The data suggest the transplantation of hNS/PCs or hADSCs cultured in PM as a promising treatment option for cell replacement therapy in TBI.
Collapse
Affiliation(s)
- Ali Jahanbazi Jahan-Abad
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Department of Clinical Biochemistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Sahab Negah
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Sedigheh Ghasemi
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | | | - Walter Stummer
- Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran. .,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany. .,Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany. .,Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Robert-Koch-Strasse 45, 48149, Münster, Germany.
| | | |
Collapse
|
16
|
Generation of Functional Dopaminergic Neurons from Reprogramming Fibroblasts by Nonviral-based Mesoporous Silica Nanoparticles. Sci Rep 2018; 8:11. [PMID: 29311646 PMCID: PMC5758610 DOI: 10.1038/s41598-017-18324-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/01/2017] [Indexed: 12/11/2022] Open
Abstract
Direct-lineage conversion of the somatic cell by reprogramming, in which mature cells were fully converted into a variety of other cell types bypassing an intermediate pluripotent state, is a promising regenerative medicine approach. Due to the risk of tumorigenesis by viral methods, a non-viral carrier for the delivery of reprogramming factors is very desirable. This study utilized the mesoporous silica nanoparticles (MSNs) as a non-viral delivery system for transduction of the three key factors to achieve conversion of mouse fibroblasts (MFs) into functional dopaminergic neuron-like cells (denoted as fDA-neurons). At the same time, a neurogenesis inducer, ISX-9, was co-delivered with the MSNs to promote the direct conversion of neuron-like cells. Good transfection efficiency of plasmid@MSN allowed repeated dosing to maintain high exogenous gene expression analyzed by qPCR and the changes in neural function markers were monitored. To further validate the dopaminergic function and the electrophysiological properties of fDA-neurons, the results of ELISA assay showed the high levels of secreted-dopamine in the conditional medium and rich Na+/K+-channels were observed in the fDA-neurons on Day 22. The results demonstrated that MSN nanocarrier is effective in delivering the reprogramming factors for the conversion of functional dopaminergic neurons from adult somatic cells.
Collapse
|
17
|
Johnsen EO, Frøen RC, Olstad OK, Nicolaissen B, Petrovski G, Moe MC, Noer A. Proliferative Cells Isolated from the Adult Human Peripheral Retina only Transiently Upregulate Key Retinal Markers upon Induced Differentiation. Curr Eye Res 2017; 43:340-349. [PMID: 29161152 DOI: 10.1080/02713683.2017.1403630] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Purpose/Aim: The adult human retina has limited regenerative potential, and severe injury will result in permanent damage. Lower vertebrates handle retinal injury by activating neural stem cells (NSCs) in the ciliary marginal zone (CMZ). Müller glia-like cells expressing markers of NSCs are also present in the peripheral retina (PR) of the adult human eye, leading to the hypothesis that a CMZ-like zone might exists also in humans. In order to shed further light on this hypothesis we investigated the in vitro differentiation potential of proliferative cells isolated from the adult human PR towards a retinal phenotype. MATERIALS AND METHODS Proliferative cells were isolated from the peripheral retina of human eyes (n = 6) within 24 to 48 hours post mortem and further expanded for 2 or 3 passages before being differentiated for 1-3 weeks. Gene expression was analyzed by microarray and qRT-PCR analysis, while protein expression was identified by immunocytochemistry. RESULTS A high density of cells co-staining with markers for progenitor cells and Müller glia was found in situ in the PR. Cells isolated from this region and cultured adherently showed fibrillary processes and were positive for the immature marker Nestin and the glial marker GFAP, while a few co-expressed PAX6. After 7 days of differentiation, there was a transient upregulation of early and mature photoreceptor markers, including NRL, CRX, RHO and RCVRN, as well as the Müller cell and retinal pigmented epithelium (RPE) marker CRALBP, and the early RPE marker MITF. However, the expression of all these markers dropped from Day 14 and onwards. CONCLUSIONS Upon exposure of proliferating cells from the adult human PR to differentiating conditions in culture, there is a widespread change in morphology and gene expression, including the upregulation of key retinal markers. However, this upregulation is only transient and decreases after 14 days of differentiation.
Collapse
Affiliation(s)
- Erik O Johnsen
- a Center for Eye Research, Department of Ophthalmology , Oslo University Hospital and University of Oslo , Oslo , Norway.,b Norwegian Center for Stem Cell Research , Oslo University Hospital and University of Oslo , Oslo , Norway
| | - Rebecca C Frøen
- a Center for Eye Research, Department of Ophthalmology , Oslo University Hospital and University of Oslo , Oslo , Norway.,b Norwegian Center for Stem Cell Research , Oslo University Hospital and University of Oslo , Oslo , Norway
| | | | - Bjørn Nicolaissen
- a Center for Eye Research, Department of Ophthalmology , Oslo University Hospital and University of Oslo , Oslo , Norway.,b Norwegian Center for Stem Cell Research , Oslo University Hospital and University of Oslo , Oslo , Norway
| | - Goran Petrovski
- a Center for Eye Research, Department of Ophthalmology , Oslo University Hospital and University of Oslo , Oslo , Norway.,b Norwegian Center for Stem Cell Research , Oslo University Hospital and University of Oslo , Oslo , Norway.,d Department of Ophthalmology, Faculty of Medicine , University of Szeged and Stem Cells and Eye Research LaboratorySzeged, Hungary.,e Department of Biochemistry and Molecular Biology , University of Debrecen , Debrecen , Hungary
| | - Morten C Moe
- a Center for Eye Research, Department of Ophthalmology , Oslo University Hospital and University of Oslo , Oslo , Norway.,b Norwegian Center for Stem Cell Research , Oslo University Hospital and University of Oslo , Oslo , Norway
| | - Agate Noer
- a Center for Eye Research, Department of Ophthalmology , Oslo University Hospital and University of Oslo , Oslo , Norway.,b Norwegian Center for Stem Cell Research , Oslo University Hospital and University of Oslo , Oslo , Norway
| |
Collapse
|
18
|
Davoust C, Plas B, Béduer A, Demain B, Salabert AS, Sol JC, Vieu C, Vaysse L, Loubinoux I. Regenerative potential of primary adult human neural stem cells on micropatterned bio-implants boosts motor recovery. Stem Cell Res Ther 2017; 8:253. [PMID: 29116017 PMCID: PMC5688800 DOI: 10.1186/s13287-017-0702-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/25/2017] [Accepted: 10/18/2017] [Indexed: 01/19/2023] Open
Abstract
Background The adult brain is unable to regenerate itself sufficiently after large injuries. Therefore, hopes rely on therapies using neural stem cell or biomaterial transplantation to sustain brain reconstruction. The aim of the present study was to evaluate the improvement in sensorimotor recovery brought about by human primary adult neural stem cells (hNSCs) in combination with bio-implants. Methods hNSCs were pre-seeded on implants micropatterned for neurite guidance and inserted intracerebrally 2 weeks after a primary motor cortex lesion in rats. Long-term behaviour was significantly improved after hNSC implants versus cell engraftment in the grip strength test. MRI and immunohistological studies were conducted to elucidate the underlying mechanisms of neuro-implant integration. Results hNSC implants promoted tissue reconstruction and limited hemispheric atrophy and glial scar expansion. After 3 months, grafted hNSCs were detected on implants and expressed mature neuronal markers (NeuN, MAP2, SMI312). They also migrated over a short distance to the reconstructed tissues and to the peri-lesional tissues, where 26% integrated as mature neurons. Newly formed host neural progenitors (nestin, DCX) colonized the implants, notably in the presence of hNSCs, and participated in tissue reconstruction. The microstructured bio-implants sustained the guided maturation of both grafted hNSCs and endogenous progenitors. Conclusions These immunohistological results are coherent with and could explain the late improvement observed in sensorimotor recovery. These findings provide novel insights into the regenerative potential of primary adult hNSCs combined with microstructured implants.
Collapse
Affiliation(s)
- Carole Davoust
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Benjamin Plas
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France.,Centre Hospitalier Universitaire de Toulouse, Pôle Neurosciences, CHU Toulouse, Toulouse, France
| | - Amélie Béduer
- LAAS-CNRS, Université de Toulouse, CNRS, INSA, UPS, Toulouse, France
| | - Boris Demain
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Anne-Sophie Salabert
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France.,Centre Hospitalier Universitaire de Toulouse, Pôle Neurosciences, CHU Toulouse, Toulouse, France
| | - Jean Christophe Sol
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France.,Centre Hospitalier Universitaire de Toulouse, Pôle Neurosciences, CHU Toulouse, Toulouse, France
| | - Christophe Vieu
- LAAS-CNRS, Université de Toulouse, CNRS, INSA, UPS, Toulouse, France
| | - Laurence Vaysse
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France
| | - Isabelle Loubinoux
- ToNIC, Toulouse NeuroImaging Center, Université de Toulouse, Inserm, UPS, Toulouse, France. .,UMR1214-Inserm/UPS-ToNIC, CHU PURPAN, Pavillon Baudot, Place du Dr Baylac, 31024, Toulouse cedex 3, France.
| |
Collapse
|
19
|
Yang K, Oh JY, Lee JS, Jin Y, Chang GE, Chae SS, Cheong E, Baik HK, Cho SW. Photoactive Poly(3-hexylthiophene) Nanoweb for Optoelectrical Stimulation to Enhance Neurogenesis of Human Stem Cells. Theranostics 2017; 7:4591-4604. [PMID: 29158847 PMCID: PMC5695151 DOI: 10.7150/thno.20169] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 09/15/2017] [Indexed: 12/19/2022] Open
Abstract
Optoelectrical manipulation has recently gained attention for cellular engineering; however, few material platforms can be used to efficiently regulate stem cell behaviors via optoelectrical stimulation. In this study, we developed nanoweb substrates composed of photoactive polymer poly(3-hexylthiophene) (P3HT) to enhance the neurogenesis of human fetal neural stem cells (hfNSCs) through photo-induced electrical stimulation. METHODS The photoactive nanoweb substrates were fabricated by self-assembled one-dimensional (1D) P3HT nanostructures (nanofibrils and nanorods). The hfNSCs cultured on the P3HT nanoweb substrates were optically stimulated with a green light (539 nm) and then differentiation of hfNSCs on the substrates with light stimulation was examined. The utility of the nanoweb substrates for optogenetic application was tested with photo-responsive hfNSCs engineered by polymer nanoparticle-mediated transfection of an engineered chimeric opsin variant (C1V1)-encoding gene. RESULTS The nanoweb substrates provided not only topographical stimulation for activating focal adhesion signaling of hfNSCs, but also generated optoelectrical stimulation via photochemical and charge-transfer reactions upon exposure to 539 nm wavelength light, leading to significantly enhanced neuronal differentiation of hfNSCs. The optoelectrically stimulated hfNSCs exhibited mature neuronal phenotypes with highly extended neurite formation and functional neuron-like electrophysiological features of sodium currents and action potentials. Optoelectrical stimulation with 539 nm light simultaneously activated both C1V1-modified hfNSCs and nanoweb substrates, which upregulated the expression and activation of voltage-gated ion channels in hfNSCs and further increased the effect of photoactive substrates on neuronal differentiation of hfNSCs. CONCLUSION The photoactive nanoweb substrates developed in this study may serve as platforms for producing stem cell therapeutics with enhanced neurogenesis and neuromodulation via optoelectrical control of stem cells.
Collapse
|
20
|
Brain tissue banking for stem cells for our future. Sci Rep 2016; 6:39394. [PMID: 27991551 PMCID: PMC5171803 DOI: 10.1038/srep39394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/23/2016] [Indexed: 01/04/2023] Open
Abstract
In our lab we study neurogenesis and the development of brain tumors. We work towards treatment strategies for glioblastoma and towards using autologous neural stem cells for tissue regeneration strategies for brain damage and neurodegenerative disorders. It has been our policy to try to establish living cell cultures from all human biopsy material that we obtain. We hypothesized that small pieces of brain tissue could be cryopreserved and that live neural stem cells could be recovered at a later time. DMSO has been shown to possess a remarkable ability to diffuse through cell membranes and pass into cell interiors. Its chemical properties prevent the formation of damaging ice crystals thus allowing cell storage at or below −180 C. We report here a protocol for successful freezing of small pieces of tissue derived from human brain and human brain tumours. Virtually all specimens could be successfully revived. Assays of phenotype and behaviour show that the cell cultures derived were equivalent to those cultures previously derived from fresh tissue.
Collapse
|
21
|
Stangeland B, Mughal AA, Grieg Z, Sandberg CJ, Joel M, Nygård S, Meling T, Murrell W, Vik Mo EO, Langmoen IA. Combined expressional analysis, bioinformatics and targeted proteomics identify new potential therapeutic targets in glioblastoma stem cells. Oncotarget 2016; 6:26192-215. [PMID: 26295306 PMCID: PMC4694895 DOI: 10.18632/oncotarget.4613] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/10/2015] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is both the most common and the most lethal primary brain tumor. It is thought that GBM stem cells (GSCs) are critically important in resistance to therapy. Therefore, there is a strong rationale to target these cells in order to develop new molecular therapies. To identify molecular targets in GSCs, we compared gene expression in GSCs to that in neural stem cells (NSCs) from the adult human brain, using microarrays. Bioinformatic filtering identified 20 genes (PBK/TOPK, CENPA, KIF15, DEPDC1, CDC6, DLG7/DLGAP5/HURP, KIF18A, EZH2, HMMR/RHAMM/CD168, NOL4, MPP6, MDM1, RAPGEF4, RHBDD1, FNDC3B, FILIP1L, MCC, ATXN7L4/ATXN7L1, P2RY5/LPAR6 and FAM118A) that were consistently expressed in GSC cultures and consistently not expressed in NSC cultures. The expression of these genes was confirmed in clinical samples (TCGA and REMBRANDT). The first nine genes were highly co-expressed in all GBM subtypes and were part of the same protein-protein interaction network. Furthermore, their combined up-regulation correlated negatively with patient survival in the mesenchymal GBM subtype. Using targeted proteomics and the COGNOSCENTE database we linked these genes to GBM signalling pathways. Nine genes: PBK, CENPA, KIF15, DEPDC1, CDC6, DLG7, KIF18A, EZH2 and HMMR should be further explored as targets for treatment of GBM.
Collapse
Affiliation(s)
- Biljana Stangeland
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway.,SFI-CAST Biomedical Innovation Center, Oslo University Hospital, Oslo, Norway
| | - Awais A Mughal
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Zanina Grieg
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway.,Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Cecilie Jonsgar Sandberg
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Mrinal Joel
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway.,Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway.,Laboratory of Neural Development and Optical Recording (NDEVOR), Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Ståle Nygård
- Bioinformatics Core Facility, Institute for Medical Informatics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Torstein Meling
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Wayne Murrell
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Einar O Vik Mo
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Iver A Langmoen
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, Oslo, Norway.,SFI-CAST Biomedical Innovation Center, Oslo University Hospital, Oslo, Norway.,Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
22
|
Squecco R, Idrizaj E, Morelli A, Gallina P, Vannelli GB, Francini F. An electrophysiological study on the effects of BDNF and FGF2 on voltage dependent Ca(2+) currents in developing human striatal primordium. Mol Cell Neurosci 2016; 75:50-62. [PMID: 27370937 DOI: 10.1016/j.mcn.2016.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 05/24/2016] [Accepted: 06/27/2016] [Indexed: 01/06/2023] Open
Abstract
Over the past decades, studies in both Huntington's disease animal models and pilot clinical trials have demonstrated that replacement of degenerated striatum and repair of circuitries by grafting fetal striatal primordium is feasible, safe and may counteract disease progression. However, a better comprehension of striatal ontogenesis is required to assess the fetal graft regenerative potential. During neuronal development, neurotrophins exert pleiotropic actions in regulating cell fate and synaptic plasticity. In this regard, brain-derived neurotrophic factor (BDNF) and fibroblast growth factor 2 (FGF2) are crucially implicated in the control of fate choice of striatal progenitor cells. In this study, we intended to refine the functional features of human striatal precursor (HSP) cells isolated from ganglionic eminence of 9-12week old human fetuses, by studying with electrophysiological methods the effect of BDNF and FGF2 on the membrane biophysical properties and the voltage-dependent Ca(2+) currents. These features are particularly relevant to evaluate neuronal cell functioning and can be considered reliable markers of the developmental phenotype of human striatal primordium. Our results have demonstrated that BDNF and FGF2 induced membrane hyperpolarization, increased the membrane capacitance and reduced the resting total and specific conductance values, suggesting a more efficient control of resting ionic fluxes. Moreover, the treatment with both neurotrophins enhanced N-type Ca(2+) current amplitude and reduced L- and T-type ones. Overall, our data indicate that BDNF and FGF2 may help HSP cells to attain a more functionally mature phenotype.
Collapse
Affiliation(s)
- Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, viale Morgagni 63, 50134 Florence, Italy.
| | - Eglantina Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, viale Morgagni 63, 50134 Florence, Italy
| | - Annamaria Morelli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Pasquale Gallina
- Department of Surgery and Translational Medicine, University of Florence, Largo Palagi 1, 50139 Florence, Italy
| | - Gabriella B Vannelli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Fabio Francini
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, viale Morgagni 63, 50134 Florence, Italy
| |
Collapse
|
23
|
Wu CC, Lien CC, Hou WH, Chiang PM, Tsai KJ. Gain of BDNF Function in Engrafted Neural Stem Cells Promotes the Therapeutic Potential for Alzheimer's Disease. Sci Rep 2016; 6:27358. [PMID: 27264956 PMCID: PMC4893631 DOI: 10.1038/srep27358] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/17/2016] [Indexed: 12/16/2022] Open
Abstract
Stem cell-based therapy is a potential treatment for neurodegenerative diseases, but its application to Alzheimer’s disease (AD) remains limited. Brain-derived neurotrophic factor (BDNF) is critical in the pathogenesis and treatment of AD. Here, we present a novel therapeutic approach for AD treatment using BDNF-overexpressing neural stem cells (BDNF-NSCs). In vitro, BDNF overexpression was neuroprotective to beta-amyloid-treated NSCs. In vivo, engrafted BDNF-NSCs-derived neurons not only displayed the Ca2+-response fluctuations, exhibited electrophysiological properties of mature neurons and integrated into local brain circuits, but recovered the cognitive deficits. Furthermore, BDNF overexpression improved the engrafted cells’ viability, neuronal fate, neurite complexity, maturation of electrical property and the synaptic density. In contrast, knockdown of the BDNF in BDNF-NSCs diminished stem cell-based therapeutic efficacy. Together, our findings indicate BDNF overexpression improves the therapeutic potential of engrafted NSCs for AD via neurogenic effects and neuronal replacement, and further support the feasibility of NSC-based ex vivo gene therapy for AD.
Collapse
Affiliation(s)
- Cheng-Chun Wu
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Chang Lien
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
| | - Wen-Hsien Hou
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
| | - Po-Min Chiang
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuen-Jer Tsai
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
24
|
Burns AJ, Goldstein AM, Newgreen DF, Stamp L, Schäfer KH, Metzger M, Hotta R, Young HM, Andrews PW, Thapar N, Belkind-Gerson J, Bondurand N, Bornstein JC, Chan WY, Cheah K, Gershon MD, Heuckeroth RO, Hofstra RMW, Just L, Kapur RP, King SK, McCann CJ, Nagy N, Ngan E, Obermayr F, Pachnis V, Pasricha PJ, Sham MH, Tam P, Vanden Berghe P. White paper on guidelines concerning enteric nervous system stem cell therapy for enteric neuropathies. Dev Biol 2016; 417:229-51. [PMID: 27059883 DOI: 10.1016/j.ydbio.2016.04.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 03/29/2016] [Accepted: 04/02/2016] [Indexed: 12/22/2022]
Abstract
Over the last 20 years, there has been increasing focus on the development of novel stem cell based therapies for the treatment of disorders and diseases affecting the enteric nervous system (ENS) of the gastrointestinal tract (so-called enteric neuropathies). Here, the idea is that ENS progenitor/stem cells could be transplanted into the gut wall to replace the damaged or absent neurons and glia of the ENS. This White Paper sets out experts' views on the commonly used methods and approaches to identify, isolate, purify, expand and optimize ENS stem cells, transplant them into the bowel, and assess transplant success, including restoration of gut function. We also highlight obstacles that must be overcome in order to progress from successful preclinical studies in animal models to ENS stem cell therapies in the clinic.
Collapse
Affiliation(s)
- Alan J Burns
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Donald F Newgreen
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville 3052, Victoria, Australia
| | - Lincon Stamp
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Karl-Herbert Schäfer
- University of Applied Sciences, Kaiserlautern, Germany; Clinic of Pediatric Surgery, University Hospital Mannheim, University Heidelberg, Germany
| | - Marco Metzger
- Fraunhofer-Institute Interfacial Engineering and Biotechnology IGB Translational Centre - Würzburg branch and University Hospital Würzburg - Tissue Engineering and Regenerative Medicine (TERM), Würzburg, Germany
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Heather M Young
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Peter W Andrews
- Centre for Stem Cell Biology, Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Jaime Belkind-Gerson
- Division of Gastroenterology, Hepatology and Nutrition, Massachusetts General Hospital for Children, Harvard Medical School, Boston, USA
| | - Nadege Bondurand
- INSERM U955, 51 Avenue du Maréchal de Lattre de Tassigny, F-94000 Créteil, France; Université Paris-Est, UPEC, F-94000 Créteil, France
| | - Joel C Bornstein
- Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Wood Yee Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Kathryn Cheah
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong
| | - Michael D Gershon
- Department of Pathology and Cell Biology, Columbia University, New York 10032, USA
| | - Robert O Heuckeroth
- Department of Pediatrics, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA; Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, PA 19104, USA
| | - Robert M W Hofstra
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Lothar Just
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Germany
| | - Raj P Kapur
- Department of Pathology, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Sebastian K King
- Department of Paediatric and Neonatal Surgery, The Royal Children's Hospital, Melbourne, Australia
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Elly Ngan
- Department of Surgery, The University of Hong Kong, Hong Kong
| | - Florian Obermayr
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital Tübingen, D-72076 Tübingen, Germany
| | | | | | - Mai Har Sham
- Department of Biochemistry, The University of Hong Kong, Hong Kong
| | - Paul Tam
- Department of Surgery, The University of Hong Kong, Hong Kong
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), TARGID, University of Leuven, Belgium
| |
Collapse
|
25
|
Nam H, Lee KH, Nam DH, Joo KM. Adult human neural stem cell therapeutics: Current developmental status and prospect. World J Stem Cells 2015; 7:126-136. [PMID: 25621112 PMCID: PMC4300923 DOI: 10.4252/wjsc.v7.i1.126] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/01/2014] [Accepted: 10/16/2014] [Indexed: 02/06/2023] Open
Abstract
Over the past two decades, regenerative therapies using stem cell technologies have been developed for various neurological diseases. Although stem cell therapy is an attractive option to reverse neural tissue damage and to recover neurological deficits, it is still under development so as not to show significant treatment effects in clinical settings. In this review, we discuss the scientific and clinical basics of adult neural stem cells (aNSCs), and their current developmental status as cell therapeutics for neurological disease. Compared with other types of stem cells, aNSCs have clinical advantages, such as limited proliferation, inborn differentiation potential into functional neural cells, and no ethical issues. In spite of the merits of aNSCs, difficulties in the isolation from the normal brain, and in the in vitro expansion, have blocked preclinical and clinical study using aNSCs. However, several groups have recently developed novel techniques to isolate and expand aNSCs from normal adult brains, and showed successful applications of aNSCs to neurological diseases. With new technologies for aNSCs and their clinical strengths, previous hurdles in stem cell therapies for neurological diseases could be overcome, to realize clinically efficacious regenerative stem cell therapeutics.
Collapse
|
26
|
Store-operated CRAC channels regulate gene expression and proliferation in neural progenitor cells. J Neurosci 2014; 34:9107-23. [PMID: 24990931 DOI: 10.1523/jneurosci.0263-14.2014] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Calcium signals regulate many critical processes during vertebrate brain development including neurogenesis, neurotransmitter specification, and axonal outgrowth. However, the identity of the ion channels mediating Ca(2+) signaling in the developing nervous system is not well defined. Here, we report that embryonic and adult mouse neural stem/progenitor cells (NSCs/NPCs) exhibit store-operated Ca(2+) entry (SOCE) mediated by Ca(2+) release-activated Ca(2+) (CRAC) channels. SOCE in NPCs was blocked by the CRAC channel inhibitors La(3+), BTP2, and 2-APB and Western blots revealed the presence of the canonical CRAC channel proteins STIM1 and Orai1. Knock down of STIM1 or Orai1 significantly diminished SOCE in NPCs, and SOCE was lost in NPCs from transgenic mice lacking Orai1 or STIM1 and in knock-in mice expressing the loss-of-function Orai1 mutant, R93W. Therefore, STIM1 and Orai1 make essential contributions to SOCE in NPCs. SOCE in NPCs was activated by epidermal growth factor and acetylcholine, the latter occurring through muscarinic receptors. Activation of SOCE stimulated gene transcription through calcineurin/NFAT (nuclear factor of activated T cells) signaling through a mechanism consistent with local Ca(2+) signaling by Ca(2+) microdomains near CRAC channels. Importantly, suppression or deletion of STIM1 and Orai1 expression significantly attenuated proliferation of embryonic and adult NPCs cultured as neurospheres and, in vivo, in the subventricular zone of adult mice. These findings show that CRAC channels serve as a major route of Ca(2+) entry in NPCs and regulate key effector functions including gene expression and proliferation, indicating that CRAC channels are important regulators of mammalian neurogenesis.
Collapse
|
27
|
A time course analysis of the electrophysiological properties of neurons differentiated from human induced pluripotent stem cells (iPSCs). PLoS One 2014; 9:e103418. [PMID: 25072157 PMCID: PMC4114788 DOI: 10.1371/journal.pone.0103418] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 07/02/2014] [Indexed: 11/19/2022] Open
Abstract
Many protocols have been designed to differentiate human embryonic stem cells (ESCs) and human induced pluripotent stem cells (iPSCs) into neurons. Despite the relevance of electrophysiological properties for proper neuronal function, little is known about the evolution over time of important neuronal electrophysiological parameters in iPSC-derived neurons. Yet, understanding the development of basic electrophysiological characteristics of iPSC-derived neurons is critical for evaluating their usefulness in basic and translational research. Therefore, we analyzed the basic electrophysiological parameters of forebrain neurons differentiated from human iPSCs, from day 31 to day 55 after the initiation of neuronal differentiation. We assayed the developmental progression of various properties, including resting membrane potential, action potential, sodium and potassium channel currents, somatic calcium transients and synaptic activity. During the maturation of iPSC-derived neurons, the resting membrane potential became more negative, the expression of voltage-gated sodium channels increased, the membrane became capable of generating action potentials following adequate depolarization and, at day 48–55, 50% of the cells were capable of firing action potentials in response to a prolonged depolarizing current step, of which 30% produced multiple action potentials. The percentage of cells exhibiting miniature excitatory post-synaptic currents increased over time with a significant increase in their frequency and amplitude. These changes were associated with an increase of Ca2+ transient frequency. Co-culturing iPSC-derived neurons with mouse glial cells enhanced the development of electrophysiological parameters as compared to pure iPSC-derived neuronal cultures. This study demonstrates the importance of properly evaluating the electrophysiological status of the newly generated neurons when using stem cell technology, as electrophysiological properties of iPSC-derived neurons mature over time.
Collapse
|
28
|
Nogueira AB, Sogayar MC, Colquhoun A, Siqueira SA, Nogueira AB, Marchiori PE, Teixeira MJ. Existence of a potential neurogenic system in the adult human brain. J Transl Med 2014; 12:75. [PMID: 24655332 PMCID: PMC3998109 DOI: 10.1186/1479-5876-12-75] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 03/13/2014] [Indexed: 01/17/2023] Open
Abstract
Background Prevailingly, adult mammalian neurogenesis is thought to occur in discrete, separate locations known as neurogenic niches that are best characterized in the subgranular zone (SGZ) of the dentate gyrus and in the subventricular zone (SVZ). The existence of adult human neurogenic niches is controversial. Methods The existence of neurogenic niches was investigated with neurogenesis marker immunostaining in histologically normal human brains obtained from autopsies. Twenty-eight adult temporal lobes, specimens from limbic structures and the hypothalamus of one newborn and one adult were examined. Results The neural stem cell marker nestin stained circumventricular organ cells and the immature neuronal marker doublecortin (DCX) stained hypothalamic and limbic structures adjacent to circumventricular organs; both markers stained a continuous structure running from the hypothalamus to the hippocampus. The cell proliferation marker Ki-67 was detected predominately in structures that form the septo-hypothalamic continuum. Nestin-expressing cells were located in the fimbria-fornix at the insertion of the choroid plexus; ependymal cells in this structure expressed the putative neural stem cell marker CD133. From the choroidal fissure in the temporal lobe, a nestin-positive cell layer spread throughout the SVZ and subpial zone. In the subpial zone, a branch of this layer reached the hippocampal sulcus and ended in the SGZ (principally in the newborn) and in the subiculum (principally in the adults). Another branch of the nestin-positive cell layer in the subpial zone returned to the optic chiasm. DCX staining was detected in the periventricular and middle hypothalamus and more densely from the mammillary body to the subiculum through the fimbria-fornix, thus running through the principal neuronal pathway from the hippocampus to the hypothalamus. The column of the fornix forms part of this pathway and appears to coincide with the zone previously identified as the human rostral migratory stream. Partial co-labeling with DCX and the neuronal marker βIII-tubulin was also observed. Conclusions Collectively, these findings suggest the existence of an adult human neurogenic system that rises from the circumventricular organs and follows, at minimum, the circuitry of the hypothalamus and limbic system.
Collapse
Affiliation(s)
- Adriano Barreto Nogueira
- Division of Neurosurgery Clinic, Hospital das Clínicas, Faculty of Medicine, University of São Paulo, Avenida Dr, Eneas de Carvalho Aguiar 255, 05403-900 São Paulo, Brazil.
| | | | | | | | | | | | | |
Collapse
|
29
|
Does the adult human ciliary body epithelium contain "true" retinal stem cells? BIOMED RESEARCH INTERNATIONAL 2013; 2013:531579. [PMID: 24286080 PMCID: PMC3826557 DOI: 10.1155/2013/531579] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 08/26/2013] [Accepted: 08/31/2013] [Indexed: 11/17/2022]
Abstract
Recent reports of retinal stem cells being present in several locations of the adult eye have sparked great hopes that they may be used to treat the millions of people worldwide who suffer from blindness as a result of retinal disease or injury. A population of proliferative cells derived from the ciliary body epithelium (CE) has been considered one of the prime stem cell candidates, and as such they have received much attention in recent years. However, the true nature of these cells in the adult human eye has still not been fully elucidated, and the stem cell claim has become increasingly controversial in light of new and conflicting reports. In this paper, we will try to answer the question of whether the available evidence is strong enough for the research community to conclude that the adult human CE indeed harbors stem cells.
Collapse
|
30
|
Expansion of multipotent stem cells from the adult human brain. PLoS One 2013; 8:e71334. [PMID: 23967194 PMCID: PMC3743777 DOI: 10.1371/journal.pone.0071334] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 06/30/2013] [Indexed: 12/22/2022] Open
Abstract
The discovery of stem cells in the adult human brain has revealed new possible scenarios for treatment of the sick or injured brain. Both clinical use of and preclinical research on human adult neural stem cells have, however, been seriously hampered by the fact that it has been impossible to passage these cells more than a very few times and with little expansion of cell numbers. Having explored a number of alternative culturing conditions we here present an efficient method for the establishment and propagation of human brain stem cells from whatever brain tissue samples we have tried. We describe virtually unlimited expansion of an authentic stem cell phenotype. Pluripotency proteins Sox2 and Oct4 are expressed without artificial induction. For the first time multipotency of adult human brain-derived stem cells is demonstrated beyond tissue boundaries. We characterize these cells in detail in vitro including microarray and proteomic approaches. Whilst clarification of these cells' behavior is ongoing, results so far portend well for the future repair of tissues by transplantation of an adult patient's own-derived stem cells.
Collapse
|
31
|
Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tønnesen P, Suso EMI, Sæbøe-Larssen S, Sandberg C, Brinchmann JE, Helseth E, Rasmussen AM, Lote K, Aamdal S, Gaudernack G, Kvalheim G, Langmoen IA. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother 2013; 62:1499-509. [PMID: 23817721 PMCID: PMC3755221 DOI: 10.1007/s00262-013-1453-3] [Citation(s) in RCA: 234] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 06/17/2013] [Indexed: 01/31/2023]
Abstract
Background The growth and recurrence of several cancers appear to be driven by a population of cancer stem cells (CSCs). Glioblastoma, the most common primary brain tumor, is invariably fatal, with a median survival of approximately 1 year. Although experimental data have suggested the importance of CSCs, few data exist regarding the potential relevance and importance of these cells in a clinical setting. Methods We here present the first seven patients treated with a dendritic cell (DC)-based vaccine targeting CSCs in a solid tumor. Brain tumor biopsies were dissociated into single-cell suspensions, and autologous CSCs were expanded in vitro as tumorspheres. From these, CSC-mRNA was amplified and transfected into monocyte-derived autologous DCs. The DCs were aliquoted to 9–18 vaccines containing 107 cells each. These vaccines were injected intradermally at specified intervals after the patients had received a standard 6-week course of post-operative radio-chemotherapy. The study was registered with the ClinicalTrials.gov identifier NCT00846456. Results Autologous CSC cultures were established from ten out of eleven tumors. High-quality RNA was isolated, and mRNA was amplified in all cases. Seven patients were able to be weaned from corticosteroids to receive DC immunotherapy. An immune response induced by vaccination was identified in all seven patients. No patients developed adverse autoimmune events or other side effects. Compared to matched controls, progression-free survival was 2.9 times longer in vaccinated patients (median 694 vs. 236 days, p = 0.0018, log-rank test). Conclusion These findings suggest that vaccination against glioblastoma stem cells is safe, well-tolerated, and may prolong progression-free survival. Electronic supplementary material The online version of this article (doi:10.1007/s00262-013-1453-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Einar Osland Vik-Mo
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research, University of Oslo, Oslo, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Sandberg CJ, Altschuler G, Jeong J, Strømme KK, Stangeland B, Murrell W, Grasmo-Wendler UH, Myklebost O, Helseth E, Vik-Mo EO, Hide W, Langmoen IA. Comparison of glioma stem cells to neural stem cells from the adult human brain identifies dysregulated Wnt- signaling and a fingerprint associated with clinical outcome. Exp Cell Res 2013; 319:2230-43. [PMID: 23791939 DOI: 10.1016/j.yexcr.2013.06.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 04/19/2013] [Accepted: 06/07/2013] [Indexed: 12/12/2022]
Abstract
Glioblastoma is the most common brain tumor. Median survival in unselected patients is <10 months. The tumor harbors stem-like cells that self-renew and propagate upon serial transplantation in mice, although the clinical relevance of these cells has not been well documented. We have performed the first genome-wide analysis that directly relates the gene expression profile of nine enriched populations of glioblastoma stem cells (GSCs) to five identically isolated and cultivated populations of stem cells from the normal adult human brain. Although the two cell types share common stem- and lineage-related markers, GSCs show a more heterogeneous gene expression. We identified a number of pathways that are dysregulated in GSCs. A subset of these pathways has previously been identified in leukemic stem cells, suggesting that cancer stem cells of different origin may have common features. Genes upregulated in GSCs were also highly expressed in embryonic and induced pluripotent stem cells. We found that canonical Wnt-signaling plays an important role in GSCs, but not in adult human neural stem cells. As well we identified a 30-gene signature highly overexpressed in GSCs. The expression of these signature genes correlates with clinical outcome and demonstrates the clinical relevance of GSCs.
Collapse
|
33
|
Reinhardt P, Glatza M, Hemmer K, Tsytsyura Y, Thiel CS, Höing S, Moritz S, Parga JA, Wagner L, Bruder JM, Wu G, Schmid B, Röpke A, Klingauf J, Schwamborn JC, Gasser T, Schöler HR, Sterneckert J. Derivation and expansion using only small molecules of human neural progenitors for neurodegenerative disease modeling. PLoS One 2013; 8:e59252. [PMID: 23533608 PMCID: PMC3606479 DOI: 10.1371/journal.pone.0059252] [Citation(s) in RCA: 286] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 02/12/2013] [Indexed: 11/18/2022] Open
Abstract
Phenotypic drug discovery requires billions of cells for high-throughput screening (HTS) campaigns. Because up to several million different small molecules will be tested in a single HTS campaign, even small variability within the cell populations for screening could easily invalidate an entire campaign. Neurodegenerative assays are particularly challenging because neurons are post-mitotic and cannot be expanded for implementation in HTS. Therefore, HTS for neuroprotective compounds requires a cell type that is robustly expandable and able to differentiate into all of the neuronal subtypes involved in disease pathogenesis. Here, we report the derivation and propagation using only small molecules of human neural progenitor cells (small molecule neural precursor cells; smNPCs). smNPCs are robust, exhibit immortal expansion, and do not require cumbersome manual culture and selection steps. We demonstrate that smNPCs have the potential to clonally and efficiently differentiate into neural tube lineages, including motor neurons (MNs) and midbrain dopaminergic neurons (mDANs) as well as neural crest lineages, including peripheral neurons and mesenchymal cells. These properties are so far only matched by pluripotent stem cells. Finally, to demonstrate the usefulness of smNPCs we show that mDANs differentiated from smNPCs with LRRK2 G2019S are more susceptible to apoptosis in the presence of oxidative stress compared to wild-type. Therefore, smNPCs are a powerful biological tool with properties that are optimal for large-scale disease modeling, phenotypic screening, and studies of early human development.
Collapse
Affiliation(s)
- Peter Reinhardt
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, North Rhine Westphalia, Germany
| | - Michael Glatza
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, North Rhine Westphalia, Germany
| | - Kathrin Hemmer
- Stem Cell Biology and Regeneration Group, Institute of Cell Biology, Center for Molecular Biology of Inflammation, Westfälische Wilhelms-Universität Münster, Münster, North Rhine-Westphalia, Germany
| | - Yaroslav Tsytsyura
- Westfälische Wilhelms-Universität Münster, Institute for Medical Physics and Biophysics, Cellular Biophysics Group, Münster, North Rhine-Westphalia, Germany
| | - Cora S. Thiel
- Westfälische Wilhelms-Universität Münster, Institute for Medical Physics and Biophysics, Cellular Biophysics Group, Münster, North Rhine-Westphalia, Germany
| | - Susanne Höing
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, North Rhine Westphalia, Germany
| | - Sören Moritz
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, North Rhine Westphalia, Germany
| | - Juan A. Parga
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, North Rhine Westphalia, Germany
- Center for Research in Molecular Medicine and Chronic Diseases at the University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Lydia Wagner
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, North Rhine Westphalia, Germany
| | - Jan M. Bruder
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, North Rhine Westphalia, Germany
| | - Guangming Wu
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, North Rhine Westphalia, Germany
| | - Benjamin Schmid
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, and German Center for Neurodegenerative Diseases, Tübingen, Baden-Württemburg, Germany
| | - Albrecht Röpke
- Institute for Human Genetics, University of Münster, Münster, North Rhine Westphalia, Germany
| | - Jürgen Klingauf
- Westfälische Wilhelms-Universität Münster, Institute for Medical Physics and Biophysics, Cellular Biophysics Group, Münster, North Rhine-Westphalia, Germany
| | - Jens C. Schwamborn
- Stem Cell Biology and Regeneration Group, Institute of Cell Biology, Center for Molecular Biology of Inflammation, Westfälische Wilhelms-Universität Münster, Münster, North Rhine-Westphalia, Germany
| | - Thomas Gasser
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, and German Center for Neurodegenerative Diseases, Tübingen, Baden-Württemburg, Germany
| | - Hans R. Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, North Rhine Westphalia, Germany
- Medical Faculty, University of Münster, Münster, North Rhine-Westphalia, Germany
- * E-mail: (HRS); (JS)
| | - Jared Sterneckert
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, North Rhine Westphalia, Germany
- * E-mail: (HRS); (JS)
| |
Collapse
|
34
|
Neurogenesis in temporal lobe epilepsy: Relationship between histological findings and changes in dentate gyrus proliferative properties. Clin Neurol Neurosurg 2013; 115:187-91. [DOI: 10.1016/j.clineuro.2012.05.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2011] [Revised: 04/02/2012] [Accepted: 05/12/2012] [Indexed: 11/24/2022]
|
35
|
Abstract
The discovery, in 1998, that the adult human brain contains at least two populations of progenitor cells and that progenitor cells are upregulated in response to a range of degenerative brain diseases has raised hopes for their use in replacing dying brain cells. Since these early findings the race has been on to understand the biology of progenitor cells in the human brain and they have now been isolated and studied in many major neurodegenerative diseases. Before these cells can be exploited for cell replacement purposes it is important to understand how to: (1) find them, (2) label them, (3) determine what receptors they express, (4) isolate them, and (5) examine their electrophysiological properties when differentiated. In this chapter we have described the methods we use for studying progenitor cells in the adult human brain and in particular the tissue processing, immunohistochemistry, autoradiography, progenitor cell culture, and electrophysiology on brain cells. The Neurological Foundation of New Zealand Human Brain Bank has been receiving human tissue for approximately 20 years during which time we have developed a number of unique ways to examine and isolate progenitor cells from resected surgical specimens as well as from postmortem brain tissue. There are ethical and technical considerations that are unique to working with human brain tissue and these, as well as the processing of this tissue and the culturing of it for the purpose of studying progenitor cells, are the topic of this chapter.
Collapse
|
36
|
Hao MM, Bornstein JC, Vanden Berghe P, Lomax AE, Young HM, Foong JPP. The emergence of neural activity and its role in the development of the enteric nervous system. Dev Biol 2012; 382:365-74. [PMID: 23261929 DOI: 10.1016/j.ydbio.2012.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 12/03/2012] [Accepted: 12/10/2012] [Indexed: 12/31/2022]
Abstract
The enteric nervous system (ENS) is a vital part of the autonomic nervous system that regulates many gastrointestinal functions, including motility and secretion. All neurons and glia of the ENS arise from neural crest-derived cells that migrate into the gastrointestinal tract during embryonic development. It has been known for many years that a subpopulation of the enteric neural crest-derived cells expresses pan-neuronal markers at early stages of ENS development. Recent studies have demonstrated that some enteric neurons exhibit electrical activity from as early as E11.5 in the mouse, with further maturation of activity during embryonic and postnatal development. This article discusses the maturation of electrophysiological and morphological properties of enteric neurons, the formation of synapses and synaptic activity, and the influence of neural activity on ENS development.
Collapse
Affiliation(s)
- Marlene M Hao
- Department of Anatomy and Neuroscience, the University of Melbourne, Victoria 3010, Australia
| | | | | | | | | | | |
Collapse
|
37
|
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
|
38
|
Abstract
Neural activity is integral to the development of the enteric nervous system (ENS). A subpopulation of neural crest-derived cells expresses pan-neuronal markers at early stages of ENS development (at E10.5 in the mouse). However, the electrical activity of these cells has not been previously characterized, and it is not known whether all cells expressing neuronal markers are capable of firing action potentials (APs). In this study, we examined the activity of "neuron"-like cells (expressing pan-neuronal markers or with neuronal morphology) in the gut of E11.5 and E12.5 mice using whole-cell patch-clamp electrophysiology and compared them to the activity of neonatal and adult enteric neurons. Around 30-40% of neuron-like cells at E11.5 and E12.5 fired APs, some of which were very similar to those of adult enteric neurons. All APs were sensitive to tetrodotoxin (TTX), indicating that they were driven by voltage-gated Na+ currents. Expression of mRNA encoding several voltage-gated Na+ channels by the E11.5 gut was detected using RT-PCR. The density of voltage-gated Na+ currents increased from E11.5 to neonates. Immature active responses, mediated in part by TTX- and lidocaine-insensitive channels, were observed in most cells at E11.5 and E12.5, but not in P0/P1 or adult neurons. However, some cells expressing neuronal markers at E11.5 or E12.5 did not exhibit an active response to depolarization. Spontaneous depolarizations resembling excitatory postsynaptic potentials were observed at E12.5. The ENS is one of the earliest parts of the developing nervous system to exhibit mature forms of electrical activity.
Collapse
|
39
|
Electrophysiological properties and synaptic function of mesenchymal stem cells during neurogenic differentiation - a mini-review. Int J Artif Organs 2012; 35:323-37. [PMID: 22505200 DOI: 10.5301/ijao.5000085] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2011] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) have gained considerable interest due to their potential use in cell therapies and tissue engineering. They have been reported to differentiate into various anchorage-dependent cell types, including bone, cartilage, and tendon. Our focus is on the differentiation of MSCs into neuron-like cells through the use of soluble chemical stimuli or specific growth factor supplements. The resulting cells appear to adopt neural phenotypes and express some typical neuronal markers, however, their electrophysiological properties and synaptic function remains unclear. RESULTS This mini-review illustrates how particular characteristics, electrophysiological properties, and synaptic functions of MSCs change during their neuronal differentiation. In particular we focus on changes in ion currents, ion channels, synaptic communication, and neurotransmitter release. We also highlight conflicting results, caused by inconsistencies in the experimental conditions used and in the methodologies adopted. CONCLUSIONS We conclude that there is insufficient data and that further, carefully controlled investigations are required in order to ascertain whether MSC-derived neuron-like cells can exhibit the necessary neuronal functions to become clinically relevant for use in neural repairs.
Collapse
|
40
|
Ma K, Fox L, Shi G, Shen J, Liu Q, Pappas JD, Cheng J, Qu T. Generation of neural stem cell-like cells from bone marrow-derived human mesenchymal stem cells. Neurol Res 2012; 33:1083-93. [PMID: 22196762 DOI: 10.1179/1743132811y.0000000053] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Under appropriate culture conditions, bone marrow (BM)-derived mesenchymal stem cells are capable of differentiating into diverse cell types unrelated to their phenotypical embryonic origin, including neural cells. Here, we report the successful generation of neural stem cell (NSC)-like cells from BM-derived human mesenchymal stem cells (hMSCs). Initially, hMSCs were cultivated in a conditioned medium of human neural stem cells. In this culture system, hMSCs were induced to become NSC-like cells, which proliferate in neurosphere-like structures and express early NSC markers. Like central nervous system-derived NSCs, these BM-derived NSC-like cells were able to differentiate into cells expressing neural markers for neurons, astrocytes, and oligodendrocytes. Whole-cell patch clamp recording revealed that neuron-like cells, differentiated from NSC-like cells, exhibited electrophysiological properties of neurons, including action potentials. Transplantation of NSC-like cells into mouse brain confirmed that these NSC-like cells retained their capability to differentiate into neuronal and glial cells in vivo. Our data show that multipotent NSC-like cells can be efficiently produced from BM-derived hMSCs in culture and that these cells may serve as a useful alternative to human neural stem cells for potential clinical applications such as autologous neuroreplacement therapies.
Collapse
Affiliation(s)
- K Ma
- College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Vaysse L, Labie C, Canolle B, Jozan S, Béduer A, Arnauduc F, Vieu C, Sol JC, Loubinoux I. Adult human progenitor cells from the temporal lobe: another source of neuronal cells. Brain Inj 2012; 26:1636-45. [PMID: 22823462 DOI: 10.3109/02699052.2012.700084] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES In the adult human brain, neurogenesis occurs in the SVZ and the dentate gyrus of the hippocampus, but it is still unclear whether persistent neural progenitor/stem cells are also present in other brain areas. The present work studies the possibility of obtaining neural progenitor/stem cells from the temporal lobe and investigates their potential to differentiate into neuronal cells. METHODS Human biopsies from the temporal lobe of epileptic patients were used to isolate potential neural progenitors. Differentiation was induced in the presence of different agents (NGF, NT3, RA) and immunocytochemistry was then performed for quantitative analysis. RESULTS It was shown that a significant number of cells in the temporal lobe are also capable of expansion and multi-potency. These cells can be amplified as neurospheres and have the potential to differentiate naturally in vitro into neurons, astrocytes and oligodendrocytes. Quantitative analyses show that the progenitor cells of the temporal lobe exhibit a better rate of neuronal differentiation in vitro than the cells from the SVZ, particularly in the presence of NGF. CONCLUSION This study indicates that neural progenitors are also present in the human temporal lobe. Studying them could be of great interest for cell therapy in neurological disorders.
Collapse
Affiliation(s)
- L Vaysse
- Inserm, Imagerie Cérébrale et Handicaps Neurologiques UMR 825, Toulouse, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Park TIH, Monzo H, Mee EW, Bergin PS, Teoh HH, Montgomery JM, Faull RLM, Curtis MA, Dragunow M. Adult human brain neural progenitor cells (NPCs) and fibroblast-like cells have similar properties in vitro but only NPCs differentiate into neurons. PLoS One 2012; 7:e37742. [PMID: 22675489 PMCID: PMC3366988 DOI: 10.1371/journal.pone.0037742] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 04/26/2012] [Indexed: 01/19/2023] Open
Abstract
The ability to culture neural progenitor cells from the adult human brain has provided an exciting opportunity to develop and test potential therapies on adult human brain cells. To achieve a reliable and reproducible adult human neural progenitor cell (AhNPC) culture system for this purpose, this study fully characterized the cellular composition of the AhNPC cultures, as well as the possible changes to this in vitro system over prolonged culture periods. We isolated cells from the neurogenic subventricular zone/hippocampus (SVZ/HP) of the adult human brain and found a heterogeneous culture population comprised of several types of post-mitotic brain cells (neurons, astrocytes, and microglia), and more importantly, two distinct mitotic cell populations; the AhNPCs, and the fibroblast-like cells (FbCs). These two populations can easily be mistaken for a single population of AhNPCs, as they both proliferate under AhNPC culture conditions, form spheres and express neural progenitor cell and early neuronal markers, all of which are characteristics of AhNPCs in vitro. However, despite these similarities under proliferating conditions, under neuronal differentiation conditions, only the AhNPCs differentiated into functional neurons and glia. Furthermore, AhNPCs showed limited proliferative capacity that resulted in their depletion from culture by 5–6 passages, while the FbCs, which appear to be from a neurovascular origin, displayed a greater proliferative capacity and dominated the long-term cultures. This gradual change in cellular composition resulted in a progressive decline in neurogenic potential without the apparent loss of self-renewal in our cultures. These results demonstrate that while AhNPCs and FbCs behave similarly under proliferative conditions, they are two different cell populations. This information is vital for the interpretation and reproducibility of AhNPC experiments and suggests an ideal time frame for conducting AhNPC-based experiments.
Collapse
Affiliation(s)
- Thomas In-Hyeup Park
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- The Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Department of Anatomy and Radiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Hector Monzo
- The Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Department of Anatomy and Radiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Edward W. Mee
- Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Peter S. Bergin
- Department of Neurology, Auckland City Hospital, Auckland, New Zealand
| | | | - Johanna M. Montgomery
- The Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Richard L. M. Faull
- The Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Department of Anatomy and Radiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Maurice A. Curtis
- The Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- Department of Anatomy and Radiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- The Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
- * E-mail:
| |
Collapse
|
43
|
Friedlander M. Advances in treatment and management: immunologic and cell-based regenerative therapies. Invest Ophthalmol Vis Sci 2012; 53:2511-4. [PMID: 22562853 DOI: 10.1167/iovs.12-9483p] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Martin Friedlander
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037, USA.
| |
Collapse
|
44
|
Johnsen EO, Frøen RC, Albert R, Omdal BK, Sarang Z, Berta A, Nicolaissen B, Petrovski G, Moe MC. Activation of neural progenitor cells in human eyes with proliferative vitreoretinopathy. Exp Eye Res 2012; 98:28-36. [DOI: 10.1016/j.exer.2012.03.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 03/03/2012] [Accepted: 03/14/2012] [Indexed: 01/19/2023]
|
45
|
Foong JPP, Nguyen TV, Furness JB, Bornstein JC, Young HM. Myenteric neurons of the mouse small intestine undergo significant electrophysiological and morphological changes during postnatal development. J Physiol 2012; 590:2375-90. [PMID: 22371477 DOI: 10.1113/jphysiol.2011.225938] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Organized motility patterns in the gut depend on circuitry within the enteric nervous system (ENS), but little is known about the development of electrophysiological properties and synapses within the ENS. We examined the electrophysiology and morphology of myenteric neurons in the mouse duodenum at three developmental stages: postnatal day (P)0, P10–11, and adult. Like adults, two main classes of neurons could be identified at P0 and P10–11 based on morphology: neurons with multiple long processes that projected circumferentially (Dogiel type II morphology) and neurons with a single long process. However, postnatal Dogiel type II neurons differed in several electrophysiological properties from adult Dogiel type II neurons. P0 and P10–11 Dogiel type II neurons exhibited very prominent Ca(2+)-mediated after depolarizing potentials (ADPs) following action potentials compared to adult neurons. Adult Dogiel type II neurons are characterized by the presence of a prolonged after hyperpolarizing potential (AHP), but AHPs were very rarely observed at P0. The projection lengths of the long processes of Dogiel type II neurons were mature by P10–11. Uniaxonal neurons in adults typically have fast excitatory postsynaptic potentials (fEPSPs, ‘S-type' electrophysiology) mainly mediated by nicotinic receptors. Nicotinic-fEPSPs were also recorded from neurons with a single long process at P0 and P10–11. However, these neurons underwent major developmental changes in morphology, from predominantly filamentous neurites at birth to lamellar dendrites in mature mice. Unlike Dogiel type II neurons, the projection lengths of neurons with a single long process matured after P10–11. Slow EPSPs were rarely observed in P0/P10–11 neurons. This work shows that, although functional synapses are present and two classes of neurons can be distinguished electrophysiologically and morphologically at P0, major changes in electrophysiological properties and morphology occur during the postnatal development of the ENS.
Collapse
Affiliation(s)
- Jaime Pei Pei Foong
- Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia.
| | | | | | | | | |
Collapse
|
46
|
Monzo HJ, Park TI, Montgomery JM, Faull RL, Dragunow M, Curtis MA. A method for generating high-yield enriched neuronal cultures from P19 embryonal carcinoma cells. J Neurosci Methods 2012; 204:87-103. [DOI: 10.1016/j.jneumeth.2011.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 11/04/2011] [Accepted: 11/04/2011] [Indexed: 10/15/2022]
|
47
|
Roper SN, Steindler DA. Stem cells as a potential therapy for epilepsy. Exp Neurol 2012; 244:59-66. [PMID: 22265818 DOI: 10.1016/j.expneurol.2012.01.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 12/29/2011] [Accepted: 01/04/2012] [Indexed: 12/16/2022]
Abstract
Neural stem cells and neural progenitors (NSC/NPs) hold great promise in neuro-restorative therapy due to their remarkable capacity for self-renewal, plasticity, and ability to integrate into host brain circuitry. Some types of epilepsy would appear to be excellent targets for this type of therapy due to known alterations in local circuitry based on loss or malfunction of specific types of neurons in specific brain structures. Potential sources for NSC/NPs include the embryonic blastocyst, the fetal brain, and adult brain and non-neural tissues. Each of these cell types has potential strengths and weaknesses as candidates for clinical therapeutic agents. This article reviews some of the major types of NSC/NPs and how they have been studied with regard to synaptic integration into host brain circuits. It also reviews how these transplanted cells develop and interact with host brain cells in animal models of epilepsy. The field is still wide open with a number of very promising results but there are also some major challenges that will need to be addressed prior to considering clinical applications for epilepsy.
Collapse
Affiliation(s)
- Steven N Roper
- Department of Neurosurgery and the McKnight Brain Institute, University of Florida, USA.
| | | |
Collapse
|
48
|
Frøen RC, Johnsen EO, Petrovski G, Berényi E, Facskó A, Berta A, Nicolaissen B, Moe MC. Pigment epithelial cells isolated from human peripheral iridectomies have limited properties of retinal stem cells. Acta Ophthalmol 2011; 89:e635-44. [PMID: 21801333 DOI: 10.1111/j.1755-3768.2011.02198.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE The identification of cells with properties of retinal progenitor cells (RPCs) in the adult human ciliary margin (CM) prompted a number of studies of their proliferative and differentiation potential. One of the remaining challenges is to find a feasible method of isolating RPCs from the patient's eye. In the human CM, only the iris pigment epithelium (IPE) is easily obtained by a minimally invasive procedure. In the light of recent studies questioning the existence of RPCs in the adult mammalian CM, we wanted to assess the potential of the adult human IPE as source of RPCs. METHODS The IPE were isolated from peripheral iridectomies during glaucoma surgery, and IPE and ciliary body (CB) epithelium were also isolated from post-mortem tissue. Cells were cultivated in sphere-promoting conditions or as monolayers. Whole-tissue samples, undifferentiated and differentiated cells were studied by immunocytochemistry, RT-PCR and transmission electron microscopy. RESULTS The adult human IPE, like the CB, expressed markers of RPCs such as Pax6, Sox2 and Nestin in vivo. Both sphere-promoting and monolayer cultures preserved this phenotype. However, both IPE/CB cultures expressed markers of differentiated epithelial cells such as Claudin, microphtalmia-associated transcription factor (MITF) and Cytokeratin-19. Ultrastructurally, IPE spheres displayed epithelial-like junctions and contained mature melanosomes. After induced differentiation, IPE-derived cells showed only partial neuronal differentiation expressing β-III-tubulin, Map-2 and Rhodopsin, whereas no mature glial markers were found. CONCLUSION Proliferative cells with some properties of RPCs can be isolated from the adult human IPE by peripheral iridectomies. Yet, many cells retain properties of differentiated epithelial cells and lack central properties of somatic stem cells.
Collapse
Affiliation(s)
- Rebecca C Frøen
- Department of Ophthalmology, Oslo University Hospital, Center for Eye Research, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Distribution and characterization of progenitor cells within the human filum terminale. PLoS One 2011; 6:e27393. [PMID: 22096566 PMCID: PMC3214055 DOI: 10.1371/journal.pone.0027393] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 10/16/2011] [Indexed: 01/19/2023] Open
Abstract
Background Filum terminale (FT) is a structure that is intimately associated with conus medullaris, the most caudal part of the spinal cord. It is well documented that certain regions of the adult human central nervous system contains undifferentiated, progenitor cells or multipotent precursors. The primary objective of this study was to describe the distribution and progenitor features of this cell population in humans, and to confirm their ability to differentiate within the neuroectodermal lineage. Methodology/Principal Findings We demonstrate that neural stem/progenitor cells are present in FT obtained from patients treated for tethered cord. When human or rat FT-derived cells were cultured in defined medium, they proliferated and formed neurospheres in 13 out of 21 individuals. Cells expressing Sox2 and Musashi-1 were found to outline the central canal, and also to be distributed in islets throughout the whole FT. Following plating, the cells developed antigen profiles characteristic of astrocytes (GFAP) and neurons (β-III-tubulin). Addition of PDGF-BB directed the cells towards a neuronal fate. Moreover, the cells obtained from young donors shows higher capacity for proliferation and are easier to expand than cells derived from older donors. Conclusion/Significance The identification of bona fide neural progenitor cells in FT suggests a possible role for progenitor cells in this extension of conus medullaris and may provide an additional source of such cells for possible therapeutic purposes. Filum terminale, human, progenitor cells, neuron, astrocytes, spinal cord.
Collapse
|
50
|
Using an adherent cell culture of the mouse subependymal zone to study the behavior of adult neural stem cells on a single-cell level. Nat Protoc 2011; 6:1847-59. [DOI: 10.1038/nprot.2011.404] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|