1
|
Ya J, Pellumbaj J, Hashmat A, Bayraktutan U. The Role of Stem Cells as Therapeutics for Ischaemic Stroke. Cells 2024; 13:112. [PMID: 38247804 PMCID: PMC10814781 DOI: 10.3390/cells13020112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Stroke remains one of the leading causes of death and disability worldwide. Current reperfusion treatments for ischaemic stroke are limited due to their narrow therapeutic window in rescuing ischaemic penumbra. Stem cell therapy offers a promising alternative. As a regenerative medicine, stem cells offer a wider range of treatment strategies, including long-term intervention for chronic patients, through the reparation and replacement of injured cells via mechanisms of differentiation and proliferation. The purpose of this review is to evaluate the therapeutic role of stem cells for ischaemic stroke. This paper discusses the pathology during acute, subacute, and chronic phases of cerebral ischaemic injury, highlights the mechanisms involved in mesenchymal, endothelial, haematopoietic, and neural stem cell-mediated cerebrovascular regeneration, and evaluates the pre-clinical and clinical data concerning the safety and efficacy of stem cell-based treatments. The treatment of stroke patients with different types of stem cells appears to be safe and efficacious even at relatively higher concentrations irrespective of the route and timing of administration. The priming or pre-conditioning of cells prior to administration appears to help augment their therapeutic impact. However, larger patient cohorts and later-phase trials are required to consolidate these findings.
Collapse
Affiliation(s)
| | | | | | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neurosciences, Queens Medical Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
2
|
Damle EB, Morrison VE, Cioma J, Volic M, Bix GJ. Co-administration of extracellular matrix-based biomaterials with neural stem cell transplantation for treatment of central nervous system injury. Front Neurosci 2023; 17:1177040. [PMID: 37255752 PMCID: PMC10225608 DOI: 10.3389/fnins.2023.1177040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/28/2023] [Indexed: 06/01/2023] Open
Abstract
Injuries and disorders of the central nervous system (CNS) present a particularly difficult challenge for modern medicine to address, given the complex nature of the tissues, obstacles in researching and implementing therapies, and barriers to translating efficacious treatments into human patients. Recent advancements in neural stem cell (NSC) transplantation, endogenous neurogenesis, and in vivo reprogramming of non-neural cells into the neuronal lineage represent multiple approaches to resolving CNS injury. However, we propose that one practice that must be incorporated universally in neuroregeneration studies is the use of extracellular matrix (ECM)-mimicking biomaterials to supply the architectural support and cellular microenvironment necessary for partial or complete restoration of function. Through consideration of developmental processes including neurogenesis, cellular migration, and establishment of functional connectivity, as well as evaluation of process-specific interactions between cells and ECM components, insights can be gained to harness and modulate native and induced neurobiological processes to promote CNS tissue repair. Further, evaluation of the current landscape of regenerative medicine and tissue engineering techniques external to the neurosciences provides key perspectives into the role of the ECM in the use of stem cell-based therapies, and the potential directions future neuroregenerative approaches may take. If the most successful of these approaches achieve wide-spread adoption, innovative paired NSC-ECM strategies for neuroregeneration may become prominent in the near future, and with the rapid advances these techniques are poised to herald, a new era of treatment for CNS injury may dawn.
Collapse
Affiliation(s)
- Eshan B. Damle
- Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Vivianne E. Morrison
- Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jozef Cioma
- Faculty of Biology, Medicine, and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Milla Volic
- Faculty of Biology, Medicine, and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Gregory J. Bix
- Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
3
|
Radoszkiewicz K, Jezierska-Woźniak K, Waśniewski T, Sarnowska A. Understanding Intra- and Inter-Species Variability in Neural Stem Cells' Biology Is Key to Their Successful Cryopreservation, Culture, and Propagation. Cells 2023; 12:cells12030488. [PMID: 36766833 PMCID: PMC9914787 DOI: 10.3390/cells12030488] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Although clinical trials on human neural stem cells (hNSCs) have already been implemented in the treatment of neurological diseases and they have demonstrated their therapeutic effects, many questions remain in the field of preclinical research regarding the biology of these cells, their therapeutic properties, and their neurorestorative potential. Unfortunately, scientific reports are inconsistent and much of the NSCs research has been conducted on rodents rather than human cells for ethical reasons or due to insufficient cell material. Therefore, a question arises as to whether or which conclusions drawn on the isolation, cell survival, proliferation, or cell fate observed in vitro in rodent NSCs can be introduced into clinical applications. This paper presents the effects of different spatial, nutritional, and dissociation conditions on NSCs' functional properties, which are highly species-dependent. Our study confirmed that the discrepancies in the available literature on NSCs survival, proliferation, and fate did not only depend on intra-species factors and applied environmental conditions, but they were also affected by significant inter-species variability. Human and rodent NSCs share one feature, i.e., the necessity to be cultured immediately after isolation, which significantly maintains their survival. Additionally, in the absence of experiments on human cells, rat NSCs biology (neurosphere formation potential and neural differentiation stage) seems closer to that of humans rather than mice in response to environmental factors.
Collapse
Affiliation(s)
- Klaudia Radoszkiewicz
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Katarzyna Jezierska-Woźniak
- Department of Neurosurgery, Laboratory for Regenerative Medicine, Stem Cells Bank, University of Warmia and Mazury in Olsztyn, 10-720 Olsztyn, Poland
| | - Tomasz Waśniewski
- Department of Obstetrics and Gynaecology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-561 Olsztyn, Poland
| | - Anna Sarnowska
- Translational Platform for Regenerative Medicine, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-608-6598
| |
Collapse
|
4
|
Mouse Neural Stem Cell Differentiation and Human Adipose Mesenchymal Stem Cell Transdifferentiation Into Neuron- and Oligodendrocyte-like Cells With Myelination Potential. Stem Cell Rev Rep 2021; 18:732-751. [PMID: 34780018 DOI: 10.1007/s12015-021-10218-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 01/09/2023]
Abstract
Stem cell therapy is an interesting approach for neural repair, once it can improve and increase processes, like angiogenesis, neurogenesis, and synaptic plasticity. In this regard, adult neural stem cells (NSC) are studied for their mechanisms of proliferation, differentiation and functionality in neural repair. Here, we describe novel neural differentiation methods. NSC from adult mouse brains and human adipose-derived stem cells (hADSC) were isolated and characterized regarding their neural differentiation potential based on neural marker expression profiles. For both cell types, their capabilities of differentiating into neuron-, astrocyte- and oligodendrocytes-like cells (NLC, ALC and OLC, respectively) were analyzed. Our methodologies were capable of producing NLC, ALC and OLC from adult murine and human transdifferentiated NSC. NSC showed augmented gene expression of NES, TUJ1, GFAP and PDGFRA/Cnp. Following differentiation induction into NLC, OLC or ALC, specific neural phenotypes were obtained expressing MAP2, GalC/O4 or GFAP with compatible morphologies, respectively. Accordingly, immunostaining for nestin+ in NSC, GFAP+ in astrocytes and GalC/O4+ in oligodendrocytes was detected. Co-cultured NLC and OLC showed excitability in 81.3% of cells and 23.5% of neuron/oligodendrocyte marker expression overlap indicating occurrence of in vitro myelination. We show here that hADSC can be transdifferentiated into NSC and distinct neural phenotypes with the occurrence of neuron myelination in vitro, providing novel strategies for CNS regeneration therapy. Superior Part: Schematic organization of obtaining and generating hNSC from hADSC and differentiation processes and phenotypic expression of neuron, astrocyte and oligodendrocyte markers (MAP2, GFAP and O4, respectively) and stem cell marker (NES) of differentiating hNSC 14 days after induction. The nuclear staining in blue corresponds to DAPI. bar = 100 μm. Inferior part: Neural phenotype fates in diverse differentiation media. NES: nestin; GFAP: Glial fibrillary acidic protein. MAP2: Microtubule-associated protein 2. TUJ1: β-III tubulin. PDGFRA: PDGF receptor alpha. Two-way ANOVA with Bonferroni post-test with n = 3. * p < 0.05 and ** p < 0.01: (NSCiM1 NSC induction medium 1) vs differentiation media.
Collapse
|
5
|
Nandakumar S, Shahani P, Datta I, Pal R. Interventional Strategies for Parkinson Disease: Can Neural Precursor Cells Forge a Path Ahead? ACS Chem Neurosci 2021; 12:3785-3794. [PMID: 34628850 DOI: 10.1021/acschemneuro.1c00525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neural precursor cells (NPCs), derived from pluripotent stem cells (PSCs), with their unique ability to generate multiple neuronal and glial cell types are extremely useful for understanding biological mechanisms in normal and diseased states. However, generation of specific neuronal subtypes (mature) from NPCs in large numbers adequate for cell therapy is challenging due to lack of a thorough understanding of the cues that govern their differentiation. Interestingly, neural stem cells (NSCs) themselves are in consideration for therapy given their potency to form different neural cell types, release of trophic factors, and immunomodulatory effects that confer neuroprotection. With the recent COVID-19 outbreak and its accompanying neurological indications, the immunomodulatory role of NSCs may gain additional significance in the prevention of disease progression in vulnerable populations. In this regard, small-molecule mediated NPC generation from PSCs via NSC formation has become an important strategy that ensures consistency and robustness of the process. The development of the mammalian brain occurs along the rostro-caudal axis, and the establishment of anterior identity is an early event. Wnt signaling, along with fibroblast growth factor and retinoic acid, acts as a caudalization signal. Further, the increasing amount of epigenetic data available from human fetal brain development has enhanced both our understanding of and ability to experimentally manipulate these developmental regulatory programs in vitro. However, the impact on homing and engraftment after transplantation and subsequently on therapeutic efficacy of NPCs based on their derivation strategy is not yet clear. Another formidable challenge in cell replacement therapy for neurodegenerative disorders is the mode of delivery. In this Perspective, we discuss these core ideas with insights from our preliminary studies exploring the role of PSC-derived NPCs in rat models of MPTP-induced Parkinson's disease following intranasal injections.
Collapse
Affiliation(s)
- Swapna Nandakumar
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru 560065, Karnataka, India
| | - Pradnya Shahani
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru 560029, Karnataka, India
| | - Indrani Datta
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru 560029, Karnataka, India
| | - Rajarshi Pal
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru 560065, Karnataka, India
| |
Collapse
|
6
|
Wu X, Shen Q, Zhang Z, Zhang D, Gu Y, Xing D. Photoactivation of TGFβ/SMAD signaling pathway ameliorates adult hippocampal neurogenesis in Alzheimer's disease model. Stem Cell Res Ther 2021; 12:345. [PMID: 34116709 PMCID: PMC8196501 DOI: 10.1186/s13287-021-02399-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
Background Adult hippocampal neurogenesis (AHN) is restricted under the pathological conditions of neurodegenerative diseases, especially in Alzheimer’s disease (AD). The drop of AHN reduces neural circuit plasticity, resulting in the decrease of the generation of newborn neurons in dentate gyrus (DG), which makes it difficult to recover from learning/memory dysfunction in AD, therefore, it is imperative to find a therapeutic strategy to promote neurogenesis and clarify its underlying mechanism involved. Methods Amyloid precursor protein/presenilin 1 (APP/PS1) mice were treated with photobiomodulation therapy (PBMT) for 0.1 mW/mm2 per day in the dark for 1 month (10 min for each day). The neural stem cells (NSCs) were isolated from hippocampus of APP/PS1 transgenic mice at E14, and the cells were treated with PBMT for 0.667 mW/mm2 in the dark (5 min for each time). Results In this study, photobiomodulation therapy (PBMT) is found to promote AHN in APP/PS1 mice. The latent transforming growth factor-β1 (LTGFβ1) was activated in vitro and in vivo during PBMT-induced AHN, which promoted the differentiation of hippocampal APP/PS1 NSCs into newborn neurons. In particular, behavioral experiments showed that PBMT enhanced the spatial learning/memory ability of APP/PS1 mice. Mechanistically, PBMT-stimulated reactive oxygen species (ROS) activates TGFβ/Smad signaling pathway to increase the interaction of the transcription factors Smad2/3 with Smad4 and competitively reduce the association of Smad1/5/9 with Smad4, thereby significantly upregulating the expression of doublecortin (Dcx)/neuronal class-III β-tubulin (Tuj1) and downregulating the expression of glial fibrillary acidic protein (GFAP). These in vitro effects were abrogated when eliminating ROS. Furthermore, specific inhibition of TGFβ receptor I (TGFβR I) attenuates the DNA-binding efficiency of Smad2/3 to the Dcx promotor triggered by PBMT. Conclusion Our study demonstrates that PBMT, as a viable therapeutic strategy, directs the adult hippocampal APP/PS1 NSCs differentiate towards neurons, which has great potential value for ameliorating the drop of AHN in Alzheimer’s disease mice. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02399-2.
Collapse
Affiliation(s)
- Xiaolei Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Qi Shen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Zhan Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Di Zhang
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Ying Gu
- Department of Laser Medicine, First Medical Center of PLA General Hospital, Beijing, 100853, China.
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China. .,Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
7
|
Joshi HP, Jo HJ, Kim YH, An SB, Park CK, Han I. Stem Cell Therapy for Modulating Neuroinflammation in Neuropathic Pain. Int J Mol Sci 2021; 22:ijms22094853. [PMID: 34063721 PMCID: PMC8124149 DOI: 10.3390/ijms22094853] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Neuropathic pain (NP) is a complex, debilitating, chronic pain state, heterogeneous in nature and caused by a lesion or disease affecting the somatosensory system. Its pathogenesis involves a wide range of molecular pathways. NP treatment is extremely challenging, due to its complex underlying disease mechanisms. Current pharmacological and nonpharmacological approaches can provide long-lasting pain relief to a limited percentage of patients and lack safe and effective treatment options. Therefore, scientists are focusing on the introduction of novel treatment approaches, such as stem cell therapy. A growing number of reports have highlighted the potential of stem cells for treating NP. In this review, we briefly introduce NP, current pharmacological and nonpharmacological treatments, and preclinical studies of stem cells to treat NP. In addition, we summarize stem cell mechanisms—including neuromodulation in treating NP. Literature searches were conducted using PubMed to provide an overview of the neuroprotective effects of stem cells with particular emphasis on recent translational research regarding stem cell-based treatment of NP, highlighting its potential as a novel therapeutic approach.
Collapse
Affiliation(s)
- Hari Prasad Joshi
- Department of Neurosurgery, School of Medicine, CHA University, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (H.P.J.); (S.-B.A.)
- Spinal Cord Research Centre, Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Hyun-Jung Jo
- Gachon Pain Center, Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Gyeonggi-do, Korea; (H.-J.J.); (Y.-H.K.)
| | - Yong-Ho Kim
- Gachon Pain Center, Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Gyeonggi-do, Korea; (H.-J.J.); (Y.-H.K.)
| | - Seong-Bae An
- Department of Neurosurgery, School of Medicine, CHA University, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (H.P.J.); (S.-B.A.)
| | - Chul-Kyu Park
- Gachon Pain Center, Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Gyeonggi-do, Korea; (H.-J.J.); (Y.-H.K.)
- Correspondence: (C.-K.P.); (I.H.)
| | - Inbo Han
- Department of Neurosurgery, School of Medicine, CHA University, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (H.P.J.); (S.-B.A.)
- Correspondence: (C.-K.P.); (I.H.)
| |
Collapse
|
8
|
Carroll JA, Foliaki ST, Haigh CL. A 3D cell culture approach for studying neuroinflammation. J Neurosci Methods 2021; 358:109201. [PMID: 33932455 DOI: 10.1016/j.jneumeth.2021.109201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/13/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neurodegenerative diseases are highly complex making them challenging to model in cell culture. All cell types of the brain have been implicated as exerting an effect on pathogenesis, and disease progression is likely influenced by the cross-talk between the different cell types. Sophisticated investigation of the cellular level consequences of cross-talk between different cells types requires three-dimensional (3D) co-culture systems. NEW METHOD Murine neural stem cells were differentiated into mixed-neuronal lineage populations in 3D culture. By seeding these differentiated cultures with microglia from adult brain, we have generated a 3D ex-vivo model of murine brain tissue populated with microglia. RESULTS Monitoring the infiltration of GFP-expressing microglia into the 3D neuronal lineage cultures showed population throughout the tissue and assumption of ramified homeostatic morphology by the microglia. The co-cultures showed good longevity and were functionally responsive to external stimuli. COMPARISON WITH EXISTING METHODS We have previously used 2-dimensional adhered cultures to model cell-cell interactions between microglia and neuronal lineage cells. While the microglia integrate well into these cultures and demonstrate inter-cellular cross-talk, it is known that adhered culture can change their activation state and therefore a 3D system better represents communication throughout a network of neuronal and support cells. CONCLUSIONS Our system offers a straight-forward and time effective way to model 3D mouse brain tissue that is responsive to external neuroinflammatory stimulus. It not only allows inter-cellular interactions to be studied in live tissue but additionally permits study of changes within any available mouse genotype.
Collapse
Affiliation(s)
- James A Carroll
- TSE/Prion and Retroviral Pathogenesis Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - Simote T Foliaki
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - Cathryn L Haigh
- Prion Cell Biology Unit, Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA.
| |
Collapse
|
9
|
Petrović A, Ban J, Tomljanović I, Pongrac M, Ivaničić M, Mikašinović S, Mladinic M. Establishment of Long-Term Primary Cortical Neuronal Cultures From Neonatal Opossum Monodelphis domestica. Front Cell Neurosci 2021; 15:661492. [PMID: 33815068 PMCID: PMC8012671 DOI: 10.3389/fncel.2021.661492] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/25/2021] [Indexed: 11/13/2022] Open
Abstract
Primary dissociated neuronal cultures have become a standard model for studying central nervous system (CNS) development. Such cultures are predominantly prepared from the hippocampus or cortex of rodents (mice and rats), while other mammals are less used. Here, we describe the establishment and extensive characterization of the primary dissociated neuronal cultures derived from the cortex of the gray South American short-tailed opossums, Monodelphis domestica. Opossums are unique in their ability to fully regenerate their CNS after an injury during their early postnatal development. Thus, we used cortex of postnatal day (P) 3-5 opossum to establish long-surviving and nearly pure neuronal cultures, as well as mixed cultures composed of radial glia cells (RGCs) in which their neurogenic and gliogenic potential was confirmed. Both types of cultures can survive for more than 1 month in vitro. We also prepared neuronal cultures from the P16-18 opossum cortex, which were composed of astrocytes and microglia, in addition to neurons. The long-surviving opossum primary dissociated neuronal cultures represent a novel mammalian in vitro platform particularly useful to study CNS development and regeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Miranda Mladinic
- Laboratory for Molecular Neurobiology, Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
10
|
Zhou H, Yang H, Lu L, Li X, Pan B, Fu Z, Chu T, Liu J, Kang Y, Liu L, Ning G, Ding W, Wu P, Kong X, Feng S. A modified protocol for the isolation, culture, and cryopreservation of rat embryonic neural stem cells. Exp Ther Med 2020; 20:156. [PMID: 33093894 DOI: 10.3892/etm.2020.9285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 05/16/2019] [Indexed: 11/05/2022] Open
Abstract
Neural stem cells (NSCs) are characterized by their potential for self-renewal and ability to differentiate into neurons, astrocytes, and oligodendrocytes. They are of great value to scientific studies and clinical applications. Culturing NSCs in vitro is important for characterizing their properties under controlled environmental conditions that may be modified and monitored accurately. The present study explored a modified, detailed and efficient protocol for the isolation, culture and cryopreservation of rat embryonic NSCs. In particular, the viability, nestin expression, and self-renewal and multi-differentiation capabilities of NSCs cryopreserved for various periods of time (7 days, or 1, 6 or 12 months) were characterized and compared. Rat embryonic NSCs were successfully obtained and maintained their self-renewal and multipotent differentiation capabilities even following long-term cryopreservation (for up to 12 months).
Collapse
Affiliation(s)
- Hengxing Zhou
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Huan Yang
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221006, P.R. China
| | - Lu Lu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Xueying Li
- Key Laboratory of Immuno Microenvironment and Disease of the Educational Ministry of China, Department of Immunology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Bin Pan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Zheng Fu
- Key Laboratory of Immuno Microenvironment and Disease of the Educational Ministry of China, Department of Immunology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Tianci Chu
- Department of Pediatrics, Kosair Children's Hospital Research Institute, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Jun Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Yi Kang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Lu Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Guangzhi Ning
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| | - Wenyuan Ding
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Ping Wu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555-1043, USA
| | - Xiaohong Kong
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China.,Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin City, Tianjin 300052, P.R. China
| |
Collapse
|
11
|
Murphy AR, Haynes JM, Laslett AL, Cameron NR, O'Brien CM. Three-dimensional differentiation of human pluripotent stem cell-derived neural precursor cells using tailored porous polymer scaffolds. Acta Biomater 2020; 101:102-116. [PMID: 31610339 DOI: 10.1016/j.actbio.2019.10.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/30/2019] [Accepted: 10/07/2019] [Indexed: 02/07/2023]
Abstract
This study investigates the utility of a tailored poly(ethylene glycol) diacrylate-crosslinked porous polymeric tissue engineering scaffold, with mechanical properties specifically optimised to be comparable to that of mammalian brain tissue for 3D human neural cell culture. Results obtained here demonstrate the attachment, proliferation and terminal differentiation of both human induced pluripotent stem cell- and embryonic stem cell-derived neural precursor cells (hPSC-NPCs) throughout the interconnected porous network within laminin-coated scaffolds. Phenotypic data and functional analyses are presented demonstrating that this material supports terminal in vitro neural differentiation of hPSC-NPCs to a mixed population of viable neuronal and glial cells for periods of up to 49 days. This is evidenced by the upregulation of TUBB3, MAP2, SYP and GFAP gene expression, as well as the presence of the proteins βIII-TUBULIN, NEUN, MAP2 and GFAP. Functional maturity of neural cells following 49 days 3D differentiation culture was tested via measurement of intracellular calcium. These analyses revealed spontaneously active, synchronous and rhythmic calcium flux, as well as response to the neurotransmitter glutamate. This tailored construct has potential application as an improved in vitro human neurogenesis model with utility in platform drug discovery programs. STATEMENT OF SIGNIFICANCE: The interconnected porosity of polyHIPE scaffolds exhibits the ability to support three-dimensional neural cell network formation due to limited resistance to cellular migration and re-organisation. The previously developed scaffold material displays mechanical properties similar to that of the mammalian brain. This research also employs the utility of pluripotent stem cell-derived neural cells which are of greater clinical relevance than primary neural cell lines. This scaffold material has future potential in better mimicking three-dimensional neural networks found in the human brain and may result in improved in vitro models for disease modelling and drug screening applications.
Collapse
|
12
|
Trehalose as glucose surrogate in proliferation and cellular mobility of adult neural progenitor cells derived from mouse hippocampus. J Neural Transm (Vienna) 2019; 126:1485-1491. [PMID: 31468180 DOI: 10.1007/s00702-019-02070-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/19/2019] [Indexed: 01/21/2023]
Abstract
The disaccharide trehalose (TRE) represents a natural energy supply for distinct non-mammalian species. Evidence has shown that TRE impacts on various properties including the stabilization of protein structure and cell membranes, which are important neuroprotective features against neurodegeneration. In this study, we tested the specific effect of TRE on cell proliferation and mobilization using an established experimental paradigm of adult neural progenitor cells (NPCs) derived from murine hippocampus. NPC proliferation, both measured by growth curve analysis over 25 days and by bromodeoxyuridine (BrdU) incorporation, was not altered by adding TRE instead of GLC to the culture media. Using Boyden chamber experiments, the mobility in regular glucose-containing media did not differ from glucose-free TRE-supplemented media. Our observation suggests that TRE has the capacity to replace glucose (GLC) as energy source in neural cells in our experimental paradigm.
Collapse
|
13
|
Massimino L, Flores-Garcia L, Di Stefano B, Colasante G, Icoresi-Mazzeo C, Zaghi M, Hamilton BA, Sessa A. TBR2 antagonizes retinoic acid dependent neuronal differentiation by repressing Zfp423 during corticogenesis. Dev Biol 2018; 434:231-248. [PMID: 29305158 PMCID: PMC7032051 DOI: 10.1016/j.ydbio.2017.12.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/26/2017] [Accepted: 12/28/2017] [Indexed: 01/14/2023]
Abstract
During cerebral cortex development, neural progenitors are required to elaborate a variety of cell differentiation signals to which they are continuously exposed. RA acid is a potent inducer of neuronal differentiation as it was found to influence cortical development. We report herein that TBR2, a transcription factor specific to Intermediate (Basal) Neural Progenitors (INPs), represses activation of the RA responsive element and expression of RA target genes in cell lines. This repressive action on RA signaling was functionally confirmed by the decrease of RA-mediated neuronal differentiation in neural stem cells stably overexpressing TBR2. In vivo mapping of RA activity in the developing cortex indicated that RA activity is detected in radial glial cells and subsequently downregulated in INPs, revealing a fine cell-type specific regulation of its signaling. Thus, TBR2 might be a molecular player in opposing RA signaling in INPs. Interestingly, this negative regulation is achieved at least in part by directly repressing the critical nuclear RA co-factor ZFP423. Indeed, we found ZFP423 to be expressed in the developing cortex and promote RA-dependent neuronal differentiation. These data indicate that TBR2 contributes to suppressing RA signaling in INPs, thereby enabling them to re-enter the cell cycle and delay neuronal differentiation.
Collapse
Affiliation(s)
- Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Lisbeth Flores-Garcia
- Departments of Cellular&Molecular Medicine and Medicine, Moores Cancer Center, and Institute for Genomic Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0644, USA
| | - Bruno Di Stefano
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gaia Colasante
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Cecilia Icoresi-Mazzeo
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mattia Zaghi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Bruce A Hamilton
- Departments of Cellular&Molecular Medicine and Medicine, Moores Cancer Center, and Institute for Genomic Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0644, USA
| | - Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.
| |
Collapse
|
14
|
Tanaka Y, Isomura T, Shimba K, Kotani K, Jimbo Y. Neurogenesis Enhances Response Specificity to Spatial Pattern Stimulation in Hippocampal Cultures. IEEE Trans Biomed Eng 2017; 64:2555-2561. [DOI: 10.1109/tbme.2016.2639468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
15
|
Tang Y, Yu P, Cheng L. Current progress in the derivation and therapeutic application of neural stem cells. Cell Death Dis 2017; 8:e3108. [PMID: 29022921 PMCID: PMC5682670 DOI: 10.1038/cddis.2017.504] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/28/2017] [Accepted: 08/31/2017] [Indexed: 12/13/2022]
Abstract
Neural stem cells (NSCs) have a unique role in neural regeneration. Cell therapy based on NSC transplantation is a promising tool for the treatment of nervous system diseases. However, there are still many issues and controversies associated with the derivation and therapeutic application of these cells. In this review, we summarize the different sources of NSCs and their derivation methods, including direct isolation from primary tissues, differentiation from pluripotent stem cells and transdifferentiation from somatic cells. We also review the current progress in NSC implantation for the treatment of various neural defects and injuries in animal models and clinical trials. Finally, we discuss potential optimization strategies for NSC derivation and propose urgent challenges to the clinical translation of NSC-based therapies in the near future.
Collapse
Affiliation(s)
- Yuewen Tang
- National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Shanghai Institute of Haematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei Yu
- Department of Orthopaedics, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Cheng
- National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Shanghai Institute of Haematology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Developmental neurogenesis in mouse and Xenopus is impaired in the absence of Nosip. Dev Biol 2017; 429:200-212. [DOI: 10.1016/j.ydbio.2017.06.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 01/01/2023]
|
17
|
Srivastava AK, Gross SK, Almad AA, Bulte CA, Maragakis NJ, Bulte JWM. Serial in vivo imaging of transplanted allogeneic neural stem cell survival in a mouse model of amyotrophic lateral sclerosis. Exp Neurol 2016; 289:96-102. [PMID: 28038988 DOI: 10.1016/j.expneurol.2016.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/27/2016] [Accepted: 12/23/2016] [Indexed: 12/13/2022]
Abstract
Neural stem cells (NSCs) are being investigated as a possible treatment for amyotrophic lateral sclerosis (ALS) through intraspinal transplantation, but no longitudinal imaging studies exist that describe the survival of engrafted cells over time. Allogeneic firefly luciferase-expressing murine NSCs (Luc+-NSCs) were transplanted bilaterally (100,000 cells/2μl) into the cervical spinal cord (C5) parenchyma of pre-symptomatic (63day-old) SOD1G93A ALS mice (n=14) and wild-type age-matched littermates (n=14). Six control SOD1G93A ALS mice were injected with saline. Mice were immunosuppressed using a combination of tacrolimus+sirolimus (1mg/kg each, i.p.) daily. Compared to saline-injected SOD1G93A ALS control mice, a transient improvement (p<0.05) in motor performance (rotarod test) was observed after NSC transplantation only at the early disease stage (weeks 2 and 3 post-transplantation). Compared to day one post-transplantation, there was a significant decline in bioluminescent imaging (BLI) signal in SOD1G93A ALS mice at the time of disease onset (71.7±17.9% at 4weeks post-transplantation, p<0.05), with a complete loss of BLI signal at endpoint (120day-old mice). In contrast, BLI signal intensity was observed in wild-type littermates throughout the entire study period, with only a 41.4±8.7% decline at the endpoint. In SOD1G93A ALS mice, poor cell survival was accompanied by accumulation of mature macrophages and the presence of astrogliosis and microgliosis. We conclude that the disease progression adversely affects the survival of engrafted murine Luc+-NSCs in SOD1G93A ALS mice as a result of the hostile ALS spinal cord microenvironment, further emphasizing the challenges that face successful cell therapy of ALS.
Collapse
Affiliation(s)
- Amit K Srivastava
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sarah K Gross
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Akshata A Almad
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Camille A Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nicholas J Maragakis
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
18
|
Singh VK, Saini A, Kalsan M, Kumar N, Chandra R. Describing the Stem Cell Potency: The Various Methods of Functional Assessment and In silico Diagnostics. Front Cell Dev Biol 2016; 4:134. [PMID: 27921030 PMCID: PMC5118841 DOI: 10.3389/fcell.2016.00134] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/02/2016] [Indexed: 12/11/2022] Open
Abstract
Stem cells are defined by their capabilities to self-renew and give rise to various types of differentiated cells depending on their potency. They are classified as pluripotent, multipotent, and unipotent as demonstrated through their potential to generate the variety of cell lineages. While pluripotent stem cells may give rise to all types of cells in an organism, Multipotent and Unipotent stem cells remain restricted to the particular tissue or lineages. The potency of these stem cells can be defined by using a number of functional assays along with the evaluation of various molecular markers. These molecular markers include diagnosis of transcriptional, epigenetic, and metabolic states of stem cells. Many reports are defining the particular set of different functional assays, and molecular marker used to demonstrate the developmental states and functional capacities of stem cells. The careful evaluation of all these methods could help in generating standard identifying procedures/markers for them.
Collapse
Affiliation(s)
- Vimal K Singh
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Abhishek Saini
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Manisha Kalsan
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Neeraj Kumar
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Ramesh Chandra
- Department of Chemistry, University of Delhi Delhi, India
| |
Collapse
|
19
|
Francipane MG, Lagasse E. Towards Organs on Demand: Breakthroughs and Challenges in Models of Organogenesis. CURRENT PATHOBIOLOGY REPORTS 2016; 4:77-85. [PMID: 28979828 DOI: 10.1007/s40139-016-0111-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In recent years, functional three-dimensional (3D) tissue generation in vitro has been significantly advanced by tissue-engineering methods, achieving better reproduction of complex native organs compared to conventional culture systems. This review will discuss traditional 3D cell culture techniques as well as newly developed technology platforms. These recent techniques provide new possibilities in the creation of human body parts and provide more accurate predictions of tissue response to drug and chemical challenges. Given the rapid advancement in the human induced pluripotent stem cell (iPSC) field, these platforms also hold great promise in the development of patient-specific, transplantable tissues and organs on demand.
Collapse
Affiliation(s)
- Maria Giovanna Francipane
- McGowan Institute for Regenerative Medicine, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
- Ri.MED Foundation, 90133 Palermo, Italy
| | - Eric Lagasse
- McGowan Institute for Regenerative Medicine, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| |
Collapse
|
20
|
SOX2 primes the epigenetic landscape in neural precursors enabling proper gene activation during hippocampal neurogenesis. Proc Natl Acad Sci U S A 2015; 112:E1936-45. [PMID: 25825708 DOI: 10.1073/pnas.1421480112] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Newborn granule neurons generated from neural progenitor cells (NPCs) in the adult hippocampus play a key role in spatial learning and pattern separation. However, the molecular mechanisms that control activation of their neurogenic program remain poorly understood. Here, we report a novel function for the pluripotency factor sex-determining region Y (SRY)-related HMG box 2 (SOX2) in regulating the epigenetic landscape of poised genes activated at the onset of neuronal differentiation. We found that SOX2 binds to bivalently marked promoters of poised proneural genes [neurogenin 2 (Ngn2) and neurogenic differentiation 1 (NeuroD1)] and a subset of neurogenic genes [e.g., SRY-box 21 (Sox21), brain-derived neurotrophic factor (Bdnf), and growth arrest and DNA-damage-inducible, beta (Gadd45b)] where it functions to maintain the bivalent chromatin state by preventing excessive polycomb repressive complex 2 activity. Conditional ablation of SOX2 in adult hippocampal NPCs impaired the activation of proneural and neurogenic genes, resulting in increased neuroblast death and functionally aberrant newborn neurons. We propose that SOX2 sets a permissive epigenetic state in NPCs, thus enabling proper activation of the neuronal differentiation program under neurogenic cue.
Collapse
|
21
|
Ray B, Chopra N, Long JM, Lahiri DK. Human primary mixed brain cultures: preparation, differentiation, characterization and application to neuroscience research. Mol Brain 2014; 7:63. [PMID: 25223359 PMCID: PMC4181361 DOI: 10.1186/s13041-014-0063-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 08/15/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Culturing primary cortical neurons is an essential neuroscience technique. However, most cultures are derived from rodent brains and standard protocols for human brain cultures are sparse. Herein, we describe preparation, maintenance and major characteristics of a primary human mixed brain culture, including neurons, obtained from legally aborted fetal brain tissue. This approach employs standard materials and techniques used in the preparation of rodent neuron cultures, with critical modifications. RESULTS This culture has distinct differences from rodent cultures. Specifically, a significant numbers of cells in the human culture are derived from progenitor cells, and the yield and survival of the cells grossly depend on the presence of bFGF. In the presence of bFGF, this culture can be maintained for an extended period. Abundant productions of amyloid-β, tau and proteins make this a powerful model for Alzheimer's research. The culture also produces glia and different sub-types of neurons. CONCLUSION We provide a well-characterized methodology for human mixed brain cultures useful to test therapeutic agents under various conditions, and to carry forward mechanistic and translational studies for several brain disorders.
Collapse
|
22
|
Adult stem cell as new advanced therapy for experimental neuropathic pain treatment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:470983. [PMID: 25197647 PMCID: PMC4147203 DOI: 10.1155/2014/470983] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 07/23/2014] [Indexed: 02/08/2023]
Abstract
Neuropathic pain (NP) is a highly invalidating disease resulting as consequence of a lesion or disease affecting the somatosensory system. All the pharmacological treatments today in use give a long lasting pain relief only in a limited percentage of patients before pain reappears making NP an incurable disease. New approaches are therefore needed and research is testing stem cell usage. Several papers have been written on experimental neuropathic pain treatment using stem cells of different origin and species to treat experimental NP. The original idea was based on the capacity of stem cell to offer a totipotent cellular source for replacing injured neural cells and for delivering trophic factors to lesion site; soon the researchers agreed that the capacity of stem cells to contrast NP was not dependent upon their regenerative effect but was mostly linked to a bidirectional interaction between the stem cell and damaged microenvironment resident cells. In this paper we review the preclinical studies produced in the last years assessing the effects induced by several stem cells in different models of neuropathic pain. The overall positive results obtained on pain remission by using stem cells that are safe, of easy isolation, and which may allow an autologous transplant in patients may be encouraging for moving from bench to bedside, although there are several issues that still need to be solved.
Collapse
|
23
|
Shelley BC, Gowing G, Svendsen CN. A cGMP-applicable expansion method for aggregates of human neural stem and progenitor cells derived from pluripotent stem cells or fetal brain tissue. J Vis Exp 2014. [PMID: 24962813 DOI: 10.3791/51219] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A cell expansion technique to amass large numbers of cells from a single specimen for research experiments and clinical trials would greatly benefit the stem cell community. Many current expansion methods are laborious and costly, and those involving complete dissociation may cause several stem and progenitor cell types to undergo differentiation or early senescence. To overcome these problems, we have developed an automated mechanical passaging method referred to as "chopping" that is simple and inexpensive. This technique avoids chemical or enzymatic dissociation into single cells and instead allows for the large-scale expansion of suspended, spheroid cultures that maintain constant cell/cell contact. The chopping method has primarily been used for fetal brain-derived neural progenitor cells or neurospheres, and has recently been published for use with neural stem cells derived from embryonic and induced pluripotent stem cells. The procedure involves seeding neurospheres onto a tissue culture Petri dish and subsequently passing a sharp, sterile blade through the cells effectively automating the tedious process of manually mechanically dissociating each sphere. Suspending cells in culture provides a favorable surface area-to-volume ratio; as over 500,000 cells can be grown within a single neurosphere of less than 0.5 mm in diameter. In one T175 flask, over 50 million cells can grow in suspension cultures compared to only 15 million in adherent cultures. Importantly, the chopping procedure has been used under current good manufacturing practice (cGMP), permitting mass quantity production of clinical-grade cell products.
Collapse
|
24
|
Yao NW, Chen CCV, Yen CT, Chang C. Promoted Growth of Brain Tumor by the Transplantation of Neural Stem/Progenitor Cells Facilitated by CXCL12. Transl Oncol 2014; 7:S1936-5233(14)00042-4. [PMID: 24862537 PMCID: PMC4145393 DOI: 10.1016/j.tranon.2014.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 04/10/2014] [Accepted: 04/14/2014] [Indexed: 12/17/2022] Open
Abstract
The targeted migration of neural stem/progenitor cells (NSPCs) is a prerequisite for the use of stem cell therapy in the treatment of pathologies. This migration is regulated mainly by C-X-C motif chemokine 12 (CXCL12). Therefore, promotion of the migratory responses of grafted cells by upregulating CXCL12 signaling has been proposed as a strategy for improving the efficacy of such cell therapies. However, the effects of this strategy on brain tumors have not yet been examined in vivo. The aim of the present study was thus to elucidate the effects of grafted rat green fluorescent protein (GFP)-labeled NSPCs (GFP-NSPCs) with CXCL12 enhancement on a model of spontaneous rat brain tumor induced by N-ethyl-N-nitrosourea. T2-weighted magnetic resonance imaging was applied to determine the changes in tumor volume and morphology over time. Postmortem histology was performed to confirm the tumor pathology, expression levels of CXCL12 and C-X-C chemokine receptor type 4, and the fate of GFP-NSPCs. The results showed that the tumor volume and hypointense areas of T2-weighted images were both significantly increased in animals treated with combined NSPC transplantation and CXCL12 induction, but not in control animals or in those with tumors that received only one of the treatments. GFP-NSPCs appear to migrate toward tumors with CXCL12 enhancement and differentiate uniquely into a neuronal lineage. These findings suggest that CXCL12 is an effective chemoattractant that facilitates exogenous NSPC migration toward brain tumors and that CXCL12 and NSPC can act synergistically to promote tumor progression with severe hemorrhage.
Collapse
Affiliation(s)
- Nai-Wei Yao
- Department of Life Science, National Taiwan University, Taipei, Taiwan; Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan
| | - Chiao-Chi V Chen
- Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan
| | - Chen-Tung Yen
- Department of Life Science, National Taiwan University, Taipei, Taiwan.
| | - Chen Chang
- Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan.
| |
Collapse
|
25
|
Abstract
As the emergence of cancer is most frequent in proliferating tissues, replication errors are considered to be at the base of this disease. This review concentrates mainly on two neural cancers, neuroblastoma and glioma, with completely different backgrounds that are well documented with respect to their ontogeny. Although clinical data on other cancers of the nervous system are available, usually little can be said about their origins. Neuroblastoma is initiated in the embryo at a moment when the nervous system (NS) is in full expansion and occasionally genomic damage can lead to neoplasia. Glioma, to the contrary, occurs in the adult brain supposed to be mostly in a postmitotic state. According to current consensus, neural stem cells located in the subventricular zone (SVZ) in the adult are thought to accumulate enough genomic mutations to diverge on a carcinogenic course leading to diverse forms of glioma. After weighing the pros and cons of this current hypothesis in this review, it will be argued that this may be improbable, yielding to the original old concept of glial origin of glioma.
Collapse
|
26
|
Mazzoccoli G, Tevy MF, Borghesan M, Delle Vergini MR, Vinciguerra M. Caloric restriction and aging stem cells: the stick and the carrot? Exp Gerontol 2013; 50:137-48. [PMID: 24211426 DOI: 10.1016/j.exger.2013.10.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/03/2013] [Accepted: 10/28/2013] [Indexed: 12/24/2022]
Abstract
Adult tissue stem cells have the ability to adjust to environmental changes and affect also the proliferation of neighboring cells, with important consequences on tissue maintenance and regeneration. Stem cell renewal and proliferation is strongly regulated during aging of the organism. Caloric restriction is the most powerful anti-aging strategy conserved throughout evolution in the animal kingdom. Recent studies relate the properties of caloric restriction to its ability in reprogramming stem-like cell states and in prolonging the capacity of stem cells to self-renew, proliferate, differentiate, and replace cells in several adult tissues. However this general paradigm presents with exceptions. The scope of this review is to highlight how caloric restriction impacts on diverse stem cell compartments and, by doing so, might differentially delay aging in the tissues of lower and higher organisms.
Collapse
Affiliation(s)
- Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", S. Giovanni Rotondo, FG, Italy.
| | - Maria Florencia Tevy
- Genomics and Bioinformatics Centre, Major University of Santiago, Santiago, Chile
| | - Michela Borghesan
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", S. Giovanni Rotondo, FG, Italy; University College London, Institute for Liver and Digestive Health, Division of Medicine, Royal Free Campus, London, United Kingdom
| | - Maria Rita Delle Vergini
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", S. Giovanni Rotondo, FG, Italy
| | - Manlio Vinciguerra
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS Scientific Institute and Regional General Hospital "Casa Sollievo della Sofferenza", S. Giovanni Rotondo, FG, Italy; Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy; University College London, Institute for Liver and Digestive Health, Division of Medicine, Royal Free Campus, London, United Kingdom.
| |
Collapse
|
27
|
Buta C, David R, Dressel R, Emgård M, Fuchs C, Gross U, Healy L, Hescheler J, Kolar R, Martin U, Mikkers H, Müller FJ, Schneider RK, Seiler AE, Spielmann H, Weitzer G. Reconsidering pluripotency tests: do we still need teratoma assays? Stem Cell Res 2013; 11:552-62. [PMID: 23611953 PMCID: PMC7615844 DOI: 10.1016/j.scr.2013.03.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/08/2013] [Accepted: 03/16/2013] [Indexed: 01/19/2023] Open
Abstract
The induction of teratoma in mice by the transplantation of stem cells into extra-uterine sites has been used as a read-out for cellular pluripotency since the initial description of this phenomenon in 1954. Since then, the teratoma assay has remained the assay of choice to demonstrate pluripotency, gaining prominence during the recent hype surrounding human stem cell research. However, the scientific significance of the teratoma assay has been debated due to the fact that transplanted cells are exposed to a non-physiological environment. Since many mice are used for a result that is heavily questioned, it is time to reconsider the teratoma assay from an ethical point of view. Candidate alternatives to the teratoma assay comprise the directed differentiation of pluripotent stem cells into organotypic cells, differentiation of cells in embryoid bodies, the analysis of pluripotency-associated biomarkers with high correlation to the teratoma forming potential of stem cells, predictive epigenetic footprints, or a combination of these technologies. Each of these assays is capable of addressing one or more aspects of pluripotency, however it is essential that these assays are validated to provide an accepted robust, reproducible alternative. In particular, the rapidly expanding number of human induced pluripotent stem cell lines, requires the development of simple, affordable standardized in vitro and in silico assays to reduce the number of animal experiments performed.
Collapse
Affiliation(s)
| | - Robert David
- Ludwig-Maximilians-Universität, München, Germany
| | | | - Mia Emgård
- Cellartis, Göteborg and Karolinska Institute Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
van Gorp S, Leerink M, Kakinohana O, Platoshyn O, Santucci C, Galik J, Joosten EA, Hruska-Plochan M, Goldberg D, Marsala S, Johe K, Ciacci JD, Marsala M. Amelioration of motor/sensory dysfunction and spasticity in a rat model of acute lumbar spinal cord injury by human neural stem cell transplantation. Stem Cell Res Ther 2013; 4:57. [PMID: 23710605 PMCID: PMC3706882 DOI: 10.1186/scrt209] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 04/25/2013] [Indexed: 12/15/2022] Open
Abstract
Introduction Intraspinal grafting of human neural stem cells represents a promising approach to promote recovery of function after spinal trauma. Such a treatment may serve to: I) provide trophic support to improve survival of host neurons; II) improve the structural integrity of the spinal parenchyma by reducing syringomyelia and scarring in trauma-injured regions; and III) provide neuronal populations to potentially form relays with host axons, segmental interneurons, and/or α-motoneurons. Here we characterized the effect of intraspinal grafting of clinical grade human fetal spinal cord-derived neural stem cells (HSSC) on the recovery of neurological function in a rat model of acute lumbar (L3) compression injury. Methods Three-month-old female Sprague–Dawley rats received L3 spinal compression injury. Three days post-injury, animals were randomized and received intraspinal injections of either HSSC, media-only, or no injections. All animals were immunosuppressed with tacrolimus, mycophenolate mofetil, and methylprednisolone acetate from the day of cell grafting and survived for eight weeks. Motor and sensory dysfunction were periodically assessed using open field locomotion scoring, thermal/tactile pain/escape thresholds and myogenic motor evoked potentials. The presence of spasticity was measured by gastrocnemius muscle resistance and electromyography response during computer-controlled ankle rotation. At the end-point, gait (CatWalk), ladder climbing, and single frame analyses were also assessed. Syrinx size, spinal cord dimensions, and extent of scarring were measured by magnetic resonance imaging. Differentiation and integration of grafted cells in the host tissue were validated with immunofluorescence staining using human-specific antibodies. Results Intraspinal grafting of HSSC led to a progressive and significant improvement in lower extremity paw placement, amelioration of spasticity, and normalization in thermal and tactile pain/escape thresholds at eight weeks post-grafting. No significant differences were detected in other CatWalk parameters, motor evoked potentials, open field locomotor (Basso, Beattie, and Bresnahan locomotion score (BBB)) score or ladder climbing test. Magnetic resonance imaging volume reconstruction and immunofluorescence analysis of grafted cell survival showed near complete injury-cavity-filling by grafted cells and development of putative GABA-ergic synapses between grafted and host neurons. Conclusions Peri-acute intraspinal grafting of HSSC can represent an effective therapy which ameliorates motor and sensory deficits after traumatic spinal cord injury.
Collapse
|
29
|
In Vivo c-Met Pathway Inhibition Depletes Human Glioma Xenografts of Tumor-Propagating Stem-Like Cells. Transl Oncol 2013; 6:104-11. [PMID: 23556031 DOI: 10.1593/tlo.13127] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 01/16/2013] [Accepted: 01/17/2013] [Indexed: 11/18/2022] Open
Abstract
Solid malignancies contain sphere-forming stem-like cells that are particularly efficient in propagating tumors. Identifying agents that target these cells will advance the development of more effective therapies. Recent converging evidence shows that c-Met expression marks tumor-initiating stem-like cells and that c-Met signaling drives human glioblastoma multiforme (GBM) cell stemness in vitro. However, the degree to which tumor-propagating stem-like cells depend on c-Met signaling in histologically complex cancers remains unknown. We examined the effects of in vivo c-Met pathway inhibitor therapy on tumor-propagating stem-like cells in human GBM xenografts. Animals bearing pre-established tumor xenografts expressing activated c-Met were treated with either neutralizing anti- hepatocyte growth factor (HGF) monoclonal antibody L2G7 or with the c-Met kinase inhibitor PF2341066 (Crizotinib). c-Met pathway inhibition inhibited tumor growth, depleted tumors of sphere-forming cells, and inhibited tumor expression of stem cell markers CD133, Sox2, Nanog, and Musashi. Withdrawing c-Met pathway inhibitor therapy resulted in a substantial rebound in stem cell marker expression concurrent with tumor recurrence. Cells derived from xenografts treated with anti-HGF in vivo were depleted of tumor-propagating potential as determined by in vivo serial dilution tumor-propagating assay. Furthermore, daughter xenografts that did form were 12-fold smaller than controls. These findings show that stem-like tumor-initiating cells are dynamically regulated by c-Met signaling in vivo and that c-Met pathway inhibitors can deplete tumors of their tumor-propagating stem-like cells.
Collapse
|
30
|
Chen CCV, Ku MC, D. M. J, Lai JS, Hueng DY, Chang C. Simple SPION incubation as an efficient intracellular labeling method for tracking neural progenitor cells using MRI. PLoS One 2013; 8:e56125. [PMID: 23468856 PMCID: PMC3585319 DOI: 10.1371/journal.pone.0056125] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 01/04/2013] [Indexed: 01/08/2023] Open
Abstract
Cellular magnetic resonance imaging (MRI) has been well-established for tracking neural progenitor cells (NPC). Superparamagnetic iron oxide nanoparticles (SPIONs) approved for clinical application are the most common agents used for labeling. Conventionally, transfection agents (TAs) were added with SPIONs to facilitate cell labeling because SPIONs in the native unmodified form were deemed inefficient for intracellular labeling. However, compelling evidence also shows that simple SPION incubation is not invariably ineffective. The labeling efficiency can be improved by prolonged incubation and elevated iron doses. The goal of the present study was to establish simple SPION incubation as an efficient intracellular labeling method. To this end, NPCs derived from the neonatal subventricular zone were incubated with SPIONs (Feridex®) and then evaluated in vitro with regard to the labeling efficiency and biological functions. The results showed that, following 48 hours of incubation at 75 µg/ml, nearly all NPCs exhibited visible SPION intake. Evidence from light microscopy, electron microscopy, chemical analysis, and magnetic resonance imaging confirmed the effectiveness of the labeling. Additionally, biological assays showed that the labeled NPCs exhibited unaffected viability, oxidative stress, apoptosis and differentiation. In the demonstrated in vivo cellular MRI experiment, the hypointensities representing the SPION labeled NPCs remained observable throughout the entire tracking period. The findings indicate that simple SPION incubation without the addition of TAs is an efficient intracellular magnetic labeling method. This simple approach may be considered as an alternative approach to the mainstream labeling method that involves the use of TAs.
Collapse
Affiliation(s)
- Chiao-Chi V. Chen
- Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan
- Functional and Micro-magnetic Resonance Imaging Center, Academic Sinica, Taipei, Taiwan
| | - Min-Chi Ku
- Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan
- Functional and Micro-magnetic Resonance Imaging Center, Academic Sinica, Taipei, Taiwan
| | - Jayaseema D. M.
- Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan
- Functional and Micro-magnetic Resonance Imaging Center, Academic Sinica, Taipei, Taiwan
| | | | - Dueng-Yuan Hueng
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
- * E-mail: (CC); (D-YH)
| | - Chen Chang
- Institute of Biomedical Sciences, Academic Sinica, Taipei, Taiwan
- Functional and Micro-magnetic Resonance Imaging Center, Academic Sinica, Taipei, Taiwan
- * E-mail: (CC); (D-YH)
| |
Collapse
|
31
|
Schumacher T, Krohn M, Hofrichter J, Lange C, Stenzel J, Steffen J, Dunkelmann T, Paarmann K, Fröhlich C, Uecker A, Plath AS, Sommer A, Brüning T, Heinze HJ, Pahnke J. ABC transporters B1, C1 and G2 differentially regulate neuroregeneration in mice. PLoS One 2012; 7:e35613. [PMID: 22545122 PMCID: PMC3335815 DOI: 10.1371/journal.pone.0035613] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 03/19/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND ATP-binding cassette (ABC) transporters are essential regulators of organismic homeostasis, and are particularly important in protecting the body from potentially harmful exogenous substances. Recently, an increasing number of in vitro observations have indicated a functional role of ABC transporters in the differentiation and maintenance of stem cells. Therefore, we sought to determine brain-related phenotypic changes in animals lacking the expression of distinct ABC transporters (ABCB1, ABCG2 or ABCC1). METHODOLOGY AND PRINCIPAL FINDINGS Analyzing adult neurogenesis in ABC transporter-deficient animals in vivo and neuronal stem/progenitor cells in vitro resulted in complex findings. In vivo, the differentiation of neuronal progenitors was hindered in ABC transporter-deficient mice (ABCB1(0/0)) as evidenced by lowered numbers of doublecortin(+) (-36%) and calretinin(+) (-37%) cells. In vitro, we confirmed that this finding is not connected to the functional loss of single neural stem/progenitor cells (NSPCs). Furthermore, assessment of activity, exploratory behavior, and anxiety levels revealed behavioral alterations in ABCB1(0/0) and ABCC1(0/0) mice, whereas ABCG2(0/0) mice were mostly unaffected. CONCLUSION AND SIGNIFICANCE Our data show that single ABC transporter-deficiency does not necessarily impair neuronal progenitor homeostasis on the single NSPC level, as suggested by previous studies. However, loss of distinct ABC transporters impacts global brain homeostasis with far ranging consequences, leading to impaired neurogenic functions in vivo and even to distinct behavioral phenotypes. In addition to the known role of ABC transporters in proteopathies such as Parkinson's disease and Alzheimer's disease, our data highlight the importance of understanding the general function of ABC transporters for the brain's homeostasis and the regeneration potential.
Collapse
Affiliation(s)
- Toni Schumacher
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
| | - Markus Krohn
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Jacqueline Hofrichter
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
| | - Cathleen Lange
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
| | - Jan Stenzel
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
| | - Johannes Steffen
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
| | - Tina Dunkelmann
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
| | - Kristin Paarmann
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
| | - Christina Fröhlich
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
| | - Annekathrin Uecker
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
| | - Anne-Sophie Plath
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Alexandra Sommer
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Thomas Brüning
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Hans-Jochen Heinze
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
| | - Jens Pahnke
- Neurodegeneration Research Laboratory (NRL), Department of Neurology, Universities of Rostock and Magdeburg, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
| |
Collapse
|
32
|
Shimizu F, Hovinga KE, Metzner M, Soulet D, Tabar V. Organotypic explant culture of glioblastoma multiforme and subsequent single-cell suspension. ACTA ACUST UNITED AC 2012; Chapter 3:Unit3.5. [PMID: 22135084 DOI: 10.1002/9780470151808.sc0305s19] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive brain tumors. GBM cell lines used in laboratory studies are frequently passaged in various culture media at high proliferation rates, resulting in significant genetic and molecular alterations. Thus, data obtained in cell lines are often inapplicable to patient tumors. Furthermore, recent studies suggest that there is a stem cell-like hierarchy among GBM cell populations and a crucial role for tumor vasculature in stem cells, as well as tumor growth, which cannot be reproduced in cell line cultures. Our laboratory has developed a novel three-dimensional (3D) organotypic "explant" system of surgical GBM specimens that preserves tumor cells in their original milieu, as well as the cytoarchitecture of the tumor stroma. Our previous study on the role of Notch inhibition has demonstrated a definitive effect on the tumor endothelium that could only be highlighted by this system. In this unit, we describe a detailed protocol for preparing GBM explants, and discuss strengths, as well as limitations of the explant system as an in vitro 3D model of GBM.
Collapse
Affiliation(s)
- Fumiko Shimizu
- Department of Neurosurgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | |
Collapse
|
33
|
Alvarez-Palazuelos LE, Robles-Cervantes MS, Castillo-Velazquez G, Rivas-Souza M, Guzman-Muniz J, Moy-Lopez N, Gonzalez-Castaneda RE, Luquin S, Gonzalez-Perez O. Regulation of neural stem cell in the human SVZ by trophic and morphogenic factors. CURRENT SIGNAL TRANSDUCTION THERAPY 2011; 6:320-326. [PMID: 22053150 PMCID: PMC3204663 DOI: 10.2174/157436211797483958] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The subventricular zone (SVZ), lining the lateral ventricular system, is the largest germinal region in mammals. In there, neural stem cells express markers related to astoglial lineage that give rise to new neurons and oligodendrocytes in vivo. In the adult human brain, in vitro evidence has also shown that astrocytic cells isolated from the SVZ can generate new neurons and oligodendrocytes. These proliferative cells are strongly controlled by a number of signals and molecules that modulate, activate or repress the cell division, renewal, proliferation and fate of neural stem cells. In this review, we summarize the cellular composition of the adult human SVZ (hSVZ) and discuss the increasing evidence showing that some trophic modulators strongly control the function of neural stem cells in the SVZ.
Collapse
Affiliation(s)
| | | | - Gabriel Castillo-Velazquez
- Department of Neurosurgery. Instituto Nacional de Neurología y Neurocirugia "Manuel Velasco Suárez" México, DF
| | - Mario Rivas-Souza
- Forensic medicine. Instituto Jalisciense de Ciencias Forenses, Guadalajara, Jalisco
| | - Jorge Guzman-Muniz
- Department of Neuroscience, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara
| | - Norma Moy-Lopez
- Department of Neuroscience, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara
| | | | - Sonia Luquin
- Department of Neuroscience, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara
| | - Oscar Gonzalez-Perez
- Department of Neuroscience, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara ; Laboratory of Neuroscience, Facultad de Psicología, Universidad de Colima, Colima, Col, México
| |
Collapse
|
34
|
Cunha C, Panseri S, Villa O, Silva D, Gelain F. 3D culture of adult mouse neural stem cells within functionalized self-assembling peptide scaffolds. Int J Nanomedicine 2011; 6:943-55. [PMID: 21720506 PMCID: PMC3124398 DOI: 10.2147/ijn.s17292] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Indexed: 12/20/2022] Open
Abstract
Three-dimensional (3D) in vitro models of cell culture aim to fill the gap between the standard two-dimensional cell studies and the in vivo environment. Especially for neural tissue regeneration approaches where there is little regenerative capacity, these models are important for mimicking the extracellular matrix in providing support, allowing the natural flow of oxygen, nutrients, and growth factors, and possibly favoring neural cell regrowth. We have previously demonstrated that a new self-assembling nanostructured biomaterial, based on matrigel, was able to support adult neural stem cell (NSC) culture. In this study, we developed a new 3D cell culture system that takes advantage of the nano- and microfiber assembling process, under physiologic conditions, of these biomaterials. The assembled scaffold forms an intricate and biologically active matrix that displays specifically designed functional motifs: RGD (Arg-Gly-Asp), BMHP1 (bone marrow homing peptide 1), and BMHP2, for the culture of adult NSCs. These scaffolds were prepared at different concentrations, and microscopic examination of the cell-embedded scaffolds showed that NSCs are viable and they proliferate and differentiate within the nanostructured environment of the scaffold. Such a model has the potential to be tailored to develop ad hoc designed peptides for specific cell lines.
Collapse
Affiliation(s)
- Carla Cunha
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| | - Silvia Panseri
- Laboratory of Biomechanics and Technology Innovation, Rizzoli Orthopaedic Institute, Bologna
- Laboratory of Nano-Biomagnetism, Institute of Science and Technology for Ceramics, National Research Council, Faenza, Italy
| | - Omar Villa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| | - Diego Silva
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| | - Fabrizio Gelain
- Department of Biotechnology and Biosciences, University of Milano-Bicocca
- Center for Nanomedicine and Tissue Engineering, CNTE – A.O. Ospedale Niguarda Ca’ Granda, Milan
| |
Collapse
|
35
|
Abstract
AbstractHuman neurospheres are free-floating spherical clusters generated from a single neural stem cell and comprising cells at different stages of maturation in the neuronal and glial lineages. Although recent findings have disproved the original idea of clonally derived neurospheres according to the paradigm of one stem cell — one neurosphere, they still represent a valid model for growing neural stem cell cultures in vitro. While the immunocytochemical approach to the identification of stem cells, progenitor cells, and mature cells has been extensively used, scant data are available about the ultrastructural arrangement of different cell types within the neurosphere. This paper provides, by means of scanning electron microscopy, some new insights into the three-dimensional assembly of human neurospheres, trying to correlate some parameters such as cell density, shape and growing strategies with the immunolocalization of some antigens such as nestin, GFAP, α-internexin and βIII-tubulin. The major findings from this study are: a) regardless of the stage of in vitro maturation, the growth of the spheres is the result of mitotic divisions producing the aspect of an irregular budding mechanism in the outermost layer look like; b) analysis of the volumetric composition of the inner core has revealed the presence of two alternative shape pattern (pyramidal vs rounded cells) possibly related to both the ongoing maturation stages and GFAP and internexin expression.
Collapse
|