1
|
Glotzbach K, Stamm N, Weberskirch R, Faissner A. Cationic Hydrogels Modulate Neural Stem and Progenitor Cell Proliferation and Differentiation Behavior in Dependence of Cationic Moiety Concentration in 2D Cell Culture. ACS Biomater Sci Eng 2024; 10:3148-3163. [PMID: 38227432 DOI: 10.1021/acsbiomaterials.3c01668] [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] [Indexed: 01/17/2024]
Abstract
The central nervous system (CNS) has a limited regenerative capacity because a hostile environment prevents tissue regeneration after damage or injury. Neural stem/progenitor cells (NSPCs) are considered a potential resource for CNS repair, which raises the issue of adequate cultivation and expansion procedures. Cationic charge supports the survival and adhesion of NSPCs. Typically, tissue culture plates with cationic coatings, such as poly-l-ornithine (PLO), have been used to culture these cell types (NSPCs). Yet presently, little is known about the impact of cationic charge concentration on the viability, proliferation, and differentiation capacity of NSPCs. Therefore, we have recently developed well-defined, fully synthetic hydrogel systems G1 (gel 1) to G6 (gel 6) that allow for the precise control of the concentration of the cationic trimethylaminoethyl acrylate (TMAEA) molecule associated with the polymer in a range from 0.06 to 0.91 μmol/mg. When murine NSPCs were cultured on these gels under differentiation conditions, we observed a strong correlation of cationic charge concentration with NSPC survival. In particular, neurons were preferentially formed on gels with a higher cationic charge concentration, whereas astrocytes and oligodendrocytes favored weakly charged or even neutral gel surfaces. To test the properties of the gels under proliferative conditions, the NSPCs were cultivated in the presence of fibroblast growth factor 2 (FGF2). The cytokine significantly increased the number of NSPCs but delayed the differentiation toward neurons and glia cells. Thus, the hydrogels are compatible with the survival, expansion, and differentiation of NSPCs and may be useful to create supportive environments in transplantation approaches.
Collapse
Affiliation(s)
- Kristin Glotzbach
- Department of Cell Morphology and Molecular Neurobiology, Ruhr Universität Bochum, Bochum 44801, Germany
| | - Nils Stamm
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund 44227, Germany
| | - Ralf Weberskirch
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund 44227, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr Universität Bochum, Bochum 44801, Germany
| |
Collapse
|
2
|
Romariz SAA, Sanabria V, da Silva KR, Quintella ML, de Melo BAG, Porcionatto M, de Almeida DC, Longo BM. High Concentrations of Cannabidiol Induce Neurotoxicity in Neurosphere Culture System. Neurotox Res 2024; 42:14. [PMID: 38349488 DOI: 10.1007/s12640-024-00692-5] [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: 08/07/2023] [Revised: 12/30/2023] [Accepted: 01/30/2024] [Indexed: 02/15/2024]
Abstract
Recent studies have demonstrated that cannabinoids are potentially effective in the treatment of various neurological conditions, and cannabidiol (CBD), one of the most studied compounds, has been proposed as a non-toxic option. However, the adverse effects of CBD on neurodevelopmental processes have rarely been studied in cell culture systems. To better understand CBD's influence on neurodevelopment, we exposed neural progenitor cells (NPCs) to different concentrations of CBD (1 µM, 5 µM, and 10 µM). We assessed the morphology, migration, differentiation, cell death, and gene expression in 2D and 3D bioprinted models to stimulate physiological conditions more effectively. Our results showed that CBD was more toxic at higher concentrations (5 µM and 10 µM) and affected the viability of NPCs than at lower concentrations (1 µM), in both 2D and 3D models. Moreover, our study revealed that higher concentrations of CBD drastically reduced the size of neurospheres and the number of NPCs within neurospheres, impaired the morphology and mobility of neurons and astrocytes after differentiation, and reduced neurite sprouting. Interestingly, we also found that CBD alters cellular metabolism by influencing the expression of glycolytic and β-oxidative enzymes in the early and late stages of metabolic pathways. Therefore, our study demonstrated that higher concentrations of CBD promote important changes in cellular functions that are crucial during CNS development.
Collapse
Affiliation(s)
- Simone A A Romariz
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Viviam Sanabria
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Karina Ribeiro da Silva
- Department of Medicine, Nephrology Division, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Miguel L Quintella
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Bruna A G de Melo
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Marimélia Porcionatto
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Danilo Candido de Almeida
- Department of Medicine, Nephrology Division, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Beatriz M Longo
- Department of Physiology, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
3
|
Ling Q, Liang JJ, Chen S, Chen CB, Ng TK, Huang Y. Continuous non-adherent culture promotes transdifferentiation of human adipose-derived stem cells into retinal lineage. Open Life Sci 2023; 18:20220760. [PMID: 38027227 PMCID: PMC10668113 DOI: 10.1515/biol-2022-0760] [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: 06/16/2023] [Revised: 09/12/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Non-adherent culture is critical for the transdifferentiation of stem cells from mesoderm to neuroectoderm. Sphere culture has been reported to directly induce the adipose tissue cells to neural stem cells. Here we aimed to evaluate continuous non-adherent culture on the transdifferentiation potential of human adipose-derived stem cells (ASCs) into retinal lineage. Human ASCs were induced into retinal lineage by the treatment of noggin, dickkopf-related protein 1, and IGF-1 (NDI) under adherent and non-adherent culture. The NDI induction treatment with the adherent culture for 21 days promoted robust expression of retinal markers in the induced ASCs as compared to those without NDI induction on the adherent culture. With continuous non-adherent culture for 21 days, human ASCs could highly express retinal marker genes even without NDI induction treatment as compared to those on the adherent culture. The combination of continuous non-adherent culture with the NDI induction did not show a significant upregulation of the retinal marker expression as compared to either NDI induction with the adherent culture or continuous non-adherent culture without NDI induction treatment. In summary, both non-adherent culture and NDI induction medium could independently promote the transdifferentiation of human ASCs into retinal lineage. Yet, their combination did not produce an enhancement effect.
Collapse
Affiliation(s)
- Qiying Ling
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, North Dongxia Road, Shantou, Guangdong, China
| | - Jia-Jian Liang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, North Dongxia Road, Shantou, Guangdong, China
| | - Shaowan Chen
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, North Dongxia Road, Shantou, Guangdong, China
| | - Chong-Bo Chen
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, North Dongxia Road, Shantou, Guangdong, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, North Dongxia Road, Shantou, Guangdong, China
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuqiang Huang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, North Dongxia Road, Shantou, Guangdong, China
| |
Collapse
|
4
|
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
|
5
|
Campos HC, Ribeiro DE, Hashiguchi D, Hukuda DY, Gimenes C, Romariz SAA, Ye Q, Tang Y, Ulrich H, Longo BM. Distinct Effects of the Hippocampal Transplantation of Neural and Mesenchymal Stem Cells in a Transgenic Model of Alzheimer's Disease. Stem Cell Rev Rep 2022; 18:781-791. [PMID: 34997526 DOI: 10.1007/s12015-021-10321-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a severe disabling condition with no cure currently available, which accounts for 60-70% of all dementia cases worldwide. Therefore, the investigation of possible therapeutic strategies for AD is necessary. To this end, animal models corresponding to the main aspects of AD in humans have been widely used. Similar to AD patients, the double transgenic APPswe/PS1dE9 (APP/PS1) mice show cognitive deficits, hyperlocomotion, amyloid-β (Αβ) plaques in the cortex and hippocampus, and exacerbated inflammatory responses. Recent studies have shown that these neuropathological features could be reversed by stem cell transplantation. However, the effects induced by neural (NSC) and mesenchymal (MSC) stem cells has never been compared in an AD animal model. Therefore, the present study aimed to investigate whether transplantation of NSC or MSC into the hippocampus of APP/PS1 mice reverses AD-induced pathological alterations, evaluated by the locomotor activity (open field test), short- and long-term memory (object recognition) tests, Αβ plaques (6-E10), microglia distribution (Iba-1), M1 (iNOS) and M2 (ARG-1) microglial phenotype frequencies. NSC and MSC engraftment reduced the number of Αβ plaques and produced an increase in M2 microglia polarization in the hippocampus of APP/PS1 mice, suggesting an anti-inflammatory effect of stem cell transplantation. NSC also reversed the hyperlocomotor activity and increased the number of microglia in the hippocampus of APP/PS1 mice. No impairment of short or long-term memory was observed in APP/PS1 mice. Overall, this study highlights the potential beneficial effects of transplanting NSC or MSC for AD treatment.
Collapse
Affiliation(s)
- Henrique C Campos
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Deidiane Elisa Ribeiro
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil
| | - Debora Hashiguchi
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil.,Laboratório de Plasticidade Sináptica, Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, 59078-900, Caixa Postal: 1524, Brazil
| | - Deborah Y Hukuda
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Christiane Gimenes
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Simone A A Romariz
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Qing Ye
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil.,International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China
| | - Yong Tang
- International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, 610075, China
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, Brazil.,International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Beatriz Monteiro Longo
- Laboratório de Neurofisiologia, Depto. Fisiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
6
|
Neurospheres obtained from the ciliary margin of the chicken eye possess positional values and retinal ganglion cells differentiated from them respond to EphA/ephrin-A system. Exp Eye Res 2022; 217:108965. [DOI: 10.1016/j.exer.2022.108965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 12/14/2021] [Accepted: 01/25/2022] [Indexed: 11/23/2022]
|
7
|
Fernandez-Muñoz B, Garcia-Delgado AB, Arribas-Arribas B, Sanchez-Pernaute R. Human Neural Stem Cells for Cell-Based Medicinal Products. Cells 2021; 10:2377. [PMID: 34572024 PMCID: PMC8469920 DOI: 10.3390/cells10092377] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells represent an attractive tool for the development of regenerative therapies and are being tested in clinical trials for several neurological disorders. Human neural stem cells can be isolated from the central nervous system or can be derived in vitro from pluripotent stem cells. Embryonic sources are ethically controversial and other sources are less well characterized and/or inefficient. Recently, isolation of NSC from the cerebrospinal fluid of patients with spina bifida and with intracerebroventricular hemorrhage has been reported. Direct reprogramming may become another alternative if genetic and phenotypic stability of the reprogrammed cells is ensured. Here, we discuss the advantages and disadvantages of available sources of neural stem cells for the production of cell-based therapies for clinical applications. We review available safety and efficacy clinical data and discuss scalability and quality control considerations for manufacturing clinical grade cell products for successful clinical application.
Collapse
Affiliation(s)
- Beatriz Fernandez-Muñoz
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Ana Belen Garcia-Delgado
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Blanca Arribas-Arribas
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Sevilla, 41012 Sevilla, Spain
| | - Rosario Sanchez-Pernaute
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| |
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
|
Zhang Z, Jhaveri D, Sharmin S, Harvey TJ, Dawson PA, Piper M, Simmons DG. Cell-extrinsic requirement for sulfate in regulating hippocampal neurogenesis. Biol Open 2020; 9:bio053132. [PMID: 32661132 PMCID: PMC7406315 DOI: 10.1242/bio.053132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/01/2020] [Indexed: 01/24/2023] Open
Abstract
Sulfate is a key anion required for a range of physiological functions within the brain. These include sulfonation of extracellular proteoglycans to facilitate local growth factor binding and to regulate the shape of morphogen gradients during development. We have previously shown that mice lacking one allele of the sulfate transporter Slc13a4 exhibit reduced sulfate transport into the brain, deficits in social behaviour, reduced performance in learning and memory tasks, and abnormal neurogenesis within the ventricular/subventricular zone lining the lateral ventricles. However, whether these mice have deficits in hippocampal neurogenesis was not addressed. Here, we demonstrate that adult Slc13a4+/- mice have increased neurogenesis within the subgranular zone (SGZ) of the hippocampal dentate gyrus, with elevated numbers of neural progenitor cells and intermediate progenitors. In contrast, by 12 months of age there were reduced numbers of neural stem cells in the SGZ of heterozygous mice. Importantly, we did not observe any changes in proliferation when we isolated and cultured progenitors in vitro in neurosphere assays, suggestive of a cell-extrinsic requirement for sulfate in regulating hippocampal neurogenesis. Collectively, these data demonstrate a requirement for sulfate transport during postnatal brain development to ensure normal adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Zhe Zhang
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Dhanisha Jhaveri
- Mater Research Institute, The University of Queensland, TRI Building, Woolloongabba, Brisbane, 4102, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia
| | - Sazia Sharmin
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Tracey J Harvey
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Paul A Dawson
- Mater Research Institute, The University of Queensland, TRI Building, Woolloongabba, Brisbane, 4102, Australia
| | - Michael Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia
| | - David G Simmons
- School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| |
Collapse
|
10
|
Human Dental Pulp Stem Cells and Gingival Mesenchymal Stem Cells Display Action Potential Capacity In Vitro after Neuronogenic Differentiation. Stem Cell Rev Rep 2020; 15:67-81. [PMID: 30324358 DOI: 10.1007/s12015-018-9854-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The potential of human mesenchymal stromal/stem cells (MSCs) including oral stem cells (OSCs) as a cell source to derive functional neurons has been inconclusive. Here we tested a number of human OSCs for their neurogenic potential compared to non-OSCs and employed various neurogenic induction methods. OSCs including dental pulp stem cells (DPSCs), gingiva-derived mesenchymal stem cells (GMSCs), stem cells from apical papilla and non-OSCs including bone marrow MSCs (BMMSCs), foreskin fibroblasts and dermal fibroblasts using non-neurosphere-mediated or neurosphere-mediated methods to guide them toward neuronal lineages. Cells were subjected to RT-qPCR, immunocytofluorescence to detect the expression of neurogenic genes or electrophysiological analysis at final stage of maturation. We found that induced DPSCs and GMSCs overall appeared to be more neurogenic compared to other cells either morphologically or levels of neurogenic gene expression. Nonetheless, of all the neural induction methods employed, only one neurosphere-mediated method yielded electrophysiological properties of functional neurons. Under this method, cells expressed increased neural stem cell markers, nestin and SOX1, in the first phase of differentiation. Neuronal-like cells expressed βIII-tubulin, CNPase, GFAP, MAP-2, NFM, pan-Nav, GAD67, Nav1.6, NF1, NSE, PSD95, and synapsin after the second phase of differentiation to maturity. Electrophysiological experiments revealed that 8.3% of DPSC-derived neuronal cells and 21.2% of GMSC-derived neuronal cells displayed action potential, although no spontaneous excitatory/inhibitory postsynaptic action potential was observed. We conclude that DPSCs and GMSCs have the potential to become neuronal cells in vitro, therefore, these cells may be used as a source for neural regeneration.
Collapse
|
11
|
Zhang L, Ye M, Zhu L, Cha J, Li C, Yao YG, Mao B. Loss of ZC4H2 and RNF220 Inhibits Neural Stem Cell Proliferation and Promotes Neuronal Differentiation. Cells 2020; 9:cells9071600. [PMID: 32630355 PMCID: PMC7408363 DOI: 10.3390/cells9071600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/22/2020] [Accepted: 06/29/2020] [Indexed: 12/31/2022] Open
Abstract
The ubiquitin E3 ligase RNF220 and its co-factor ZC4H2 are required for multiple neural developmental processes through different targets, including spinal cord patterning and the development of the cerebellum and the locus coeruleus. Here, we explored the effects of loss of ZC4H2 and RNF220 on the proliferation and differentiation of neural stem cells (NSCs) derived from mouse embryonic cortex. We showed that loss of either ZC4H2 or RNF220 inhibits the proliferation and promotes the differentiation abilities of NSCs in vitro. RNA-Seq profiling revealed 132 and 433 differentially expressed genes in the ZC4H2−/− and RNF220−/− NSCs, compared to wild type (WT) NSCs, respectively. Specifically, Cend1, a key regulator of cell cycle exit and differentiation of neuronal precursors, was found to be upregulated in both ZC4H2−/− and RNF220−/− NSCs at the mRNA and protein levels. The targets of Cend1, such as CyclinD1, Notch1 and Hes1, were downregulated both in ZC4H2−/− and RNF220−/− NSCs, whereas p53 and p21 were elevated. ZC4H2−/− and RNF220−/− NSCs showed G0/G1 phase arrest compared to WT NSCs in cell cycle analysis. These results suggested that ZC4H2 and RNF220 are likely involved in the regulation of neural stem cell proliferation and differentiation through Cend1.
Collapse
Affiliation(s)
- Longlong Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (L.Z.); (L.Z.); (J.C.); (C.L.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China; (M.Y.); (Y.-G.Y.)
| | - Maosen Ye
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China; (M.Y.); (Y.-G.Y.)
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, and KIZ – CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Liang Zhu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (L.Z.); (L.Z.); (J.C.); (C.L.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China; (M.Y.); (Y.-G.Y.)
| | - Jingmei Cha
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (L.Z.); (L.Z.); (J.C.); (C.L.)
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China; (M.Y.); (Y.-G.Y.)
| | - Chaocui Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (L.Z.); (L.Z.); (J.C.); (C.L.)
| | - Yong-Gang Yao
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, China; (M.Y.); (Y.-G.Y.)
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, and KIZ – CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bingyu Mao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (L.Z.); (L.Z.); (J.C.); (C.L.)
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
- Correspondence: ; Tel.: +86-871-68125418
| |
Collapse
|
12
|
Carroll JA, Groveman BR, Williams K, Moore R, Race B, Haigh CL. Prion protein N1 cleavage peptides stimulate microglial interaction with surrounding cells. Sci Rep 2020; 10:6654. [PMID: 32313035 PMCID: PMC7171115 DOI: 10.1038/s41598-020-63472-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/29/2020] [Indexed: 01/06/2023] Open
Abstract
Microglia act as the protective immune cell of the brain. By surveying the tissue to identify and rectify problems, they function to maintain the health of brain cells. The prion protein N-terminal cleavage fragment, N1, has demonstrated neuroprotective activities in vitro and in vivo. This study aimed to elucidate whether N1 could modulate microglial function and, if so, determine the consequences for the surrounding tissue. Using a mixed neuronal lineage and microglia co-culture system, we showed that N1 stimulation changed overall morphology and metabolism, suggesting enhanced cellular viability. Furthermore, N1 induced an increase in Cxcl10 secretion in the co-cultures. Recombinant Cxcl10, administered exogenously, mediated the changes in the mixed neuronal lineage culture morphology and metabolism in the absence of microglia, but no effect of Cxcl10 was observed on microglia cultured on their own. Direct cell-to-cell contact was required for N1 to influence microglia in the co-cultures, and this was linked with restructuring of microglial membrane composition to include a higher GM1 content at interaction sites with surrounding cells. Our findings show that N1 can play a regulatory role in microglial function in the context of an inter-connected network of cells by changing both cellular interaction sites and cytokine secretion.
Collapse
Affiliation(s)
- J A Carroll
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - B R Groveman
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - K Williams
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - R Moore
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - B Race
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA
| | - C L Haigh
- Laboratory of Persistent Viral Diseases, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, 903 South 4th Street, Hamilton, MT, 59840, USA.
| |
Collapse
|
13
|
O'Grady BJ, Lippmann ES. Recent Advancements in Engineering Strategies for Manipulating Neural Stem Cell Behavior. ACTA ACUST UNITED AC 2020; 1:41-47. [PMID: 33748772 DOI: 10.1007/s43152-020-00003-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Purpose of Review Stem cells are exquisitely sensitive to biophysical and biochemical cues within the native microenvironment. This review focuses on emerging strategies to manipulate neural cell behavior using these influences in three-dimensional (3D) culture systems. Recent Findings Traditional systems for neural cell differentiation typically produce heterogeneous populations with limited diversity rather than the complex, organized tissue structures observed in vivo. Advancements in developing engineering tools to direct neural cell fates can enable new applications in basic research, disease modeling, and regenerative medicine. Summary This review article highlights engineering strategies that facilitate controlled presentation of biophysical and biochemical cues to guide differentiation and impart desired phenotypes on neural cell populations. Specific highlighted examples include engineered biomaterials and microfluidic platforms for spatiotemporal control over the presentation of morphogen gradients.
Collapse
Affiliation(s)
- Brian J O'Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
14
|
Nesan D, Thornton HF, Sewell LC, Kurrasch DM. An Efficient Method for Generating Murine Hypothalamic Neurospheres for the Study of Regional Neural Progenitor Biology. Endocrinology 2020; 161:5802442. [PMID: 32154873 PMCID: PMC7105385 DOI: 10.1210/endocr/bqaa035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 02/25/2020] [Accepted: 03/02/2020] [Indexed: 12/18/2022]
Abstract
The hypothalamus is a key homeostatic brain region and the primary effector of neuroendocrine signaling. Recent studies show that early embryonic developmental disruption of this region can lead to neuroendocrine conditions later in life, suggesting that hypothalamic progenitors might be sensitive to exogenous challenges. To study the behavior of hypothalamic neural progenitors, we developed a novel dissection methodology to isolate murine hypothalamic neural stem and progenitor cells at the early timepoint of embryonic day 12.5, which coincides with peak hypothalamic neurogenesis. Additionally, we established and optimized a culturing protocol to maintain multipotent hypothalamic neurospheres that are capable of sustained proliferation or differentiation into neurons, oligodendrocytes, and astrocytes. We characterized media requirements, appropriate cell seeding density, and the role of growth factors and sonic hedgehog (Shh) supplementation. Finally, we validated the use of fluorescence activated cell sorting of either Sox2GFPKI or Nkx2.1GFPKI transgenic mice as an alternate cellular isolation approach to enable enriched selection of hypothalamic progenitors for growth into neurospheres. Combined, we present a new technique that yields reliable culturing of hypothalamic neural stem and progenitor cells that can be used to study hypothalamic development in a controlled environment.
Collapse
Affiliation(s)
- Dinushan Nesan
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Hayley F Thornton
- Department of Neuroscience, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Laronna C Sewell
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Department of Neuroscience, University of Calgary, Calgary, Alberta, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Correspondence: Deborah M Kurrasch, Department of Medical Genetics, University of Calgary, 3330 Hospital Drive NW, HSC 2215, Calgary, AB T2N 4N1. E-mail:
| |
Collapse
|
15
|
Zhang L, Yu H, Yuan Y, Yu JS, Lou Z, Xue Y, Liu Y. The necessity for standardization of glioma stem cell culture: a systematic review. Stem Cell Res Ther 2020; 11:84. [PMID: 32102678 PMCID: PMC7045630 DOI: 10.1186/s13287-020-01589-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/15/2019] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The cancer stem cell hypothesis is an old idea which has been revived in recent years for many cancers, including gliomas. However, this concept has become controversial due to a series of studies with conflicting results. METHODS A systematic literature search was conducted in PubMed and the Web of Science database to analyze studies using serum-free medium and its components in glioma stem cells, glioma stem-like cells, glioma-initiating cells, or glioma neurosphere cultures. All the studies reviewed were published between 1970 and 2019. We found that no standardized culture method was used, and the data were incomparable due to differing culture conditions and the use of media with different components. CONCLUSIONS Here, we review the most commonly used serum-free media and added components for glioma stem cell culture while highlighting the function of each component used in the media. We emphasize the necessity for standardization of glioma stem cell culture and propose a standard culture medium to prevent bias in glioma stem cell research.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China.,Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, USA.,Department of Oncology, Mayo Clinic, Rochester, USA
| | - Hongwei Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China
| | - Yuhui Yuan
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China
| | - John S Yu
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, Rochester, USA
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, # 36 Sanhao Street, Heping District, Shenyang, China.
| |
Collapse
|
16
|
Yang S, Cao Z, Zhu J, Zhang Z, Zhao H, Zhao L, Sun X, Wang X. In Vitro Monolayer Culture of Dispersed Neural Stem Cells on the E-Cadherin-Based Substrate with Long-Term Stemness Maintenance. ACS OMEGA 2019; 4:18136-18146. [PMID: 31720516 PMCID: PMC6843705 DOI: 10.1021/acsomega.9b02053] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/13/2019] [Indexed: 05/08/2023]
Abstract
Neural stem cells (NSCs) play an important role in neural tissue engineering because of their capacity of self-renewal and differentiation to multiple cell lineages. The in vitro conventional neurosphere culture protocol has some limitations such as limited nutrition and oxygen penetration and distribution causing the heterogeneity of cells inside, inaccessibility of internal cells, and inhomogeneous cellular morphology and properties. As a result, cultivation as a monolayer is a better way to study NSCs and obtain a homogeneous cell population. The cadherins are a classical family of homophilic cell adhesion molecules mediating cell-cell adhesion. Here, we used a recombinant human E-cadherin mouse IgG Fc chimera protein that self-assembles on a hydrophobic polystyrene surface via hydrophobic interaction to obtain an E-cadherin-coated culture plate (ECP). The rat fetal NSCs were cultured on the ECP and routine tissue culture plate (TCP) from passage 2 to passage 5. NSCs on TCP formed uniform floating neurospheres and grew up over time, while cells on the ECP adhered on the bottom of the plate and exhibited individual cells with scattering morphology, forming intercellular connections between cells. The cell proliferation and differentiation behaviors that were evaluated by Cell Counting Kit-8 assay (CCK-8), immunofluorescence staining, and real-time quantitative polymerase chain reaction showed NSCs could maintain the capacity for self-renewal and ability to differentiate into neurons, oligodendrocytes, and astrocytes after the long-term in vitro cell culture and passaging. Therefore, our study indicated that hE-cad-Fc could provide a homogeneous environment for individual cells in monolayer conditions to maintain the capacity of self-renewal and differentiation by mimicking the cell-cell interaction.
Collapse
Affiliation(s)
- Shuhui Yang
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Zheng Cao
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Jinjin Zhu
- Department
of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College
of Zhejiang University, Sir Run Run Shaw
Institute of Clinical Medicine of Zhejiang University, 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, China
| | - Zhe Zhang
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - He Zhao
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Lingyun Zhao
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaodan Sun
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiumei Wang
- State
Key Laboratory of New Ceramics and Fine Processing, Key Laboratory
of Advanced Materials of Ministry of Education, School of Materials
Science and Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
17
|
da Silva BR, Santos LE, de Melo Reis RA, de Mello FG, Ribeiro-Resende VT. Müller Cells Derived from Adult Chicken and Mouse Retina Neurospheres Acquire the Dopaminergic Phenotype. Cell Mol Neurobiol 2019; 39:99-109. [PMID: 30430378 DOI: 10.1007/s10571-018-0636-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/08/2018] [Indexed: 10/27/2022]
Abstract
Neurospheres prepared from multipotent progenitors in the retina obtained from postnatal mice differentiate into neurons and Müller glia (De Melo Reis et al., in Cell Mol Neurobiol 31:835-846, 2011). Here, we investigated whether neurospheres prepared from adult chickens (ciliary marginal zone, CMZ) or (ciliary body) retina could also lead to differentiated neurons and glia. Neurospheres were prepared from post-hatched chickens or from adult mice after 7 days in the presence of mitogenic factors (FGFb, insulin, and EGF), generating neurons and glial cells. In addition, Müller (2M6 or glutamine synthetase positive cells) derived from post-hatch chicken CMZ neurospheres displayed the dopaminergic phenotype. Furthermore, we observed that Müller cells derived from adult chickens and mice retina neurospheres released significant amounts of dopamine as well as of its metabolites. Taken together, our data lead us to conclude that as for embryonic (chick) or newborn (mouse), the dopaminergic phenotype is a default condition of Müller glial cells obtained from neurospheres prepared from mature retina. Our data raise the possibility that Müller cells from differentiated tissue could be used to ameliorate neurodegenerative diseases involving dopaminergic dysfunction as in Parkinson's disease as shown previously (Stutz et al., in J Neurochem 128:829-840, 2014).
Collapse
Affiliation(s)
- Bárbara Rangel da Silva
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Sala, C1-27, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Luis Eduardo Santos
- Laboratório de Doenças Neurodegenerativas, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, CCS, Sala C1-31, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Ricardo A de Melo Reis
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Sala, C1-27, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Fernando Garcia de Mello
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Sala, C1-27, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Victor T Ribeiro-Resende
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Sala, C1-27, Rio de Janeiro, RJ, 21941-902, Brazil.
- Núcleo Multidisciplinar de Pesquisa em Biologia (Numpex-Bio), Campus de Duque de Caxias Geraldo Guerra Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias, RJ, 25255-030, Brazil.
| |
Collapse
|
18
|
Neural stem cells deriving from chick embryonic hindbrain recapitulate hindbrain development in culture. Sci Rep 2018; 8:13920. [PMID: 30224755 PMCID: PMC6141497 DOI: 10.1038/s41598-018-32203-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 09/03/2018] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells (NSCs) are self-renewing multipotent cells that line the neural-tube and generate all the nervous system. Understanding NSC biology is fundamental for neurodevelopmental research and therapy. Many studies emphasized the need to culture NSCs, which are typically purified from mammalian embryonic/adult brains. These sources are somewhat limited in terms of quantity, availability and animal ethical guidelines. Therefore, new sources are needed. The chick is a powerful system for experimental embryology which contributed enormously to neurodevelopmental concepts. Its accessibility, genetic/molecular manipulations, and homology to other vertebrates, makes it valuable for developmental biology research. Recently, we identified a population of NSCs in the chick hindbrain. It resides in rhombomere-boundaries, expresses Sox2 and generates progenitors and neurons. Here, we investigated whether these cells can recapitulate hindbrain development in culture. By developing approaches to propagate and image cells, manipulate their growth-conditions and separate them into subpopulations, we demonstrate the ordered formation of multipotent and self-renewing neurospheres that maintain regional identity and display differential stem/differentiation/proliferation properties. Live imaging revealed new cellular dynamics in the culture. Collectively, these NSC cultures reproduce major aspects of hindbrain development in-vitro, proposing the chick as a model for culturing hindbrain-NSCs that can be directly applied to other neural-tube domains and species.
Collapse
|
19
|
Lakshman N, Xu W, Morshead CM. A Neurosphere Assay to Evaluate Endogenous Neural Stem Cell Activation in a Mouse Model of Minimal Spinal Cord Injury. J Vis Exp 2018. [PMID: 30272658 DOI: 10.3791/57727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Neural stem cells (NSCs) in the adult mammalian spinal cord are a relatively mitotically quiescent population of periventricular cells that can be studied in vitro using the neurosphere assay. This colony-forming assay is a powerful tool to study the response of NSCs to exogenous factors in a dish; however, this can also be used to study the effect of in vivo manipulations with the proper understanding of the strengths and limitations of the assay. One manipulation of the clinical interest is the effect of injury on endogenous NSC activation. Current models of spinal cord injury provide a challenge to study this as the severity of common contusion, compression, and transection models cause the destruction of the NSC niche at the site of the injury where the stem cells reside. Here, we describe a minimal injury model that causes localized damage at the superficial dorsolateral surface of the lower thoracic level (T7/8) of the adult mouse spinal cord. This injury model spares the central canal at the level of injury and permits analysis of the NSCs that reside at the level of the lesion at various time points following injury. Here, we show how the neurosphere assay can be utilized to study the activation of the two distinct, lineally-related, populations of NSCs that reside in the spinal cord periventricular region - primitive and definitive NSCs (pNSCs and dNSCs, respectively). We demonstrate how to isolate and culture these NSCs from the periventricular region at the level of injury and the white matter injury site. Our post-surgical spinal cord dissections show increased numbers of pNSC and dNSC-derived neurospheres from the periventricular region of injured cords compared to controls, speaking to their activation via injury. Furthermore, following injury, dNSC-derived neurospheres can be isolated from the injury site - demonstrating the ability of NSCs to migrate from their periventricular niche to sites of injury.
Collapse
Affiliation(s)
- Nishanth Lakshman
- Institute of Medical Science, University of Toronto; Department of Surgery, University of Toronto
| | - Wenjun Xu
- Department of Surgery, University of Toronto
| | - Cindi M Morshead
- Institute of Medical Science, University of Toronto; Department of Surgery, University of Toronto; Institute of Biomaterials and Biomedical Engineering, University of Toronto;
| |
Collapse
|
20
|
Hwang DW, Jaganathan A, Shrestha P, Jin Y, El-Amine N, Wang SH, Hammell M, Mills AA. Chromatin-mediated translational control is essential for neural cell fate specification. Life Sci Alliance 2018; 1:e201700016. [PMID: 30456361 PMCID: PMC6238594 DOI: 10.26508/lsa.201700016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/28/2022] Open
Abstract
Neural cell fate specification is a multistep process in which stem cells undergo sequential changes in states, giving rise to particular lineages such as neurons and astrocytes. This process is accompanied by dynamic changes of chromatin and in transcription, thereby orchestrating lineage-specific gene expression programs. A pressing question is how these events are interconnected to sculpt cell fate. We show that altered chromatin due to loss of the chromatin remodeler Chd5 causes neural stem cell activation to occur ahead of time. This premature activation is accompanied by transcriptional derepression of ribosomal subunits, enhanced ribosome biogenesis, and increased translation. These untimely events deregulate cell fate decisions, culminating in the generation of excessive numbers of astrocytes at the expense of neurons. By monitoring the proneural factor Mash1, we further show that translational control is crucial for appropriate execution of cell fate specification, thereby providing new insight into the interplay between transcription and translation at the initial stages of neurogenesis.
Collapse
Affiliation(s)
- Dong-Woo Hwang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.,Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, USA
| | | | - Padmina Shrestha
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.,Molecular and Cellular Biology Program, Stony Brook University, Stony Brook, NY, USA
| | - Ying Jin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Nour El-Amine
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Sidney H Wang
- Center for Human Genetics, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Molly Hammell
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Alea A Mills
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| |
Collapse
|
21
|
Tse MK, Hung TS, Chan CM, Wong T, Dorothea M, Leclerc C, Moreau M, Miller AL, Webb SE. Identification of Ca 2+ signaling components in neural stem/progenitor cells during differentiation into neurons and glia in intact and dissociated zebrafish neurospheres. SCIENCE CHINA-LIFE SCIENCES 2018; 61:1352-1368. [PMID: 29931586 DOI: 10.1007/s11427-018-9315-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/03/2018] [Indexed: 01/30/2023]
Abstract
The development of the CNS in vertebrate embryos involves the generation of different sub-types of neurons and glia in a complex but highly-ordered spatio-temporal manner. Zebrafish are commonly used for exploring the development, plasticity and regeneration of the CNS, and the recent development of reliable protocols for isolating and culturing neural stem/progenitor cells (NSCs/NPCs) from the brain of adult fish now enables the exploration of mechanisms underlying the induction/specification/differentiation of these cells. Here, we refined a protocol to generate proliferating and differentiating neurospheres from the entire brain of adult zebrafish. We demonstrated via RT-qPCR that some isoforms of ip3r, ryr and stim are upregulated/downregulated significantly in differentiating neurospheres, and via immunolabelling that 1,4,5-inositol trisphosphate receptor (IP3R) type-1 and ryanodine receptor (RyR) type-2 are differentially expressed in cells with neuron- or radial glial-like properties. Furthermore, ATP but not caffeine (IP3R and RyR agonists, respectively), induced the generation of Ca2+ transients in cells exhibiting neuron- or glial-like morphology. These results indicate the differential expression of components of the Ca2+-signaling toolkit in proliferating and differentiating cells. Thus, given the complexity of the intact vertebrate brain, neurospheres might be a useful system for exploring neurodegenerative disease diagnosis protocols and drug development using Ca2+ signaling as a read-out.
Collapse
Affiliation(s)
- Man Kit Tse
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, China
| | - Ting Shing Hung
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, China
| | - Ching Man Chan
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, China
| | - Tiffany Wong
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, China
| | - Mike Dorothea
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, China
| | - Catherine Leclerc
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, F-31062, France
| | - Marc Moreau
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, F-31062, France
| | - Andrew L Miller
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, China
| | - Sarah E Webb
- Division of Life Science & State Key Laboratory of Molecular Neuroscience, HKUST, Clear Water Bay, Hong Kong, China.
| |
Collapse
|
22
|
Stamegna JC, Sadelli K, Escoffier G, Girard SD, Veron AD, Bonnet A, Khrestchatisky M, Gauthier P, Roman FS. Grafts of Olfactory Stem Cells Restore Breathing and Motor Functions after Rat Spinal Cord Injury. J Neurotrauma 2018; 35:1765-1780. [PMID: 29357739 DOI: 10.1089/neu.2017.5383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The transplantation of olfactory ecto-mesenchymal stem cells (OEMSCs) could be a helpful therapeutic strategy for spinal cord repair. Using an acute rat model of high cervical contusion that provokes a persistent hemidiaphragmatic and foreleg paralysis, we evaluated the therapeutic effect of a delayed syngeneic transplantation (two days post-contusion) of OEMSCs within the injured spinal cord. Respiratory function was assessed using diaphragmatic electromyography and neuroelectrophysiological recordings of phrenic nerves (innervating the diaphragm). Locomotor function was evaluated using the ladder-walking locomotor test. Cellular reorganization in the injured area was also studied using immunohistochemical and microscopic techniques. We report a substantial improvement in breathing movements, in activities of the ipsilateral phrenic nerve and ipsilateral diaphragm, and also in locomotor abilities four months post-transplantation with nasal OEMSCs. Moreover, in the grafted spinal cord, axonal disorganization and inflammation were reduced. Some grafted stem cells adopted a neuronal phenotype, and axonal sparing was observed in the injury site. The therapeutic effect on the supraspinal command is presumably because of both neuronal replacements and beneficial paracrine effects on the injury area. Our study provides evidence that nasal OEMSCs could be a first step in clinical application, particularly in patients with reduced breathing/locomotor movements.
Collapse
Affiliation(s)
- Jean-Claude Stamegna
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| | - Kevin Sadelli
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| | - Guy Escoffier
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| | - Stéphane D Girard
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| | - Antoine D Veron
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France .,2 IRSEA, Research Institute in Semiochemistry and Applied Ethology , Apt, France
| | - Amandine Bonnet
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| | | | - Patrick Gauthier
- 3 Laboratoire de Neurosciences et Cognitives, Aix-Marseille Université , Marseille, France
| | - François S Roman
- 1 Institut de Neurophysiopathologie, Aix-Marseille Université , Marseille, France
| |
Collapse
|
23
|
Pauly MG, Krajka V, Stengel F, Seibler P, Klein C, Capetian P. Adherent vs. Free-Floating Neural Induction by Dual SMAD Inhibition for Neurosphere Cultures Derived from Human Induced Pluripotent Stem Cells. Front Cell Dev Biol 2018; 6:3. [PMID: 29468156 PMCID: PMC5807902 DOI: 10.3389/fcell.2018.00003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/19/2018] [Indexed: 01/16/2023] Open
Abstract
Keeping neural stem cells under proliferation, followed by terminal differentiation, can substantially increase the number of neurons generated. With regard to the usability of proliferating neurospheres (NSPHs) cultures, adherent induction protocols have not yet been studied in comparison to embryoid body (EB)-based protocols. To compare these proctocols, neural induction of human induced pluripotent stem cells was performed by dual SMAD inhibition under both adherent and free-floating EB culture conditions. After 10 days, we transferred cells to low-attachment culture plates and proliferated them as free-floating neurospheres. RNA was collected, transcribed to cDNA and analyzed for sonic hedgehog expression that plays an important role during proliferation process. NSPHs were analyzed by immunofluorescence imaging directly and upon continued differentiation. The EB-based approach yielded in higher numbers of cells expressing the neural stem cell marker Nestin, and showed in contrast to the adherent induction protocol increased expression levels of sonic hedgehog. Although improvements to culture consistency and reliability are desirable, the EB-based protocol appears to be superior to the adherent protocol for both, the proliferation and differentiation capacity.
Collapse
Affiliation(s)
- Martje G Pauly
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Victor Krajka
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Felix Stengel
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Philipp Capetian
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
24
|
Harrill JA. Human-Derived Neurons and Neural Progenitor Cells in High Content Imaging Applications. Methods Mol Biol 2018; 1683:305-338. [PMID: 29082500 DOI: 10.1007/978-1-4939-7357-6_18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Due to advances in the fields of stem cell biology and cellular engineering, a variety of commercially available human-derived neurons and neural progenitor cells (NPCs) are now available for use in research applications, including small molecule efficacy or toxicity screening. The use of human-derived neural cells is anticipated to address some of the uncertainties associated with the use of nonhuman culture models or transformed cell lines derived from human tissues. Many of the human-derived neurons and NPCs currently available from commercial sources recapitulate critical process of nervous system development including NPC proliferation, neurite outgrowth, synaptogenesis, and calcium signaling, each of which can be evaluated using high content image analysis (HCA). Human-derived neurons and NPCs are also amenable to culture in multiwell plate formats and thus may be adapted for use in HCA-based screening applications. This article reviews various types of HCA-based assays that have been used in conjunction with human-derived neurons and NPC cultures. This article also highlights instances where lower throughput analysis of neurodevelopmental processes has been performed and which demonstrate a potential for adaptation to higher-throughout imaging methods. Finally, a generic protocol for evaluating neurite outgrowth in human-derived neurons using a combination of immunocytochemistry and HCA is presented. The information provided in this article is intended to serve as a resource for cell model and assay selection for those interested in evaluating neurodevelopmental processes in human-derived cells.
Collapse
Affiliation(s)
- Joshua A Harrill
- Center for Toxicology and Environmental Health, LLC, 5120 Northshore Drive, Little Rock, AR, 72118, USA.
| |
Collapse
|
25
|
Kim JY, Chun SY, Park JS, Chung JW, Ha YS, Lee JN, Kwon TG. Laminin and Platelet-Derived Growth Factor-BB Promote Neuronal Differentiation of Human Urine-Derived Stem Cells. Tissue Eng Regen Med 2017; 15:195-209. [PMID: 30603547 DOI: 10.1007/s13770-017-0102-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/15/2017] [Accepted: 11/20/2017] [Indexed: 11/24/2022] Open
Abstract
Urine-derived stem cells (USCs) are considered as a promising cell source capable of neuronal differentiation. In addition, specific growth factors and extracellular matrix are essential for enhancing their neuronal differentiation efficiency. In this study, we investigated the possibility of neuronal differentiation of USCs and the role of laminin and platelet-derived growth factor BB (PDGF-BB) as promoting factors. USCs were isolated from fresh urine of healthy donors. Cultured USCs were adherent to the plate and their morphology was similar to the cobblestone. In addition, they showed chromosome stability, rapid proliferation rate, colony forming capacity, and mesenchymal stem cell characteristics. For inducing the neuronal differentiation, USCs were cultured for 14 days in neuronal differentiation media supplemented with/without laminin and/or PDGF-BB. To identify the expression of neuronal markers, RT-PCR, flow cytometry analysis and immunocytochemistry were used. After neuronal induction, the cells showed neuron-like morphological change and high expression level of neuronal markers. In addition, laminin and PDGF-BB respectively promoted the neuronal differentiation of USCs and the combination of laminin and PDGF-BB showed a synergistic effect for the neuronal differentiation of USCs. In conclusion, USCs are noteworthy cell source in the field of neuronal regeneration and laminin and PDGF-BB promote their neuronal differentiation efficiency.
Collapse
Affiliation(s)
- Jung Yeon Kim
- 1Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, 135 Dongdeok-ro, Jung-gu, Daegu, 41940 Korea
| | - So Young Chun
- 1Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, 135 Dongdeok-ro, Jung-gu, Daegu, 41940 Korea
| | - Jin-Sung Park
- 2Department of Neurology, School of Medicine, Kyungpook National University, 807 Hoguk-ro, Buk-gu, Daegu, 41404 Korea
| | - Jae-Wook Chung
- 3Department of Urology, School of Medicine, Kyungpook National University, 807 Hoguk-ro, Buk-gu, Daegu, 41404 Korea
| | - Yun-Sok Ha
- 3Department of Urology, School of Medicine, Kyungpook National University, 807 Hoguk-ro, Buk-gu, Daegu, 41404 Korea
| | - Jun Nyung Lee
- 3Department of Urology, School of Medicine, Kyungpook National University, 807 Hoguk-ro, Buk-gu, Daegu, 41404 Korea
| | - Tae Gyun Kwon
- 1Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, 135 Dongdeok-ro, Jung-gu, Daegu, 41940 Korea.,3Department of Urology, School of Medicine, Kyungpook National University, 807 Hoguk-ro, Buk-gu, Daegu, 41404 Korea
| |
Collapse
|
26
|
Waschek JA, Cohen JR, Chi GC, Proszynski TJ, Niewiadomski P. PACAP Promotes Matrix-Driven Adhesion of Cultured Adult Murine Neural Progenitors. ASN Neuro 2017; 9:1759091417708720. [PMID: 28523979 PMCID: PMC5439654 DOI: 10.1177/1759091417708720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
New neurons are born throughout the life of mammals in germinal zones of the brain known as neurogenic niches: the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus of the hippocampus. These niches contain a subpopulation of cells known as adult neural progenitor cells (aNPCs), which self-renew and give rise to new neurons and glia. aNPCs are regulated by many factors present in the niche, including the extracellular matrix (ECM). We show that the neuropeptide PACAP (pituitary adenylate cyclase-activating polypeptide) affects subventricular zone-derived aNPCs by increasing their surface adhesion. Gene array and reconstitution assays indicate that this effect can be attributed to the regulation of ECM components and ECM-modifying enzymes in aNPCs by PACAP. Our work suggests that PACAP regulates a bidirectional interaction between the aNPCs and their niche: PACAP modifies ECM production and remodeling, in turn the ECM regulates progenitor cell adherence. We speculate that PACAP may in this manner help restrict adult neural progenitors to the stem cell niche in vivo, with potential significance for aNPC function in physiological and pathological states.
Collapse
Affiliation(s)
- James A Waschek
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Joseph R Cohen
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Gloria C Chi
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Tomasz J Proszynski
- 2 Department of Cell Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel Niewiadomski
- 1 Intellectual Development and Disabilities Research Center, The David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,2 Department of Cell Biology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.,3 Centre of New Technologies, University of Warsaw, Warsaw, Poland
| |
Collapse
|
27
|
Roles of Nitric Oxide Synthase Isoforms in Neurogenesis. Mol Neurobiol 2017; 55:2645-2652. [PMID: 28421538 DOI: 10.1007/s12035-017-0513-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/04/2017] [Indexed: 10/19/2022]
Abstract
Nitric oxide (NO), a free radical gas, acts as a neurotransmitter or neuromodulator in the central nervous system (CNS). It has been widely explored as a mediator of neuroinflammation, neuronal damages, and neurodegeneration at its pathological levels. Recently, increasing evidence suggests that NO plays key roles in mediating adult neurogenesis, the process of neural stem cells (NSCs) to generate newborn neurons for replacing damaged neurons or maintaining the function of the brain. NO synthase (NOS) is a major enzyme catalyzing the generation of NO in the brain. Recent studies indicate that three homologous NOS isoforms are involved in the proliferation of NSCs and neurogenesis. Therefore, the impact of NOS isoforms on NSC functions needs to be elucidated. Here, we summarize the studies on the role of NO and NOS with different isoforms in NSC proliferation and neurogenesis with the focus on introducing action mechanisms involved in the regulation of NSC function. This growing research area provides the new insight into controlling NSC function via regulating NO microenvironment in the brain. It also provides the evidence on targeting NOS for the treatment of brain diseases.
Collapse
|
28
|
Afelik S, Rovira M. Pancreatic β-cell regeneration: Facultative or dedicated progenitors? Mol Cell Endocrinol 2017; 445:85-94. [PMID: 27838399 DOI: 10.1016/j.mce.2016.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 10/21/2016] [Accepted: 11/08/2016] [Indexed: 12/19/2022]
Abstract
The adult pancreas is only capable of limited regeneration. Unlike highly regenerative tissues such as the skin, intestinal crypts and hematopoietic system, no dedicated adult stem cells or stem cell niche have so far been identified within the adult pancreas. New β cells have been shown to form in the adult pancreas, in response to high physiological demand or experimental β-cell ablation, mostly by replication of existing β cells. The possibility that new β cells are formed from other sources is currently a point of major controversy. Under particular injury conditions, fully differentiated pancreatic duct and acinar cells have been shown to dedifferentiate into a progenitor-like state, however the extent, to which ductal, acinar or other endocrine cells contribute to restoring pancreatic β-cell mass remains to be resolved. In this review we focus on regenerative events in the pancreas with emphasis on the restoration of β-cell mass. We present an overview of regenerative responses noted within the different pancreatic lineages, following injury. We also highlight the intrinsic plasticity of the adult pancreas that allows for inter-conversion of fully differentiated pancreatic lineages through manipulation of few genes or growth factors. Taken together, evidence from a number of studies suggest that differentiated pancreatic lineages could act as facultative progenitor cells, but the extent to which these contribute to β-cell regeneration in vivo is still a matter of contention.
Collapse
Affiliation(s)
- Solomon Afelik
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago, 840 South Wood Street, CSB 920 (Rm 502), Chicago, IL 60612, USA.
| | - Meritxell Rovira
- Genomic Programming of Beta-Cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
| |
Collapse
|
29
|
Pesce M, Messina E, Chimenti I, Beltrami AP. Cardiac Mechanoperception: A Life-Long Story from Early Beats to Aging and Failure. Stem Cells Dev 2016; 26:77-90. [PMID: 27736363 DOI: 10.1089/scd.2016.0206] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The life-long story of the heart starts concomitantly with primary differentiation events occurring in multipotent progenitors located in the so-called heart tube. This initially tubular structure starts a looping process, which leads to formation of the final four-chambered heart with a primary contribution of geometric and position-associated cell sensing. While this establishes the correct patterning of the final cardiac structure, it also provides feedbacks to fundamental cellular machineries controlling proliferation and differentiation, thus ensuring a coordinated restriction of cell growth and a myocyte terminal differentiation. Novel evidences provided by embryological and cell engineering studies have clarified the relevance of mechanics-supported position sensing for the correct recognition of cell fate inside developing embryos and multicellular aggregates. One of the main components of this pathway, the Hippo-dependent signal transduction machinery, is responsible for cell mechanics intracellular transduction with important consequences for gene transcription and cell growth control. Being the Hippo pathway also directly connected to stress responses and altered metabolism, it is tempting to speculate that permanent alterations of mechanosensing may account for modifying self-renewal control in tissue homeostasis. In the present contribution, we translate these concepts to the aging process and the failing of the human heart, two pathophysiologic conditions that are strongly affected by stress responses and altered metabolism.
Collapse
Affiliation(s)
- Maurizio Pesce
- 1 Tissue Engineering Research Unit, Centro Cardiologico Monzino, IRCCS , Milan, Italy
| | - Elisa Messina
- 2 Department of Pediatric Cardiology, "Sapienza" University , Rome, Italy
| | - Isotta Chimenti
- 3 Department of Medical Surgical Science and Biotechnology, "Sapienza" University , Rome, Italy
| | | |
Collapse
|
30
|
Capetian P, Azmitia L, Pauly MG, Krajka V, Stengel F, Bernhardi EM, Klett M, Meier B, Seibler P, Stanslowsky N, Moser A, Knopp A, Gillessen-Kaesbach G, Nikkhah G, Wegner F, Döbrössy M, Klein C. Plasmid-Based Generation of Induced Neural Stem Cells from Adult Human Fibroblasts. Front Cell Neurosci 2016; 10:245. [PMID: 27822179 PMCID: PMC5075569 DOI: 10.3389/fncel.2016.00245] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/06/2016] [Indexed: 01/01/2023] Open
Abstract
Direct reprogramming from somatic to neural cell types has become an alternative to induced pluripotent stem cells. Most protocols employ viral expression systems, posing the risk of random genomic integration. Recent developments led to plasmid-based protocols, lowering this risk. However, these protocols either relied on continuous presence of a variety of small molecules or were only able to reprogram murine cells. We therefore established a reprogramming protocol based on vectors containing the Epstein-Barr virus (EBV)-derived oriP/EBNA1 as well as the defined expression factors Oct3/4, Sox2, Klf4, L-myc, Lin28, and a small hairpin directed against p53. We employed a defined neural medium in combination with the neurotrophins bFGF, EGF and FGF4 for cultivation without the addition of small molecules. After reprogramming, cells demonstrated a temporary increase in the expression of endogenous Oct3/4. We obtained induced neural stem cells (iNSC) 30 days after transfection. In contrast to previous results, plasmid vectors as well as a residual expression of reprogramming factors remained detectable in all cell lines. Cells showed a robust differentiation into neuronal (72%) and glial cells (9% astrocytes, 6% oligodendrocytes). Despite the temporary increase of pluripotency-associated Oct3/4 expression during reprogramming, we did not detect pluripotent stem cells or non-neural cells in culture (except occasional residual fibroblasts). Neurons showed electrical activity and functional glutamatergic synapses. Our results demonstrate that reprogramming adult human fibroblasts to iNSC by plasmid vectors and basic neural medium without small molecules is possible and feasible. However, a full set of pluripotency-associated transcription factors may indeed result in the acquisition of a transient (at least partial) pluripotent intermediate during reprogramming. In contrast to previous reports, the EBV-based plasmid system remained present and active inside the cells at all time points.
Collapse
Affiliation(s)
- Philipp Capetian
- Institute of Neurogenetics, University of LübeckLübeck, Germany; Department of Neurology, University of LübeckLübeck, Germany
| | - Luis Azmitia
- Department of Neurosurgery, University of Kiel Kiel, Germany
| | - Martje G Pauly
- Institute of Neurogenetics, University of Lübeck Lübeck, Germany
| | - Victor Krajka
- Institute of Neurogenetics, University of Lübeck Lübeck, Germany
| | - Felix Stengel
- Institute of Neurogenetics, University of Lübeck Lübeck, Germany
| | | | - Mariana Klett
- Laboratory of Stereotaxy and Interventional Neuroscience, Department of Stereotactic and Functional Neuroscience, University Medical Center Freiburg Freiburg im Breisgau, Germany
| | - Britta Meier
- Institute of Neurogenetics, University of Lübeck Lübeck, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck Lübeck, Germany
| | | | - Andreas Moser
- Department of Neurology, University of Lübeck Lübeck, Germany
| | - Andreas Knopp
- Institute of Physiology, University of Kiel Kiel, Germany
| | | | - Guido Nikkhah
- Department of Neurosurgery, University of Erlangen-Nuremberg Erlangen, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School Hanover, Germany
| | - Máté Döbrössy
- Laboratory of Stereotaxy and Interventional Neuroscience, Department of Stereotactic and Functional Neuroscience, University Medical Center Freiburg Freiburg im Breisgau, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck Lübeck, Germany
| |
Collapse
|
31
|
Vascular Transdifferentiation in the CNS: A Focus on Neural and Glioblastoma Stem-Like Cells. Stem Cells Int 2016; 2016:2759403. [PMID: 27738435 PMCID: PMC5055959 DOI: 10.1155/2016/2759403] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 09/05/2016] [Indexed: 01/12/2023] Open
Abstract
Glioblastomas are devastating and extensively vascularized brain tumors from which glioblastoma stem-like cells (GSCs) have been isolated by many groups. These cells have a high tumorigenic potential and the capacity to generate heterogeneous phenotypes. There is growing evidence to support the possibility that these cells are derived from the accumulation of mutations in adult neural stem cells (NSCs) as well as in oligodendrocyte progenitors. It was recently reported that GSCs could transdifferentiate into endothelial-like and pericyte-like cells both in vitro and in vivo, notably under the influence of Notch and TGFβ signaling pathways. Vascular cells derived from GBM cells were also observed directly in patient samples. These results could lead to new directions for designing original therapeutic approaches against GBM neovascularization but this specific reprogramming requires further molecular investigations. Transdifferentiation of nontumoral neural stem cells into vascular cells has also been described and conversely vascular cells may generate neural stem cells. In this review, we present and discuss these recent data. As some of them appear controversial, further validation will be needed using new technical approaches such as high throughput profiling and functional analyses to avoid experimental pitfalls and misinterpretations.
Collapse
|
32
|
Ababon MR, Li BI, Matteson PG, Millonig JH. Quantitative Measurement of Relative Retinoic Acid Levels in E8.5 Embryos and Neurosphere Cultures Using the F9 RARE-Lacz Cell-based Reporter Assay. J Vis Exp 2016. [PMID: 27684594 PMCID: PMC5091987 DOI: 10.3791/54443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Retinoic acid (RA) is an important developmental morphogen that coordinates anteroposterior and dorsoventral axis patterning, somitic differentiation, neurogenesis, patterning of the hindbrain and spinal cord, and the development of multiple organ systems. Due to its chemical nature as a small amphipathic lipid, direct detection and visualization of RA histologically remains technically impossible. Currently, methods used to infer the presence and localization of RA make use of reporter systems that detect the biological activity of RA. Most established reporter systems, both transgenic mice and cell lines, make use of the highly potent RA response element (RARE) upstream of the RAR-beta gene to drive RA-inducible expression of reporter genes, such as beta-galactosidase or luciferase. The transgenic RARE-LacZ mouse is useful in visualizing spatiotemporal changes in RA signaling especially during embryonic development. However, it does not directly measure overall RA levels. As a reporter system, the F9 RARE-LacZ cell line can be used in a variety of ways, from simple detection of RA to quantitative measurements of RA levels in tissue explants. Here we describe the quantitative determination of relative RA levels generated in embryos and neurosphere cultures using the F9 RARE-LacZ reporter cell line.
Collapse
Affiliation(s)
- Myka R Ababon
- Center for Advanced Biotechnology and Medicine, Rutgers University
| | - Bo I Li
- Department of Neuroscience and Cell Biology, Rutgers University
| | - Paul G Matteson
- Center for Advanced Biotechnology and Medicine, Rutgers University
| | - James H Millonig
- Center for Advanced Biotechnology and Medicine, Rutgers University; Department of Neuroscience and Cell Biology, Rutgers University;
| |
Collapse
|
33
|
Peretz Y, Eren N, Kohl A, Hen G, Yaniv K, Weisinger K, Cinnamon Y, Sela-Donenfeld D. A new role of hindbrain boundaries as pools of neural stem/progenitor cells regulated by Sox2. BMC Biol 2016; 14:57. [PMID: 27392568 PMCID: PMC4938926 DOI: 10.1186/s12915-016-0277-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/21/2016] [Indexed: 01/28/2023] Open
Abstract
Background Compartment boundaries are an essential developmental mechanism throughout evolution, designated to act as organizing centers and to regulate and localize differently fated cells. The hindbrain serves as a fascinating example for this phenomenon as its early development is devoted to the formation of repetitive rhombomeres and their well-defined boundaries in all vertebrates. Yet, the actual role of hindbrain boundaries remains unresolved, especially in amniotes. Results Here, we report that hindbrain boundaries in the chick embryo consist of a subset of cells expressing the key neural stem cell (NSC) gene Sox2. These cells co-express other neural progenitor markers such as Transitin (the avian Nestin), GFAP, Pax6 and chondroitin sulfate proteoglycan. The majority of the Sox2+ cells that reside within the boundary core are slow-dividing, whereas nearer to and within rhombomeres Sox2+ cells are largely proliferating. In vivo analyses and cell tracing experiments revealed the contribution of boundary Sox2+ cells to neurons in a ventricular-to-mantle manner within the boundaries, as well as their lateral contribution to proliferating Sox2+ cells in rhombomeres. The generation of boundary-derived neurospheres from hindbrain cultures confirmed the typical NSC behavior of boundary cells as a multipotent and self-renewing Sox2+ cell population. Inhibition of Sox2 in boundaries led to enhanced and aberrant neural differentiation together with inhibition in cell-proliferation, whereas Sox2 mis-expression attenuated neurogenesis, confirming its significant function in hindbrain neuronal organization. Conclusions Data obtained in this study deciphers a novel role of hindbrain boundaries as repetitive pools of neural stem/progenitor cells, which provide proliferating progenitors and differentiating neurons in a Sox2-dependent regulation. Electronic supplementary material The online version of this article (doi:10.1186/s12915-016-0277-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuval Peretz
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel
| | - Noa Eren
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel
| | - Gideon Hen
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Karina Yaniv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Karen Weisinger
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Yuval Cinnamon
- Institute of Animal Sciences, Department of Poultry and Aquaculture Sciences, Agricultural Research Organization, The Volcani Center, Bet Dagan, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 76100, Israel.
| |
Collapse
|
34
|
Karout M, Miesch M, Geoffroy P, Kraft S, Hofmann HD, Mensah-Nyagan AG, Kirsch M. Novel analogs of allopregnanolone show improved efficiency and specificity in neuroprotection and stimulation of proliferation. J Neurochem 2016; 139:782-794. [PMID: 27256158 DOI: 10.1111/jnc.13693] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 05/04/2016] [Accepted: 05/24/2016] [Indexed: 01/28/2023]
Abstract
The natural neurosteroid allopregnanolone exerts beneficial effects in animal models of neurodegenerative diseases, nervous system injury and peripheral neuropathies. It not only has anti-apoptotic activity, but also promotes proliferation of progenitor cells. With respect to using it as a therapeutic tool, such pleiotropic actions might create unwanted side effects. Therefore, we have synthesized allopregnanolone analogs and analyzed their neuroprotective and proliferative effects to identify compounds with higher efficiency and less ambiguous biological actions. Proliferation-promoting effects of 3α and 3β isomers of 3-O-allyl-allopregnanolone and 12 oxo-allopregnanolone were studied in adult subventricular zone stem cell cultures and in primary hippocampal cultures by measuring 5-ethynyl-2'-deoxyuridine incorporation. Neuroprotective activity against amyloid beta 42-induced cell death was determined by quantifying caspase 3/7 activity. The 3α isomers significantly stimulated proliferation in all culture systems, whereas the 3β isomers were ineffective. The stimulatory effect of 3α-O-allyl-allopregnanolone was significantly higher than that of allopregnanolone. In neural stem cell cultures, 3α-O-allyl-allopregnanolone specifically enhanced proliferation of Nestin-positive progenitors. In addition, it promoted the differentiation of doublecortin-positive neurons. In neural stem cell cultures treated with amyloid beta 42, both the α and β isomers of O-allyl- allopregnanolone showed increased neuroprotective activity as compared to allopregnanolone, completely preventing amyloid-induced caspase 3/7 activation. The 12 oxo-allopregnanolone analogs were ineffective. These results identify structural allopregnanolone analogs with higher anti-apoptotic and proliferation-promoting activity than the natural neurosteroid. Interestingly, stereoisomers of the analogs were found to have distinct profiles of activity raising the possibility of exploiting the neuroprotective properties of neurosteroids with or without simultaneously stimulating neurogenesis. Cover Image for this issue: doi: 10.1111/jnc.13344.
Collapse
Affiliation(s)
- Mona Karout
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, ZfN, Faculty of Medicine, University of Freiburg, Albertstr. 23, D-79104, Freiburg, Germany.,Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Michel Miesch
- Laboratoire de Chimie Organique Synthétique, UMR 7177, Institut de Chimie de l'Université de Strasbourg, Strasbourg, France
| | - Philippe Geoffroy
- Laboratoire de Chimie Organique Synthétique, UMR 7177, Institut de Chimie de l'Université de Strasbourg, Strasbourg, France
| | - Stephanie Kraft
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, ZfN, Faculty of Medicine, University of Freiburg, Albertstr. 23, D-79104, Freiburg, Germany
| | - Hans-Dieter Hofmann
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, ZfN, Faculty of Medicine, University of Freiburg, Albertstr. 23, D-79104, Freiburg, Germany
| | - Ayikoe Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Matthias Kirsch
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, ZfN, Faculty of Medicine, University of Freiburg, Albertstr. 23, D-79104, Freiburg, Germany
| |
Collapse
|
35
|
Aleksandrova MA, Poltavtseva RA, Marei MV, Sukhikh GT. Analysis of Neural Stem Cells from Human Cortical Brain Structures In Vitro. Bull Exp Biol Med 2016; 161:197-208. [PMID: 27279101 DOI: 10.1007/s10517-016-3375-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Indexed: 12/12/2022]
Abstract
Comparative immunohistochemical analysis of the neocortex from human fetuses showed that neural stem and progenitor cells are present in the brain throughout the gestation period, at least from week 8 through 26. At the same time, neural stem cells from the first and second trimester fetuses differed by the distribution, morphology, growth, and quantity. Immunocytochemical analysis of neural stem cells derived from fetuses at different gestation terms and cultured under different conditions showed their differentiation capacity. Detailed analysis of neural stem cell populations derived from fetuses on gestation weeks 8-9, 18-20, and 26 expressing Lex/SSEA1 was performed.
Collapse
Affiliation(s)
- M A Aleksandrova
- N. K. Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia.,V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - R A Poltavtseva
- V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - M V Marei
- V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - G T Sukhikh
- V. I. Kulakov Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
36
|
Watters AK, Rom S, Hill JD, Dematatis MK, Zhou Y, Merkel SF, Andrews AM, Cena J, Potula R, Skuba A, Son YJ, Persidsky Y, Ramirez SH. Identification and dynamic regulation of tight junction protein expression in human neural stem cells. Stem Cells Dev 2016; 24:1377-89. [PMID: 25892136 DOI: 10.1089/scd.2014.0497] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Recent reports indicate that neural stem cells (NSCs) exist in a cluster-like formation in close proximity to cerebral microvessels. Similar appearing clusters can be seen ex vivo in NSC cultures termed neurospheres. It is known that this neurosphere configuration is important for preserving stemness and a proliferative state. How NSCs form neurospheres or neuroclusters remains largely undetermined. In this study, we show that primary human NSCs express the tight junction proteins (TJPs): zonula occludens-1 (ZO-1), occludin, claudin-1, -3, -5, and -12. The relative mRNA expression was measured by quantitative polymerase chain reaction, and protein expression was confirmed by flow cytometry and immunofluorescence microscopy. Our results show that downregulation of TJPs occurs as neuronal differentiation is induced, suggesting that control of TJPs may be tied to the neuronal differentiation program. Importantly, upon specific knockdown of the accessory TJP, ZO-1, undifferentiated NSCs showed decreased levels of key stem cell markers. Taken together, our results indicate that TJPs possibly aid in maintaining the intercellular configuration of NSCs and that reduction in TJP expression consequently affects the stemness status.
Collapse
Affiliation(s)
- Andrea K Watters
- 1Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Slava Rom
- 1Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Jeremy D Hill
- 1Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Marie K Dematatis
- 1Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Yu Zhou
- 1Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Steven F Merkel
- 1Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Allison M Andrews
- 1Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Jonathan Cena
- 1Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Raghava Potula
- 1Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania.,2Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Andrew Skuba
- 3Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Young-Jin Son
- 3Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Yuri Persidsky
- 1Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania.,2Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Servio H Ramirez
- 1Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania.,2Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, Pennsylvania.,3Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
37
|
Neurogenic Effects of Cell-Free Extracts of Adipose Stem Cells. PLoS One 2016; 11:e0148691. [PMID: 26859291 PMCID: PMC4747593 DOI: 10.1371/journal.pone.0148691] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/20/2016] [Indexed: 11/19/2022] Open
Abstract
Stem-cell-based therapies are regarded as promising treatments for neurological disorders, and adipose-derived stem cells (ASCs) are a feasible source of clinical application of stem cell. Recent studies have shown that stem cells have a therapeutic potential for use in the treatment of various illnesses through paracrine action. To examine the effects of cell components of ASCs on neural stem cells (NSCs), we treated cell-free extracts of ASCs (CFE-ASCs) containing various components with brain-derived NSCs. To elucidate the effects of CFE-ASCs in NSC proliferation, we treated mouse subventricular zone-derived cultured NSCs with various doses of CFE-ASCs. As a result, CFE-ASCs were found to induce the proliferation of NSCs under conditions of growth factor deprivation in a dose-dependent manner (p<0.01). CFE-ASCs increase the expression of neuron and astrocyte differentiation markers including Tuj-1 (p<0.05) and glial fibrillary acidic protein (p<0.01) without altering the cell’s fate in differentiating NSCs. In addition, treatment with CFE-ASCs induces an increase in neurite numbers (p<0.01) and lengths of NSCs (p<0.05). Furthermore, CFE-ASCs rescue the hydrogen peroxide-induced reduction of NSCs’ viability (p<0.05) and neurite branching (p<0.01). Findings from our study indicate that CFE-ASCs support the survival, proliferation and differentiation of NSCs accompanied with neurite outgrowth, suggesting that CFE-ASCs can modulate neurogenesis in the central nervous system.
Collapse
|
38
|
Glioma Stem Cells. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
39
|
Baskin R, Woods NT, Mendoza-Fandiño G, Forsyth P, Egan KM, Monteiro ANA. Functional analysis of the 11q23.3 glioma susceptibility locus implicates PHLDB1 and DDX6 in glioma susceptibility. Sci Rep 2015; 5:17367. [PMID: 26610392 PMCID: PMC4661592 DOI: 10.1038/srep17367] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/28/2015] [Indexed: 12/04/2022] Open
Abstract
Glioma is the most common malignant primary brain tumor and is associated with poor prognosis. Genetic factors contributing to glioma risk have recently been investigated through genome-wide association studies (GWAS), implicating seven independent glioma risk loci in six chromosomal regions. Here, we performed an in-depth functional analysis of the risk locus proximal to the PHLDB1 gene on 11q23.3. We retrieved all SNPs in linkage disequilibrium (r2 ≥ 0.2) with the glioma-associated SNP (rs498872) and performed a comprehensive bioinformatics and experimental functional analysis for the region. After testing candidate SNPs for allele-specific activity in a luciferase-based enhancer scanning assay, we established a subset of 10 functional SNPs in the promoters of PHLDB1 and DDX6, and in a putative enhancer element. Chromatin conformation capture (3C) identified a physical interaction between the enhancer element containing a functional SNP (rs73001406) and the promoter of the DDX6 gene. Knockdown experiments in cell culture and 3D assays to evaluate the role of PHLDB1 and DDX6 suggest that both genes may contribute to the phenotype. These studies reveal the functional landscape of the 11q23.3 glioma susceptibility locus and identify a network of functional SNPs in regulatory elements and two target genes as a possible mechanism driving glioma risk association.
Collapse
Affiliation(s)
- Rebekah Baskin
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Nicholas T Woods
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.,Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Gustavo Mendoza-Fandiño
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Peter Forsyth
- Department of Neuro-oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kathleen M Egan
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Alvaro N A Monteiro
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
40
|
Poly-L-ornithine promotes preferred differentiation of neural stem/progenitor cells via ERK signalling pathway. Sci Rep 2015; 5:15535. [PMID: 26503112 PMCID: PMC4622086 DOI: 10.1038/srep15535] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 09/28/2015] [Indexed: 11/09/2022] Open
Abstract
Neural stem/progenitor cells (NSPCs) replacement therapies are the most attractive strategies to restore an injured brain. Key challenges of such therapies are enriching NSPCs and directing them differentiation into specific neural cell types. Here, three biomaterial substrates Poly-L-ornithine (PO), Poly-L-lysine (PLL) and fibronectin (FN) were investigated for their effects on proliferation and differentiation of rat NSPCs, and the underlying mechanisms were also explored. The results showed PO significantly increased NSPCs proliferation and induced preferred differentiation, compared with PLL and FN. Checking protein markers of several neural cell subtypes, it is showed PO significantly induced NSPCs expressing Doublecortin (DCX) and Olig2, one for neuroblasts and young neurons and the other for young oligodendrocytes. It is suggested the ERK signaling pathway was involving in this process because an ERK antagonist U0126 could inhibit PO’s effects mentioned above, as well as an ERK pathway agonist Ceramide C6 could enhance them. Given that both neurons and oligodendrocytes are the most vulnerable cells in many neurological diseases, PO-induced preferred differentiation into neurons and oligodendrocytes is a potential paradigm for NSPCs-based therapies.
Collapse
|
41
|
Chatzi C, Schnell E, Westbrook GL. Localized hypoxia within the subgranular zone determines the early survival of newborn hippocampal granule cells. eLife 2015; 4:e08722. [PMID: 26476335 PMCID: PMC4714973 DOI: 10.7554/elife.08722] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 10/16/2015] [Indexed: 01/09/2023] Open
Abstract
The majority of adult hippocampal newborn cells die during early differentiation from intermediate progenitors (IPCs) to immature neurons. Neural stem cells in vivo are located in a relative hypoxic environment, and hypoxia enhances their survival, proliferation and stemness in vitro. Thus, we hypothesized that migration of IPCs away from hypoxic zones within the SGZ might result in oxidative damage, thus triggering cell death. Hypoxic niches were observed along the SGZ, composed of adult NSCs and early IPCs, and oxidative byproducts were present in adjacent late IPCs and neuroblasts. Stabilizing hypoxia inducible factor-1α with dimethyloxallyl glycine increased early survival, but not proliferation or differentiation, in neurospheres in vitro and in newly born SGZ cells in vivo. Rescue experiments in Bax(fl/fl) mutants supported these results. We propose that localized hypoxia within the SGZ contributes to the neurogenic microenvironment and determines the early, activity-independent survival of adult hippocampal newborn cells.
Collapse
Affiliation(s)
- Christina Chatzi
- The Vollum Institute, Oregon Health and Science University, Portland, United States
| | - Eric Schnell
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, United States.,United States Department of Veterans Affairs, VA Portland Health Care System, Portland, United States
| | - Gary L Westbrook
- The Vollum Institute, Oregon Health and Science University, Portland, United States
| |
Collapse
|
42
|
Henry E, Cores J, Hensley MT, Anthony S, Vandergriff A, de Andrade JBM, Allen T, Caranasos TG, Lobo LJ, Cheng K. Adult Lung Spheroid Cells Contain Progenitor Cells and Mediate Regeneration in Rodents With Bleomycin-Induced Pulmonary Fibrosis. Stem Cells Transl Med 2015; 4:1265-74. [PMID: 26359426 DOI: 10.5966/sctm.2015-0062] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/29/2015] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Lung diseases are devastating conditions and ranked as one of the top five causes of mortality worldwide according to the World Health Organization. Stem cell therapy is a promising strategy for lung regeneration. Previous animal and clinical studies have focused on the use of mesenchymal stem cells (from other parts of the body) for lung regenerative therapies. We report a rapid and robust method to generate therapeutic resident lung progenitors from adult lung tissues. Outgrowth cells from healthy lung tissue explants are self-aggregated into three-dimensional lung spheroids in a suspension culture. Without antigenic sorting, the lung spheroids recapitulate the stem cell niche and contain a natural mixture of lung stem cells and supporting cells. In vitro, lung spheroid cells can be expanded to a large quantity and can form alveoli-like structures and acquire mature lung epithelial phenotypes. In severe combined immunodeficiency mice with bleomycin-induced pulmonary fibrosis, intravenous injection of human lung spheroid cells inhibited apoptosis, fibrosis, and infiltration but promoted angiogenesis. In a syngeneic rat model of pulmonary fibrosis, lung spheroid cells outperformed adipose-derived mesenchymal stem cells in reducing fibrotic thickening and infiltration. Previously, lung spheroid cells (the spheroid model) had only been used to study lung cancer cells. Our data suggest that lung spheroids and lung spheroid cells from healthy lung tissues are excellent sources of regenerative lung cells for therapeutic lung regeneration. SIGNIFICANCE The results from the present study will lead to future human clinical trials using lung stem cell therapies to treat various incurable lung diseases, including pulmonary fibrosis. The data presented here also provide fundamental knowledge regarding how injected stem cells mediate lung repair in pulmonary fibrosis.
Collapse
Affiliation(s)
- Eric Henry
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, USA
| | - Jhon Cores
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, USA
| | - M Taylor Hensley
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Shirena Anthony
- Department of Biology, North Carolina State University, Raleigh, North Carolina, USA
| | - Adam Vandergriff
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, USA
| | - James B M de Andrade
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Tyler Allen
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Thomas G Caranasos
- Division of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Leonard J Lobo
- Division of Pulmonary Diseases and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Center for Comparative Medicine and Translational Research, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
43
|
Lang H, Xing Y, Brown LN, Samuvel DJ, Panganiban CH, Havens LT, Balasubramanian S, Wegner M, Krug EL, Barth JL. Neural stem/progenitor cell properties of glial cells in the adult mouse auditory nerve. Sci Rep 2015; 5:13383. [PMID: 26307538 PMCID: PMC4549618 DOI: 10.1038/srep13383] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/24/2015] [Indexed: 12/14/2022] Open
Abstract
The auditory nerve is the primary conveyor of hearing information from sensory hair cells to the brain. It has been believed that loss of the auditory nerve is irreversible in the adult mammalian ear, resulting in sensorineural hearing loss. We examined the regenerative potential of the auditory nerve in a mouse model of auditory neuropathy. Following neuronal degeneration, quiescent glial cells converted to an activated state showing a decrease in nuclear chromatin condensation, altered histone deacetylase expression and up-regulation of numerous genes associated with neurogenesis or development. Neurosphere formation assays showed that adult auditory nerves contain neural stem/progenitor cells (NSPs) that were within a Sox2-positive glial population. Production of neurospheres from auditory nerve cells was stimulated by acute neuronal injury and hypoxic conditioning. These results demonstrate that a subset of glial cells in the adult auditory nerve exhibit several characteristics of NSPs and are therefore potential targets for promoting auditory nerve regeneration.
Collapse
Affiliation(s)
- Hainan Lang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Yazhi Xing
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - LaShardai N Brown
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Devadoss J Samuvel
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Clarisse H Panganiban
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Luke T Havens
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | | | - Michael Wegner
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Edward L Krug
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Jeremy L Barth
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| |
Collapse
|
44
|
Kim JT, Chung HJ, Seo JY, Yang YI, Choi MY, Kim HI, Yang TH, Lee WJ, Youn YC, Kim HJ, Kim YM, Lee H, Jang YS, Lee SJ. A fibrin-supported myocardial organ culture for isolation of cardiac stem cells via the recapitulation of cardiac homeostasis. Biomaterials 2015; 48:66-83. [DOI: 10.1016/j.biomaterials.2015.01.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 01/11/2015] [Accepted: 01/20/2015] [Indexed: 12/22/2022]
|
45
|
Demir M, Laywell ED. Neurotoxic effects of AZT on developing and adult neurogenesis. Front Neurosci 2015; 9:93. [PMID: 25852464 PMCID: PMC4367529 DOI: 10.3389/fnins.2015.00093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/05/2015] [Indexed: 11/18/2022] Open
Abstract
Azidothymidine (AZT) is a synthetic, chain-terminating nucleoside analog used to treat HIV-1 infection. While AZT is not actively transported across the blood brain barrier, it does accumulate at high levels in cerebrospinal fluid, and subsequently diffuses into the overlying parenchyma. Due to the close anatomical proximity of the neurogenic niches to the ventricular system, we hypothesize that diffusion from CSF exposes neural stem/progenitor cells and their progeny to biologically relevant levels of AZT sufficient to perturb normal cell functions. We employed in vitro and in vivo models of mouse neurogenesis in order to assess the effects of AZT on developing and adult neurogenesis. Using in vitro assays we show that AZT reduces the population expansion potential of neural stem/progenitor cells by inducing senescence. Additionally, in a model of in vitro neurogenesis AZT severely attenuates neuroblast production. These effects are mirrored in vivo by clinically-relevant animal models. We show that in utero AZT exposure perturbs both population expansion and neurogenesis among neural stem/progenitor cells. Additionally, a short-term AZT regimen in adult mice suppresses subependymal zone neurogenesis. These data reveal novel negative effects of AZT on neural stem cell biology. Given that the sequelae of HIV infection often include neurologic deficits—subsumed under AIDS Dementia Complex (Brew, 1999)—it is important to determine to what extent AZT negatively affects neurological function in ways that contribute to, or exacerbate, ADC in order to avoid attributing iatrogenic drug effects to the underlying disease process, and thereby skewing the risk/benefit analysis of AZT therapy.
Collapse
Affiliation(s)
- Meryem Demir
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida Gainesville, FL, USA
| | - Eric D Laywell
- Department of Biomedical Sciences, College of Medicine, Florida State University Tallahassee, FL, USA
| |
Collapse
|
46
|
Wu CH, Hong BH, Ho CT, Yen GC. Targeting cancer stem cells in breast cancer: potential anticancer properties of 6-shogaol and pterostilbene. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:2432-2441. [PMID: 25686711 DOI: 10.1021/acs.jafc.5b00002] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Breast cancer stem cells (BCSCs) constitute a small fraction of the primary tumor that can self-renew and become a drug-resistant cell population, thus limiting the treatment effects of chemotherapeutic drugs. The present study evaluated the cytotoxic effects of five phytochemicals including 6-gingerol (6-G), 6-shogaol (6-S), 5-hydroxy-3,6,7,8,3',4'-hexamethoxyflavone (5-HF), nobiletin (NOL), and pterostilbene (PTE) on MCF-7 breast cancer cells and BCSCs. The results showed that 6-G, 6-S, and PTE selectively killed BCSCs and had high sensitivity for BCSCs isolated from MCF-7 cells that expressed the surface antigen CD44(+)/CD24(-). 6-S and PTE induced cell necrosis phenomena such as membrane injury and bleb formation in BCSCs and inhibited mammosphere formation. In addition, 6-S and PTE increased the sensitivity of isolated BCSCs to chemotherapeutic drugs and significantly increased the anticancer activity of paclitaxel. Analysis of the underlying mechanism showed that 6-S and PTE decreased the expression of the surface antigen CD44 on BCSCs and promoted β-catenin phosphorylation through the inhibition of hedgehog/Akt/GSK3β signaling, thus decreasing the protein expression of downstream c-Myc and cyclin D1 and reducing BCSC stemness.
Collapse
Affiliation(s)
- Chi-Hao Wu
- School of Nutrition and Health Sciences, Taipei Medical University , 250 Wu-Hsing Street, Taipei 110, Taiwan
| | | | | | | |
Collapse
|
47
|
Abstract
Cancer stem cells (CSCs) have been identified in a growing list of malignancies and are believed to be responsible for cancer initiation, metastasis and relapse following certain therapies, even though they may only represent a small fraction of the cells in a given cancer. Like somatic stem cells and embryonic stem cells, CSCs are capable of self-renewal and differentiation into more mature, less tumorigenic cells that make up the bulk populations of cancer cells. Elimination of CSCs promises intriguing therapeutic potential and this concept has been adopted in preclinical drug discovery programs. Herein we will discuss the progress of these efforts, general considerations in practice, major challenges and possible solutions.
Collapse
|
48
|
Jasty S, Suriyanarayanan S, Krishnakumar S. Influence of self-assembling peptide nanofibre scaffolds on retinal differentiation potential of stem/progenitor cells derived from ciliary pigment epithelial cells. J Tissue Eng Regen Med 2014; 11:509-518. [PMID: 25066608 DOI: 10.1002/term.1947] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 06/08/2014] [Accepted: 06/16/2014] [Indexed: 11/12/2022]
Abstract
Aim of the study was to investigate the influence of the self-assembling peptide nanofibre scaffolds (SAPNs) on the growth, proliferation and retinal neuronal differentiation of the stem/progenitor cells (SCs) derived from the ciliary pigment epithelium (CPE) of human cadaveric eye. Here SAPNs (RADA16-I, PM), which is well described in previous studies, commercially available and xeno-free. The CPE cells isolated were cultured in DMEM/F12 supplemented with N2 and growth factors such as basic fibroblast growth factor and epidermal growth factor, encapsulated in the scaffolds. The entrapped SCs actively expanded and formed clone-like clusters in the scaffolds. Many cells in the cluster were proliferating, as revealed by 5-bromo-2-deoxyuridine uptake and could be maintained for up to 6 days and expressed neural progenitor markers such as β-III tubulin, Nestin, Pax6 and Musashi1. Upon differentiation of these cells in conditioned medium, the cells exhibited retinal neuronal markers such as s-Opsin, rhodopsin and Recoverin. The RT2 profiler polymerase chain reaction array experiments showed selective gene expression, possibly involved in neural stem/progenitor cell adhesion and differentiation. These findings suggest the suitability of the three-dimensional culture system for the proliferation and maintenance of CPE stem/progenitor cells (CPE-NS) and for possible use in ex vivo studies of small molecules, drug deliveries for retinal diseases and for use in combination with directed stem/progenitor cell differentiation. and ultimately for tissue replacement therapies. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Srilatha Jasty
- L&T Department of Ocular Pathology.,Radheshyam Kanoi Stem Cell Laboratory, Vision Research Foundation, Sankara Nethralaya, Tamilnadu, India
| | | | | |
Collapse
|
49
|
Thakker-Varia S, Behnke J, Doobin D, Dalal V, Thakkar K, Khadim F, Wilson E, Palmieri A, Antila H, Rantamaki T, Alder J. VGF (TLQP-62)-induced neurogenesis targets early phase neural progenitor cells in the adult hippocampus and requires glutamate and BDNF signaling. Stem Cell Res 2014; 12:762-77. [PMID: 24747217 PMCID: PMC4991619 DOI: 10.1016/j.scr.2014.03.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 02/24/2014] [Accepted: 03/18/2014] [Indexed: 01/19/2023] Open
Abstract
The neuropeptide VGF (non-acronymic), which has antidepressant-like effects, enhances adult hippocampal neurogenesis as well as synaptic activity and plasticity in the hippocampus, however the interaction between these processes and the mechanism underlying this regulation remain unclear. In this study, we demonstrate that VGF-derived peptide TLQP-62 specifically enhances the generation of early progenitor cells in nestin-GFP mice. Specifically, TLQP-62 significantly increases the number of Type 2a neural progenitor cells (NPCs) while reducing the number of more differentiated Type 3 cells. The effect of TLQP-62 on proliferation rather than differentiation was confirmed using NPCs in vitro; TLQP-62 but not scrambled peptide PEHN-62 increases proliferation in a cell line as well as in primary progenitors from adult hippocampus. Moreover, TLQP-62 but not scrambled peptide increases Cyclin D mRNA expression. The proliferation of NPCs induced by TLQP-62 requires synaptic activity, in particular through NMDA and metabotropic glutamate receptors. The activation of glutamate receptors by TLQP-62 activation induces phosphorylation of CaMKII through NMDA receptors and protein kinase D through metabotropic glutamate receptor 5 (mGluR5). Furthermore, pharmacological antagonists to CaMKII and PKD inhibit TLQP-62-induced proliferation of NPCs indicating that these signaling molecules downstream of glutamate receptors are essential for the actions of TLQP-62 on neurogenesis. We also show that TLQP-62 gradually activates Brain-Derived Neurotrophic Factor (BDNF)-receptor TrkB in vitro and that Trk signaling is required for TLQP-62-induced proliferation of NPCs. Understanding the precise molecular mechanism of how TLQP-62 influences neurogenesis may reveal mechanisms by which VGF-derived peptides act as antidepressant-like agents.
Collapse
Affiliation(s)
- Smita Thakker-Varia
- Department of Neuroscience and Cell Biology, Rutgers University - Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | - Joseph Behnke
- Department of Neuroscience and Cell Biology, Rutgers University - Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | - David Doobin
- Department of Neuroscience and Cell Biology, Rutgers University - Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | - Vidhi Dalal
- Department of Neuroscience and Cell Biology, Rutgers University - Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | - Keya Thakkar
- Department of Neuroscience and Cell Biology, Rutgers University - Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | - Farah Khadim
- Department of Neuroscience and Cell Biology, Rutgers University - Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | - Elizabeth Wilson
- Department of Neuroscience and Cell Biology, Rutgers University - Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | - Alicia Palmieri
- Department of Neuroscience and Cell Biology, Rutgers University - Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | - Hanna Antila
- Neuroscience Center, University of Helsinki, P.O. Box 56, Viikinkaari 4, 00014 Helsinki, Finland.
| | - Tomi Rantamaki
- Neuroscience Center, University of Helsinki, P.O. Box 56, Viikinkaari 4, 00014 Helsinki, Finland.
| | - Janet Alder
- Department of Neuroscience and Cell Biology, Rutgers University - Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| |
Collapse
|
50
|
Hagelkruys A, Lagger S, Krahmer J, Leopoldi A, Artaker M, Pusch O, Zezula J, Weissmann S, Xie Y, Schöfer C, Schlederer M, Brosch G, Matthias P, Selfridge J, Lassmann H, Knoblich JA, Seiser C. A single allele of Hdac2 but not Hdac1 is sufficient for normal mouse brain development in the absence of its paralog. Development 2014; 141:604-616. [PMID: 24449838 PMCID: PMC4773893 DOI: 10.1242/dev.100487] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The histone deacetylases HDAC1 and HDAC2 are crucial regulators of chromatin structure and gene expression, thereby controlling important developmental processes. In the mouse brain, HDAC1 and HDAC2 exhibit different developmental stage- and lineage-specific expression patterns. To examine the individual contribution of these deacetylases during brain development, we deleted different combinations of Hdac1 and Hdac2 alleles in neural cells. Ablation of Hdac1 or Hdac2 by Nestin-Cre had no obvious consequences on brain development and architecture owing to compensation by the paralog. By contrast, combined deletion of Hdac1 and Hdac2 resulted in impaired chromatin structure, DNA damage, apoptosis and embryonic lethality. To dissect the individual roles of HDAC1 and HDAC2, we expressed single alleles of either Hdac1 or Hdac2 in the absence of the respective paralog in neural cells. The DNA-damage phenotype observed in double knockout brains was prevented by expression of a single allele of either Hdac1 or Hdac2. Strikingly, Hdac1−/−Hdac2+/− brains showed normal development and no obvious phenotype, whereas Hdac1+/−Hdac2−/− mice displayed impaired brain development and perinatal lethality. Hdac1+/−Hdac2−/− neural precursor cells showed reduced proliferation and premature differentiation mediated by overexpression of protein kinase C, delta, which is a direct target of HDAC2. Importantly, chemical inhibition or knockdown of protein kinase C delta was sufficient to rescue the phenotype of neural progenitor cells in vitro. Our data indicate that HDAC1 and HDAC2 have a common function in maintaining proper chromatin structures and show that HDAC2 has a unique role by controlling the fate of neural progenitors during normal brain development.
Collapse
Affiliation(s)
- Astrid Hagelkruys
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Sabine Lagger
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Julia Krahmer
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Alexandra Leopoldi
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Matthias Artaker
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Oliver Pusch
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Jürgen Zezula
- Institute of Pharmacology, Medical University of Vienna, Vienna 1090, Austria
| | - Simon Weissmann
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| | - Yunli Xie
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Christian Schöfer
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Michaela Schlederer
- Ludwig Boltzmann Institute for Cancer Research (LBICR), Vienna 1090, Austria
| | - Gerald Brosch
- Division of Molecular Biology, Biocenter Innsbruck, Medical University, Innsbruck 6020, Austria
| | - Patrick Matthias
- Friedrich Miescher Institute for Biomedical Research, Novartis Research Foundation, Basel 4058, Switzerland
| | - Jim Selfridge
- Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3QR, UK
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna 1090, Austria
| | - Jürgen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Christian Seiser
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna 1030, Austria
| |
Collapse
|