1
|
Fofie CK, Granja-Vazquez R, Truong V, Walsh P, Price T, Biswas S, Dussor G, Pancrazio J, Kolber B. Profiling human iPSC-derived sensory neurons for analgesic drug screening using a multi-electrode array. CELL REPORTS METHODS 2025; 5:101051. [PMID: 40367946 DOI: 10.1016/j.crmeth.2025.101051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/16/2025] [Accepted: 04/21/2025] [Indexed: 05/16/2025]
Abstract
Chronic pain is a global health issue, yet effective treatments remain limited due to poor preclinical-to-human translation. To address this, we developed a high-content screening (HCS) platform using hiPSC-derived nociceptors to identify analgesics targeting the peripheral nervous system. These cells, cultured on multi-well microelectrode arrays, achieved nearly 100% active electrodes by week 2, maintaining stable activity for at least 2 weeks. After 28 days, we assessed drug effects on neuronal activity, achieving strong assay performance (robust Z' > 0.5). Pharmacological tests confirmed responses to key analgesic targets, including ion channels (Nav, Cav, Kv, and TRPV1), neurotransmitter receptors (AMPAR and GABA-R), and kinase inhibitors (tyrosine and JAK1/2). Transcriptomic analysis validated target expression, though levels differed from primary human DRG cells. The platform was used to screen over 700 natural compounds, demonstrating its potential for analgesic discovery. This HCS platform facilitates the rapid discovery of uncharacterized analgesics, reducing preclinical-to-human translation failure.
Collapse
Affiliation(s)
- Christian Kuete Fofie
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Rafael Granja-Vazquez
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | | | | | - Theodore Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Swati Biswas
- Department of Mathematical Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Joseph Pancrazio
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Benedict Kolber
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA.
| |
Collapse
|
2
|
Nutt K, Dombros-Ryan Z, Birea R, Franks EV, Eastham S, Godwin M, Adams CF, Chari DM, Jenkins SI. Electrospun Polycaprolactone (PCL) Nanofibers Induce Elongation and Alignment of Co-Cultured Primary Cortical Astrocytes and Neurons. MICROMACHINES 2025; 16:256. [PMID: 40141867 PMCID: PMC11946388 DOI: 10.3390/mi16030256] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/27/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025]
Abstract
Neuromimetic in vitro models, simulating in vivo architecture/organization, are urgently needed to reduce experimental reliance on live animals. Our group recently reported a novel brain tissue derivation protocol, simultaneously deriving all major cortical cell types (including immune cells) in a facile protocol, generating a network of neurons in a single growth medium, which was interfaced with nanomaterials. This represents a significant advance, as tissue engineers overwhelmingly use diverse methods to derive and combine individual brain cells for materials-interfacing. However, this multicellular model lacked cellular directionality/structural organization (unlike the highly organized cortical circuits in vivo). Synthetic nanofiber constructs are of high value in tissue engineering, providing directional cues for cells. Most neuro-nanofiber studies employ simple monocultures of astrocytes/neurons and commonly use peripheral neurons rather than central nervous system populations. Here, we have interfaced our complex brain model (neurons/astrocytes derived simultaneously) with randomly oriented or aligned polycaprolactone (PCL) fiber meshes. Both cell types showed targeted extension along aligned fibers versus coverslips or random fibers. A new analysis method developed in-house demonstrated that peak orientations for astrocytes and neurons correlated with aligned nanofibers. Our data support the concept that nanofiber scaffolds can achieve organized growth of mixed cortical neural cell populations, mimicking neural architecture.
Collapse
Affiliation(s)
- Kayleigh Nutt
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Zoe Dombros-Ryan
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Ruxandra Birea
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Emily Victoria Franks
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Medicine, Keele University, Keele ST5 5BG, UK
| | - Sarah Eastham
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- Department of Biomedical Engineering, University of Strathclyde, Glasgow G4 0NW, UK
| | - Morgan Godwin
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Chris F. Adams
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Divya Maitreyi Chari
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Medicine, Keele University, Keele ST5 5BG, UK
| | - Stuart Iain Jenkins
- Neural Tissue Engineering: Keele (NTEK), Keele University, Keele ST5 5BG, UK (Z.D.-R.); (E.V.F.); (C.F.A.)
- School of Medicine, Keele University, Keele ST5 5BG, UK
| |
Collapse
|
3
|
Kuete CF, Granja-Vazquez R, Truong V, Walsh P, Price T, Biswas S, Dussor G, Pancrazio J, Kolber B. Profiling Human iPSC-Derived Sensory Neurons for Analgesic Drug Screening Using a Multi-Electrode Array. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.623405. [PMID: 39605708 PMCID: PMC11601878 DOI: 10.1101/2024.11.18.623405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Chronic pain is a major global health issue, yet effective treatments are limited by poor translation from preclinical studies to humans. To address this, we developed a high-content screening (HCS) platform for analgesic discovery using hiPSC-derived nociceptors. These cells were cultured on multi-well micro-electrode arrays to monitor activity, achieving nearly 100% active electrodes by week two, maintaining stable activity for at least two weeks. After maturation (28 days), we exposed the nociceptors to various drugs, assessing their effects on neuronal activity, with excellent assay performance (Z' values >0.5). Pharmacological tests showed responses to analgesic targets, including ion channels (Nav, Cav, Kv, TRPV1), neurotransmitter receptors (AMPAR, GABA-R), and kinase inhibitors (tyrosine, JAK1/2). Transcriptomic analysis confirmed the presence of these drug targets, although expression levels varied compared to primary human dorsal root ganglion cells. This HCS platform facilitates the rapid discovery of novel analgesics, reducing the risk of preclinical-to-human translation failure. Motivation Chronic pain affects approximately 1.5 billion people worldwide, yet effective treatments remain elusive. A significant barrier to progress in analgesic drug discovery is the limited translation of preclinical findings to human clinical outcomes. Traditional rodent models, although widely used, often fail to accurately predict human responses, while human primary tissues are limited by scarcity, technical difficulties, and ethical concerns. Recent advancements have identified human induced pluripotent stem cell (hiPSC)-derived nociceptors as promising alternatives; however, current differentiation protocols produce cells with inconsistent and physiologically questionable phenotypes.To address these challenges, our study introduces a novel high-content screening (HCS) platform using hiPSC-derived nociceptors cultured on multi-well micro-electrode arrays (MEAs). The "Anatomic" protocol, used to generate these nociceptors, ensures cells with transcriptomic profiles closely matching human primary sensory neurons. Our platform achieves nearly 100% active electrode yield within two weeks and demonstrates sustained, stable activity over time. Additionally, robust Z' factor analysis (exceeding 0.5) confirms the platform's reliability, while pharmacological validation establishes the functional expression of critical analgesic targets. This innovative approach improves both the efficiency and clinical relevance of analgesic drug screening, potentially bridging the translational gap between preclinical studies and human clinical trials, and offering new hope for effective pain management.
Collapse
|
4
|
Fasano C, Cavaliere A, Tiranti V, Peron C. Protocol for evaluating mitochondrial respiration in iPSC-derived neurons by the Seahorse XF analyzer. STAR Protoc 2024; 5:103127. [PMID: 39012842 PMCID: PMC11283151 DOI: 10.1016/j.xpro.2024.103127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/06/2024] [Accepted: 05/23/2024] [Indexed: 07/18/2024] Open
Abstract
Here, we present a protocol describing the quantification of oxygen consumption rate (OCR) and maximal respiration rate (MRR) in living induced pluripotent stem cell (iPSC)-derived neurons using the Seahorse analyzer. We guide you through the whole process: culture amplification and seeding of neural progenitor cells (NPCs), their differentiation into neurons, and normalization of the results to cell number in the analytical phase. The assessment of cellular mitochondrial function, by analyzing mitochondrial respiration, could be useful in various diseases as well as in drug screening. For complete details on the use and execution of this protocol, please refer to Aleo et al.1.
Collapse
Affiliation(s)
- Chiara Fasano
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Andrea Cavaliere
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Valeria Tiranti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Camille Peron
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| |
Collapse
|
5
|
Steiner K, Humpel C. Brain Slice Derived Nerve Fibers Grow along Microcontact Prints and are Stimulated by Beta-Amyloid(42). FRONT BIOSCI-LANDMRK 2024; 29:232. [PMID: 38940051 DOI: 10.31083/j.fbl2906232] [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: 02/27/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Alzheimer's disease is characterized by extracellular beta-amyloid plaques, intraneuronal tau neurofibrillary tangles and excessive neurodegeneration. The mechanisms of neuron degeneration and the potential of these neurons to form new nerve fibers for compensation remain elusive. The present study aimed to evaluate the impact of beta-amyloid and tau on new formations of nerve fibers from mouse organotypic brain slices connected to collagen-based microcontact prints. METHODS Organotypic brain slices of postnatal day 8-10 wild-type mice were connected to established collagen-based microcontact prints loaded with polyornithine to enhance nerve fiber outgrowth. Human beta-amyloid(42) or P301S mutated aggregated tau was co-loaded to the prints. Nerve fibers were immunohistochemically stained with neurofilament antibodies. The physiological activity of outgrown neurites was tested with neurotracer MiniRuby, voltage-sensitive dye FluoVolt, and calcium-sensitive dye Rhod-4. RESULTS Immunohistochemical staining revealed newly formed nerve fibers extending along the prints derived from the brain slices. While collagen-only microcontact prints stimulated nerve fiber growth, those loaded with polyornithine significantly enhanced nerve fiber outgrowth. Beta-amyloid(42) significantly increased the neurofilament-positive nerve fibers, while tau had only a weak effect. MiniRuby crystals, retrogradely transported along these newly formed nerve fibers, reached the hippocampus, while FluoVolt and Rhod-4 monitored electrical activity in newly formed nerve fibers. CONCLUSIONS Our data provide evidence that intact nerve fibers can form along collagen-based microcontact prints from mouse brain slices. The Alzheimer's peptide beta-amyloid(42) stimulates this growth, hinting at a neuroprotective function when physiologically active. This "brain-on-chip" model may offer a platform for screening bioactive factors or testing drug effects on nerve fiber growth.
Collapse
Affiliation(s)
- Katharina Steiner
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
6
|
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
|
7
|
Cho Y, Choi Y, Seong H. Nanoscale surface coatings and topographies for neural interfaces. Acta Biomater 2024; 175:55-75. [PMID: 38141934 DOI: 10.1016/j.actbio.2023.12.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/28/2023] [Accepted: 12/14/2023] [Indexed: 12/25/2023]
Abstract
With the lack of minimally invasive tools for probing neuronal systems across spatiotemporal scales, understanding the working mechanism of the nervous system and limited assessments available are imperative to prevent or treat neurological disorders. In particular, nanoengineered neural interfaces can provide a solution to this technological barrier. This review covers recent surface engineering approaches, including nanoscale surface coatings, and a range of topographies from the microscale to the nanoscale, primarily focusing on neural-interfaced biosystems. Specifically, the immobilization of bioactive molecules to fertilize the neural cell lineage, topographical engineering to induce mechanotransduction in neural cells, and enhanced cell-chip coupling using three-dimensional structured surfaces are highlighted. Advances in neural interface design will help us understand the nervous system, thereby achieving the effective treatments for neurological disorders. STATEMENT OF SIGNIFICANCE: • This review focuses on designing bioactive neural interface with a nanoscale chemical modification and topographical engineering at multiscale perspective. • Versatile nanoscale surface coatings and topographies for neural interface are summarized. • Recent advances in bioactive materials applicable for neural cell culture, electrophysiological sensing, and neural implants are reviewed.
Collapse
Affiliation(s)
- Younghak Cho
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Yunyoung Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyejeong Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea.
| |
Collapse
|
8
|
Duan HZ, Zhou X, Hu Q, Liu ML, Wang SH, Zhang J, Jiang XH, Zhang TX, Yu AY. Mannitol inhibits the proliferation of neural stem cell by a p38 mitogen-activated protein kinase-dependent signaling pathway. Chin J Traumatol 2024; 27:42-52. [PMID: 37953130 PMCID: PMC10859289 DOI: 10.1016/j.cjtee.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/14/2023] Open
Abstract
PURPOSE Mannitol is one of the first-line drugs for reducing cerebral edema through increasing the extracellular osmotic pressure. However, long-term administration of mannitol in the treatment of cerebral edema triggers damage to neurons and astrocytes. Given that neural stem cell (NSC) is a subpopulation of main regenerative cells in the central nervous system after injury, the effect of mannitol on NSC is still elusive. The present study aims to elucidate the role of mannitol in NSC proliferation. METHODS C57 mice were derived from the animal house of Zunyi Medical University. A total of 15 pregnant mice were employed for the purpose of isolating NSCs in this investigation. Initially, mouse primary NSCs were isolated from the embryonic cortex of mice and subsequently identified through immunofluorescence staining. In order to investigate the impact of mannitol on NSC proliferation, both cell counting kit-8 assays and neurospheres formation assays were conducted. The in vitro effects of mannitol were examined at various doses and time points. In order to elucidate the role of Aquaporin 4 (AQP4) in the suppressive effect of mannitol on NSC proliferation, various assays including reverse transcription polymerase chain reaction, western blotting, and immunocytochemistry were conducted on control and mannitol-treated groups. Additionally, the phosphorylated p38 (p-p38) was examined to explore the potential mechanism underlying the inhibitory effect of mannitol on NSC proliferation. Finally, to further confirm the involvement of the p38 mitogen-activated protein kinase-dependent (MAPK) signaling pathway in the observed inhibition of NSC proliferation by mannitol, SB203580 was employed. All data were analyzed using SPSS 20.0 software (SPSS, Inc., Chicago, IL). The statistical analysis among multiple comparisons was performed using one-way analysis of variance (ANOVA), followed by Turkey's post hoc test in case of the data following a normal distribution using a Shapiro-Wilk normality test. Comparisons between 2 groups were determined using Student's t-test, if the data exhibited a normal distribution using a Shapiro-Wilk normality test. Meanwhile, data were shown as median and interquartile range and analyzed using the Mann-Whitney U test, if the data failed the normality test. A p < 0.05 was considered as significant difference. RESULTS Primary NSC were isolated from the mice, and the characteristics were identified using immunostaining analysis. Thereafter, the results indicated that mannitol held the capability of inhibiting NSC proliferation in a dose-dependent and time-dependent manner using cell counting kit-8, neurospheres formation, and immunostaining of Nestin and Ki67 assays. During the process of mannitol suppressing NSC proliferation, the expression of AQP4 mRNA and protein was downregulated, while the gene expression of p-p38 was elevated by reverse transcription polymerase chain reaction, immunostaining, and western blotting assays. Subsequently, the administration of SB203580, one of the p38 MAPK signaling pathway inhibitors, partially abrogated this inhibitory effect resulting from mannitol, supporting the fact that the p38 MAPK signaling pathway participated in curbing NSC proliferation induced by mannitol. CONCLUSIONS Mannitol inhibits NSC proliferation through downregulating AQP4, while upregulating the expression of p-p38 MAPK.
Collapse
Affiliation(s)
- Hai-Zhen Duan
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Xin Zhou
- Dazhou Vocational College of Chinese Medicine, Dazhou, 635000, Sichuan province, China; Dachuan District Traditional Chinese Medicine Hospital, Dazhou, 635000, Sichuan province, China
| | - Quan Hu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Meng-Long Liu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Shu-Hong Wang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Ji Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Xu-Heng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Tian-Xi Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - An-Yong Yu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China.
| |
Collapse
|
9
|
Jimenez-Vergara AC, Avina J, Block TJ, Sheldrake A, Koch C, Gonzalez A, Steele J, Díaz-Lasprilla AM, Munoz-Pinto DJ. A Bioinspired Astrocyte-Derived Coating Promotes the In Vitro Proliferation of Human Neural Stem Cells While Maintaining Their Stemness. Biomimetics (Basel) 2023; 8:589. [PMID: 38132528 PMCID: PMC10741944 DOI: 10.3390/biomimetics8080589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023] Open
Abstract
The repair of neuronal tissue is a challenging process due to the limited proliferative capacity of neurons. Neural stem cells (NSCs) can aid in the regeneration process of neural tissue due to their high proliferation potential and capacity to differentiate into neurons. The therapeutic potential of these cells can only be achieved if sufficient cells are obtained without losing their differentiation potential. Toward this end, an astrocyte-derived coating (HAc) was evaluated as a promising substrate to promote the proliferation of NSCs. Mass spectroscopy and scanning electron microscopy were used to characterize the HAc. The proliferation rate and the expression of stemness and differentiation markers in NSCs cultured on the HAc were evaluated and compared to the responses of these cells to commonly used coating materials including Poly-L-Ornithine (PLO), and a Human Induced Pluripotent Stem Cell (HiPSC)-based coating. The use of the HAc promotes the in vitro cell growth of NSCs. The expression of the stemness markers Sox2 and Nestin, and the differentiation marker DCX in the HAc group was akin to the expression of these markers in the controls. In summary, HAc supported the proliferation of NSCs while maintaining their stemness and neural differentiation potential.
Collapse
Affiliation(s)
- Andrea C. Jimenez-Vergara
- Engineering Science Department, Trinity University, San Antonio, TX 78212, USA; (A.C.J.-V.); (J.A.); (A.G.); (A.M.D.-L.)
| | - Jacob Avina
- Engineering Science Department, Trinity University, San Antonio, TX 78212, USA; (A.C.J.-V.); (J.A.); (A.G.); (A.M.D.-L.)
| | | | - Anne Sheldrake
- StemBioSys, San Antonio, TX 78229, USA; (T.J.B.); (A.S.)
| | - Carson Koch
- Neuroscience Program, Trinity University, San Antonio, TX 78212, USA;
| | - Anna Gonzalez
- Engineering Science Department, Trinity University, San Antonio, TX 78212, USA; (A.C.J.-V.); (J.A.); (A.G.); (A.M.D.-L.)
| | - Jennifer Steele
- Physics and Astronomy Department, Trinity University, San Antonio, TX 78212, USA;
| | - Ana M. Díaz-Lasprilla
- Engineering Science Department, Trinity University, San Antonio, TX 78212, USA; (A.C.J.-V.); (J.A.); (A.G.); (A.M.D.-L.)
| | - Dany J. Munoz-Pinto
- Engineering Science Department, Trinity University, San Antonio, TX 78212, USA; (A.C.J.-V.); (J.A.); (A.G.); (A.M.D.-L.)
- Neuroscience Program, Trinity University, San Antonio, TX 78212, USA;
| |
Collapse
|
10
|
Couvrette LJ, Walker KLA, Bui TV, Pelling AE. Plant Cellulose as a Substrate for 3D Neural Stem Cell Culture. Bioengineering (Basel) 2023; 10:1309. [PMID: 38002433 PMCID: PMC10669287 DOI: 10.3390/bioengineering10111309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/06/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
Neural stem cell (NSC)-based therapies are at the forefront of regenerative medicine strategies for various neural defects and injuries such as stroke, traumatic brain injury, and spinal cord injury. For several clinical applications, NSC therapies require biocompatible scaffolds to support cell survival and to direct differentiation. Here, we investigate decellularized plant tissue as a novel scaffold for three-dimensional (3D), in vitro culture of NSCs. Plant cellulose scaffolds were shown to support the attachment and proliferation of adult rat hippocampal neural stem cells (NSCs). Further, NSCs differentiated on the cellulose scaffold had significant increases in their expression of neuron-specific beta-III tubulin and glial fibrillary acidic protein compared to 2D culture on a polystyrene plate, indicating that the scaffold may enhance the differentiation of NSCs towards astrocytic and neuronal lineages. Our findings suggest that plant-derived cellulose scaffolds have the potential to be used in neural tissue engineering and can be harnessed to direct the differentiation of NSCs.
Collapse
Affiliation(s)
- Lauren J. Couvrette
- Department of Biology, University of Ottawa, Gendron Hall, 30 Marie Curie, Ottawa, ON K1N 5N5, Canada
| | - Krystal L. A. Walker
- Department of Physics, University of Ottawa, STEM Complex, 150 Louis Pasteur Pvt., Ottawa, ON K1N 5N5, Canada
| | - Tuan V. Bui
- Department of Biology, University of Ottawa, Gendron Hall, 30 Marie Curie, Ottawa, ON K1N 5N5, Canada
| | - Andrew E. Pelling
- Department of Biology, University of Ottawa, Gendron Hall, 30 Marie Curie, Ottawa, ON K1N 5N5, Canada
- Department of Physics, University of Ottawa, STEM Complex, 150 Louis Pasteur Pvt., Ottawa, ON K1N 5N5, Canada
| |
Collapse
|
11
|
Chen X, Li L, Chen L, Shao W, Chen Y, Fan X, Liu Y, Tang C, Ding S, Xu X, Zhou G, Feng X. Tea polyphenols coated sodium alginate-gelatin 3D edible scaffold for cultured meat. Food Res Int 2023; 173:113267. [PMID: 37803580 DOI: 10.1016/j.foodres.2023.113267] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 10/08/2023]
Abstract
This study aimed to use edible scaffolds as a platform for animal stem cell expansion, thus constructing block-shaped cell culture meat. The tea polyphenols (TP)-coated 3D scaffolds were constructed of sodium alginate (SA) and gelatin (Gel) with good biocompatibility and mechanical support. Initially, the physicochemical properties and mechanical properties of SA-Gel-TP scaffolds were measured, and the biocompatibility of the scaffolds was evaluated by C2C12 cells. SEM results showed that the scaffold had a porous laminar structure with TP particles attached to the surface, while FT-IR results also demonstrated the encapsulation of TP coating on the scaffold. In addition, the porosity of all scaffolds was higher than 40% and the degradation rate during the incubation cycle was less than 40% and the S2-G1-TP0.1-3 h scaffold has excellent cell adhesion and extension. Subsequently, we inoculated rabbit skeletal muscle myoblasts (RbSkMC) on the scaffold and induced differentiation. The results showed good adhesion and extension behavior of RbSkMC on S2-G1-TP0.1-3 h scaffolds with high expression of myogenic differentiation proteins and genes, and SEM results confirmed the formation of myotubes. Additionally, the adhesion rate of cells on scaffolds with TP coating was 1.5 times higher than that on scaffolds without coating, which significantly improved the cell proliferation rate and the morphology of cells with extension on the scaffolds. Furthermore, rabbit-derived cultured meat had similar appearance and textural characteristics to fresh meat. These conclusions indicate the high potential of the scaffolds with TP coating as a platform for the production of cultured meat products.
Collapse
Affiliation(s)
- Xiaohong Chen
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Linzi Li
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Lin Chen
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China.
| | - Wei Shao
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Yan Chen
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Xiaojing Fan
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Yaping Liu
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China
| | - Changbo Tang
- Lab of Meat Processing and Quality Control of EDU, College of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Shijie Ding
- Lab of Meat Processing and Quality Control of EDU, College of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Xinglian Xu
- Lab of Meat Processing and Quality Control of EDU, College of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Guanghong Zhou
- Lab of Meat Processing and Quality Control of EDU, College of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Xianchao Feng
- College of Food Science and Engineering, Northwest A&F University, No. 22 Xinong Road, Yangling, Shaanxi 712100, China.
| |
Collapse
|
12
|
Patel KD, Gulick AM. Structural and functional insights into δ-poly-L-ornithine polymer biosynthesis from Acinetobacter baumannii. Commun Biol 2023; 6:982. [PMID: 37752201 PMCID: PMC10522769 DOI: 10.1038/s42003-023-05362-4] [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: 03/22/2023] [Accepted: 09/15/2023] [Indexed: 09/28/2023] Open
Abstract
Cationic homo-polyamino acid (CHPA) peptides containing isopeptide bonds of diamino acids have been identified from Actinomycetes strains. However, none has been reported from other bacteria. Here, we report a δ-poly-L-ornithine synthetase from Acinetobacter baumannii, which we name PosA. Surprisingly, structural analysis of the adenylation domain and biochemical assay shows L-ornithine as the substrate for PosA. The product from the enzymatic reaction was purified and identified as poly-L-ornithine composed of 7-12 amino acid units. Chemical labeling of the polymer confirmed the isopeptide linkage of δ-poly-L-ornithine. We examine the biological activity of chemically synthesized 12-mer δ-poly-L-ornithine, illustrating that the polymer may act as an anti-fungal agent. Structures of the isolated adenylation domain from PosA are presented with several diamino acids and biochemical assays identify important substrate binding residues. Structurally-guided genome-mining led to the identification of homologs with different substrate binding residues that could activate additional substrates. A homolog from Bdellovibrionales sp. shows modest activity with L-arginine but not with any diamino acids observed to be substrates for previously examined CHPA synthetases. Our study indicates the possibility that additional CHPAs may be produced by various microbes, supporting the further exploration of uncharacterized natural products.
Collapse
Affiliation(s)
- Ketan D Patel
- Department of Structural Biology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, 14203, USA
| | - Andrew M Gulick
- Department of Structural Biology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY, 14203, USA.
| |
Collapse
|
13
|
Kovacevic B, Jones M, Wagle SR, Ionescu CM, Foster T, Đanić M, Mikov M, Mooranian A, Al-Salami H. Influence of poly-L-ornithine-bile acid nano hydrogels on cellular bioactivity and potential pharmacological applications. Ther Deliv 2023. [PMID: 37667908 DOI: 10.4155/tde-2023-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023] Open
Abstract
Aim: Cellular bioactivity and pathophysiological changes associated with chronic disorders are considered pivotal detrimental factors when developing novel formulations for biomedical applications. Methods: This paper investigates the use of bile acids and synthetic polypeptide poly-L-ornithine (PLO) in formulations and their impacts on a variety of cell lines, with a particular focus on their cellular bioactivity. Results: The hepatic cell line was the most negatively affected by the presence of PLO, while the muscle and beta-pancreatic cell lines did not show as profound of a negative impact of PLO on cellular viability. PLO was the least disruptive regarding mitochondrial function for muscle and beta cells. Conclusion: The addition of bile acids generally decreased mitochondrial respiration and altered bioenergetic parameters in all cell lines.
Collapse
Affiliation(s)
- Bozica Kovacevic
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Melissa Jones
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Susbin Raj Wagle
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Corina Mihaela Ionescu
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Thomas Foster
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Maja Đanić
- Department of Pharmacology, Toxicology & Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, 21101, Serbia
| | - Momir Mikov
- Department of Pharmacology, Toxicology & Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, 21101, Serbia
| | - Armin Mooranian
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
- School of Pharmacy, University of Otago, Dunedin, Otago, 9016, New Zealand
| | - Hani Al-Salami
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
- Medical School, University of Western Australia, Perth, 6000, Australia
| |
Collapse
|
14
|
Harley-Troxell ME, Dhar M. Assembling Spheroids of Rat Primary Neurons Using a Stress-Free 3D Culture System. Int J Mol Sci 2023; 24:13506. [PMID: 37686310 PMCID: PMC10488062 DOI: 10.3390/ijms241713506] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Neural injuries disrupt the normal functions of the nervous system, whose complexities limit current treatment options. Because of their enhanced therapeutic effects, neurospheres have the potential to advance the field of regenerative medicine and neural tissue engineering. Methodological steps can pose challenges for implementing neurosphere assemblies; for example, conventional static cultures hinder yield and throughput, while the presence of the necrotic core, time-consuming methodology, and high variability can slow their progression to clinical application. Here we demonstrate the optimization of primary neural cell-derived neurospheres, developed using a high-throughput, stress-free, 3D bioreactor. This process provides a necessary baseline for future studies that could develop co-cultured assemblies of stem cells combined with endothelial cells, and/or biomaterials and nanomaterials for clinical therapeutic use. Neurosphere size and neurite spreading were evaluated under various conditions using Image J software. Primary neural cells obtained from the hippocampi of three-day-old rat pups, when incubated for 24 h in a reactor coated with 2% Pluronic and seeded on Poly-D-Lysine-coated plates establish neurospheres suitable for therapeutic use within five days. Most notably, neurospheres maintained high cell viability of ≥84% and expressed the neural marker MAP2, neural marker β-Tubulin III, and glial marker GFAP at all time points when evaluated over seven days. Establishing these factors reduces the variability in developing neurospheres, while increasing the ease and output of the culture process and maintaining viable cellular constructs.
Collapse
Affiliation(s)
| | - Madhu Dhar
- Tissue Engineering and Regenerative Medicine, Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA;
| |
Collapse
|
15
|
Fan Y, Xiong Y, Wang X, Chen J, Fang D, Huang J, Yuan G. Poly (lactic-co-glycolic acid)-encapsulated iodine-131 nanoparticles fabricated with rhTSH induce apoptosis and immobilization of thyroid cancer cells. Front Oncol 2023; 13:1030105. [PMID: 36776316 PMCID: PMC9911809 DOI: 10.3389/fonc.2023.1030105] [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: 08/28/2022] [Accepted: 01/02/2023] [Indexed: 01/28/2023] Open
Abstract
Background Aggressive thyroid carcinoma (ATC) usually loses radioiodine avidity to iodine-131 (131I) due to the downregulation of sodium/iodide symporter (NIS). The expression of thyroid stimulating hormone receptor (TSHR) is more persistent than NIS and the administration of recombinant human thyroid stimulating hormone (rhTSH) promotes de novo NIS synthesis. Hence, exploring methods integrating 131I with rhTSH might be a feasible therapeutic strategy for selective delivery of 131I into thyroid cancer to fortify the effect of radioiodine ablation. Methods The 131I, poly (lactic-co-glycolic acid) (PLGA) and rhTSH were used to synthesize of the 131I-PLGA-rhTSH nanoparticles. The characteristics of the 131I-PLGA-rhTSH nanoparticles was determined using a light microscopy, scanning electron microscopy (SEM), autoradiography and immunofluorescence (IF) staining. The diameter of the 131I-PLGA-rhTSH nanoparticles was measured with a Mastersizer 3000, and the encapsulation efficiency (EF) of 131I in 131I-PLGA-rhTSH nanoparticles and the radioactivity of a single nanoparticle were determined. Then, the mouse tumor xenograft model was established, and the biodistribution and effect of 131I-PLGA-rhTSH nanoparticles on apoptosis of thyroid cance cells were investigated in vivo. Thereafter, the role of 131I-PLGA-rhTSH nanoparticles in cell viability using cell counting kit-8 and lactate dehydrogenase (LDH) release assays. Subsequently, the underlying mechanism of 131I-PLGA-rhTSH nanoparticles in reducing cell viability was assessed using immunostaining, boyden invasion assays and phalloidin staining. Results Our results showed that the method of developing nanoparticles-encapsulated 131I using poly (lactic-co-glycolic acid) (PLGA) and modified with rhTSH (131I-PLGA-rhTSH), was a feasible avenue for the integration of 131I and rhTSH. Meanwhile, the encapsulation efficiency (EF) of 131I-PLGA-rhTSH nanoparticles was approximately 60%, and the radioactivity of a single nanoparticle was about 1.1×10-2 Bq. Meanwhile, the 131I-PLGA-rhTSH nanoparticles were selectively delivered into, gradually enriched and slowly downregulated in xenograft tumor after the administration of 131I-PLGA-rhTSH nanoparticles through tail vein in mouse tumor xenograft model. Thereafter, the tumor weight was significantly reduced after the administration of 131I-PLGA-rhTSH nanoparticles. Subsequently, the application of 131I-PLGA-rhTSH nanoparticles facilitated apoptosis and attenuated immobilization via inhibiting F-actin assembling of FTC-133 cells. Conclusion The present study develops a suitable approach integrating 131I and rhTSH, and this strategy is a feasible regimen enhancing the effect of radioiodine ablation for the treatment of thyroid cancer.
Collapse
Affiliation(s)
- Yongzeng Fan
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yalan Xiong
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinhong Wang
- Department of Emergency, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiahao Chen
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Danzhou Fang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiahui Huang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gengbiao Yuan
- Department of Nuclear Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Gengbiao Yuan,
| |
Collapse
|
16
|
Ping J, Fu H, Xiong YJ, Soomro S, Huang ZH, Yu PP. Poly-L-ornithine blocks the inhibitory effects of fibronectin on oligodendrocyte differentiation and promotes myelin repair. Neural Regen Res 2023; 18:832-839. [DOI: 10.4103/1673-5374.353493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
17
|
Nazneen F, Bai F. Isolate and Culture Mouse Primary Neurons for West Nile Virus Infection. Methods Mol Biol 2023; 2585:23-31. [PMID: 36331762 DOI: 10.1007/978-1-0716-2760-0_4] [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] [Indexed: 06/16/2023]
Abstract
Primary neurons are very valuable cells to study the pathogenesis of neurotropic viruses, such as West Nile virus. The mouse primary neurons can be used to assess viral infection profiles and cellular immune responses to a viral infection. However, successful isolation and culture of mouse neurons can be challenging. Here, we report a step-by-step method to prepare a primary neuron culture from adult mice.
Collapse
Affiliation(s)
- Farzana Nazneen
- Department of Cell and Molecular Biology, Center for Molecular and Cellular Biosciences, The University of Southern Mississippi, Hattiesburg, MS, USA
| | - Fengwei Bai
- Department of Cell and Molecular Biology, Center for Molecular and Cellular Biosciences, The University of Southern Mississippi, Hattiesburg, MS, USA.
| |
Collapse
|
18
|
Lainé A, Brot S, Gaillard A. Beneficial Effects of Hyaluronan-Based Hydrogel Implantation after Cortical Traumatic Injury. Cells 2022; 11:cells11233831. [PMID: 36497093 PMCID: PMC9735891 DOI: 10.3390/cells11233831] [Citation(s) in RCA: 7] [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: 07/22/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022] Open
Abstract
Traumatic brain injury (TBI) causes cell death mainly in the cerebral cortex. We have previously reported that transplantation of embryonic cortical neurons immediately after cortical injury allows the anatomical reconstruction of injured pathways and that a delay between cortical injury and cell transplantation can partially improve transplantation efficiency. Biomaterials supporting repair processes in combination with cell transplantation are in development. Hyaluronic acid (HA) hydrogel has attracted increasing interest in the field of tissue engineering due to its attractive biological properties. However, before combining the cell with the HA hydrogel for transplantation, it is important to know the effects of the implanted hydrogel alone. Here, we investigated the therapeutic effect of HA on host tissue after a cortical trauma. For this, we implanted HA hydrogel into the lesioned motor cortex of adult mice immediately or one week after a lesion. Our results show the vascularization of the implanted hydrogel. At one month after HA implantation, we observed a reduction in the glial scar around the lesion and the presence of the newly generated oligodendrocytes, immature and mature neurons within the hydrogel. Implanted hydrogel provides favorable environments for the survival and maturation of the newly generated neurons. Collectively, these results suggest a beneficial effect of biomaterial after a cortical traumatic injury.
Collapse
|
19
|
Carvalho MS, Nogueira DE, Cabral JM, Rodrigues CA. Neural progenitor cell-derived extracellular matrix as a new platform for neural differentiation of human induced pluripotent stem cells. BIOMATERIALS AND BIOSYSTEMS 2022; 8:100070. [PMID: 36824374 PMCID: PMC9934470 DOI: 10.1016/j.bbiosy.2022.100070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/07/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022] Open
Abstract
The culture microenvironment has been demonstrated to regulate stem cell fate and to be a crucial aspect for quality-controlled stem cell maintenance and differentiation to a specific lineage. In this context, extracellular matrix (ECM) proteins are particularly important to mediate the interactions between the cells and the culture substrate. Human induced pluripotent stem cells (hiPSCs) are usually cultured as anchorage-dependent cells and require adhesion to an ECM substrate to support their survival and proliferation in vitro. Matrigel, a common substrate for hiPSC culture is a complex and undefined mixture of ECM proteins which are expensive and not well suited to clinical application. Decellularized cell-derived ECM has been shown to be a promising alternative to the common protein coatings used in stem cell culture. However, very few studies have used this approach as a niche for neural differentiation of hiPSCs. Here, we developed a new stem cell culture system based on decellularized cell-derived ECM from neural progenitor cells (NPCs) for expansion and neural differentiation of hiPSCs, as an alternative to Matrigel and poly-l-ornithine/laminin-coated well plates. Interestingly, hiPSCs were able to grow and maintain their pluripotency when cultured on decellularized ECM from NPCs (NPC ECM). Furthermore, NPC ECM enhanced the neural differentiation of hiPSCs compared to poly-l-ornithine/laminin-coated wells, which are used in most neural differentiation protocols, presenting a statistically significant enhancement of neural gene expression markers, such as βIII-Tubulin and MAP2. Taken together, our results demonstrate that NPC ECM provides a functional microenvironment, mimicking the neural niche, which may have interesting future applications for the development of new strategies in neural stem cell research.
Collapse
Affiliation(s)
- Marta S. Carvalho
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal,Associate Laboratory i4HB – Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal,Corresponding author.
| | - Diogo E.S. Nogueira
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal,Associate Laboratory i4HB – Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Joaquim M.S. Cabral
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal,Associate Laboratory i4HB – Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Carlos A.V. Rodrigues
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal,Associate Laboratory i4HB – Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
20
|
Samara A, Falck M, Spildrejorde M, Leithaug M, Acharya G, Lyle R, Eskeland R. Robust neuronal differentiation of human embryonic stem cells for neurotoxicology. STAR Protoc 2022; 3:101533. [PMID: 36123835 PMCID: PMC9485591 DOI: 10.1016/j.xpro.2022.101533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/23/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022] Open
Abstract
Here, we describe a protocol for rapid neuronal differentiation from human embryonic stem cells (hESCs) toward a heterogenous population of telencephalic progenitors, immature and mature neurons, for drug-screening and early-brain differentiation studies. hESC neuronal differentiation depends on adhesion and minimal cell-passaging to avert monolayer cross-connectivity rupture. In this protocol, we detail optimized cell-seeding densities and coating conditions with high cell viability suitable for neurotoxicology and high-resolution single-cell omics studies. Daily media changes reduce compound instability and degradation for optimal screening. For complete details on the use and execution of this protocol, please refer to Samara et al. (2022).
Collapse
Affiliation(s)
- Athina Samara
- Division of Clinical Paediatrics, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden; Astrid Lindgren Children's Hospital Karolinska University Hospital, 17177 Stockholm, Sweden.
| | - Martin Falck
- Department of Biosciences, University of Oslo, Blindern, PO Box 1066, 0316 Oslo, Norway; PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway
| | - Mari Spildrejorde
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway; Department of Medical Genetics and Norwegian Sequencing Centre, Oslo University Hospital, Kirkeveien 166, 0450 Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0450 Oslo, Norway
| | - Magnus Leithaug
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway; Department of Medical Genetics and Norwegian Sequencing Centre, Oslo University Hospital, Kirkeveien 166, 0450 Oslo, Norway
| | - Ganesh Acharya
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Alfred Nobels Allé 8, 14152 Stockholm, Sweden; Center for Fetal Medicine, Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden
| | - Robert Lyle
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway; Department of Medical Genetics and Norwegian Sequencing Centre, Oslo University Hospital, Kirkeveien 166, 0450 Oslo, Norway; Centre for Fertility and Health, Norwegian Institute of Public Health, PO 222 Skøyen, 0213 Oslo, Norway
| | - Ragnhild Eskeland
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway; Institute of Basic Medical Sciences, Department of Molecular Medicine, Faculty of Medicine, University of Oslo, Blindern, PO Box 1112, 0317 Oslo, Norway.
| |
Collapse
|
21
|
Liu LL, van Rijn RM, Zheng W. Copper Modulates Adult Neurogenesis in Brain Subventricular Zone. Int J Mol Sci 2022; 23:ijms23179888. [PMID: 36077284 PMCID: PMC9456150 DOI: 10.3390/ijms23179888] [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: 08/08/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/18/2022] Open
Abstract
The subventricular zone (SVZ) in lateral ventricles is the largest neurogenic region in adult brain containing high amounts of copper (Cu). This study aims to define the role of Cu in adult neurogenesis by chelating labile Cu ions using a well-established Cu chelator D-Penicillamine (D-Pen). A neurosphere model derived from adult mouse SVZ tissues was established and characterized for its functionality with regards to neural stem/progenitor cells (NSPCs). Applying D-Pen in cultured neurospheres significantly reduced intracellular Cu levels and reversed the Cu-induced suppression of NSPC’s differentiation and migration. An in vivo intracerebroventricular (ICV) infusion model was subsequently established to infuse D-Pen directly into the lateral ventricle. Metal analyses revealed a selective reduction of Cu in SVZ by 13.1% (p = 0.19) and 21.4% (p < 0.05) following D-Pen infusions at low (0.075 μg/h) and high (0.75 μg/h) doses for 28 days, respectively, compared to saline-infused controls. Immunohistochemical studies revealed that the 7-day, low-dose D-Pen infusion significantly increased Ki67(+)/Nestin(+) cell counts in SVZ by 28% (p < 0.05). Quantification of BrdU(+)/doublecortin (DCX)(+) newborn neuroblasts in the rostral migration stream (RMS) and olfactory bulb (OB) further revealed that the short-term, low-dose D-Pen infusion, as compared with saline-infused controls, resulted in more newborn neuroblasts in OB, while the high-dose D-Pen infusion showed fewer newborn neuroblasts in OB but with more arrested in the RMS. Long-term (28-day) infusion revealed similar outcomes. The qPCR data from neurosphere experiments revealed altered expressions of mRNAs encoding key proteins known to regulate SVZ adult neurogenesis, including, but not limited to, Shh, Dlx2, and Slit1, in response to the changed Cu level in neurospheres. Further immunohistochemical data indicated that Cu chelation also altered the expression of high-affinity copper uptake protein 1 (CTR1) and metallothionein-3 (MT3) in the SVZ as well as CTR1 in the choroid plexus, a tissue regulating brain Cu homeostasis. Taken together, this study provides first-hand evidence that a high Cu level in SVZ appears likely to maintain the stability of adult neurogenesis in this neurogenic zone.
Collapse
Affiliation(s)
- Luke L. Liu
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Richard M. van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, West Lafayette, IN 47907, USA
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA
- Correspondence:
| |
Collapse
|
22
|
Generation of Human iPSC-Derived Astrocytes with a mature star-shaped phenotype for CNS modeling. Stem Cell Rev Rep 2022; 18:2494-2512. [PMID: 35488987 PMCID: PMC9489586 DOI: 10.1007/s12015-022-10376-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2022] [Indexed: 11/23/2022]
Abstract
The generation of astrocytes from human induced pluripotent stem cells has been hampered by either prolonged differentiation—spanning over two months—or by shorter protocols that generate immature astrocytes, devoid of salient mature astrocytic traits pivotal for central nervous system (CNS) modeling. We directed stable hiPSC-derived neuroepithelial stem cells to human iPSC-derived Astrocytes (hiAstrocytes) with a high percentage of star-shaped cells by orchestrating an astrocytic-tuned culturing environment in 28 days. We employed RT-qPCR and ICC to validate the astrocytic commitment of the neuroepithelial stem cells. To evaluate the inflammatory phenotype, we challenged the hiAstrocytes with the pro-inflammatory cytokine IL-1β (interleukin 1 beta) and quantitatively assessed the secretion profile of astrocyte-associated cytokines and the expression of intercellular adhesion molecule 1 (ICAM-1). Finally, we quantitatively assessed the capacity of hiAstrocytes to synthesize and export the antioxidant glutathione. In under 28 days, the generated cells express canonical and mature astrocytic markers, denoted by the expression of GFAP, AQP4 and ALDH1L1. In addition, the notion of a mature phenotype is reinforced by the expression of both astrocytic glutamate transporters EAAT1 and EAAT2. Thus, hiAstrocytes have a mature phenotype that encompasses traits critical in CNS modeling, including glutathione synthesis and secretion, upregulation of ICAM-1 and a cytokine secretion profile on a par with human fetal astrocytes. This protocol generates a multifaceted astrocytic model suitable for in vitro CNS disease modeling and personalized medicine.
Collapse
|
23
|
Tetrahydrofolate Alleviates the Inhibitory Effect of Oxidative Stress on Neural Stem Cell Proliferation through PTEN/Akt/mTOR Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9021474. [PMID: 35265266 PMCID: PMC8898800 DOI: 10.1155/2022/9021474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/10/2022] [Indexed: 12/14/2022]
Abstract
Neural stem cell (NSC) proliferation is the initial step for NSC participating in neurorehabilitation after central nervous system (CNS) injury. During this process, oxidative stress is always involved in restricting the regenerative ability of NSC. Tetrahydrofolate (THF) is susceptible to oxidative stress and exhibits a high antioxidant activity. While its effect on NSC proliferation under oxidative stress condition remains obscure. Here, NSC were isolated from embryonic mice and identified using immunofluorescent staining. Meanwhile, the results showed that THF (5 μM and 10 μM) attenuated oxidative stress induced by 50 μM hydrogen peroxide (H2O2) in NSC using mitochondrial hydroxyl radical detection and Western blotting assays. Afterward, administration of THF markedly alleviated the inhibitory effect of oxidative stress on NSC proliferation, which was evidenced by Cell Counting Kit-8 (CCK8), neurosphere formation, and immunofluorescence of Ki67 assays. Thereafter, the results revealed that PTEN/Akt/mTOR signaling pathway played a pivotal role in counteracting oxidative stress to rescue the inhibitory effect of oxidative stress on NSC proliferation using Western blotting assays and gene knockdown techniques. Collectively, these results demonstrate that THF mitigates the inhibitory effect of oxidative stress on NSC proliferation via PTEN/Akt/mTOR signaling pathway, which provides evidence for administrating THF to potentiate the neuro-reparative capacity of NSC in the treatment of CNS diseases with the presence of oxidative stress.
Collapse
|
24
|
Song Y, Chen W, Gai K, Lin F, Sun W. Culture models produced via biomanufacturing for neural tissue-like constructs based on primary neural and neural stem cells. BRAIN SCIENCE ADVANCES 2021. [DOI: 10.26599/bsa.2021.9050021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
25
|
A carboxymethyl lentinan layer by layer self-assembly system as a promising drug chemotherapeutic platform. Carbohydr Polym 2021; 261:117847. [PMID: 33766343 DOI: 10.1016/j.carbpol.2021.117847] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/18/2022]
Abstract
Surface functionalization of mesoporous silica nanoparticles (MSNs) has been proposed as an efficient strategy for enhancing the biocompatibility and efficiency of an MSN-based carrier platform. Herein, natural polyelectrolyte multilayers composed of poly-l-ornithine (PLO) and carboxymethyl lentinan (LC) were coated on the surface of MSNs through a layer-by-layer (LbL) self-assembly technique, and were characterized by ζ-potential, FTIR, 13C NMR, SEM, TEM, XRD, and TG. The prepared carrier presented alternating positive and negative potentials when coated with the polyelectrolytes, and the surface of MSN-PLO/LC was rougher compared to the naked MSNs. The biocompatibility tests, including cytocompatibility, hemocompatibility, and histocompatibility, showed that MSNs biocompatibility could be improved by modifying LC. A high loading and sustained release drug delivery system was constructed after loading doxorubicin (DOX) into the prepared MSN-PLO/LC, which exhibited significant anti-proliferative efficiency in human cervical cancer cell lines (Hela). Therefore, the PLO/LC LbL NPs (layer-by-layer self-assembled nanoparticles coated with PLO/LC layers) based on MSNs, which is easily prepared by electrostatic interactions, can be considered a promising drug chemotherapeutic platform and delivery technique for future human cervical cancer therapy.
Collapse
|
26
|
Zhong J, Li RW, Wang J, Wang Y, Ge HF, Xian JS, Feng H, Tan L. Neuroprotection by cattle encephalon glycoside and ignotin beyond the time window of thrombolysis in ischemic stroke. Neural Regen Res 2021; 16:312-318. [PMID: 32859790 PMCID: PMC7896241 DOI: 10.4103/1673-5374.290899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/02/2019] [Accepted: 03/03/2020] [Indexed: 01/01/2023] Open
Abstract
Cattle encephalon glycoside and ignotin (CEGI) injection is known as a multi-target neuroprotective drug that contains numerous liposoluble molecules, such as polypeptides, monosialotetrahexosyl ganglioside (GM-1), free amino acids, hypoxanthine and carnosine. CEGI has been approved by the Chinese State Food and Drug Administration and widely used in the treatments of various diseases, such as stroke and Alzheimer's disease. However, the neuroprotective effects of CEGI beyond the time window of thrombolysis (within 4.5 hours) on acute ischemic stroke remain unclear. This study constructed a rat middle cerebral artery occlusion model by suture-occluded method to simulate ischemic stroke. The first daily dose was intraperitoneally injected at 8 hours post-surgery and the CEGI treatments continued for 14 days. Results of the modified five-point Bederson scale, beam balance test and rotameric test showed the neurological function of ischemic stroke rats treated with 4 mL/kg/d CEGI improved significantly, but the mortality within 14 days did not change significantly. Brain MRI and 2,3,5-triphenyltetrazolium chloride staining confirmed that the infarct size in the 4 mL/kg/d CEGI-treated rats was significantly reduced compared with ischemic insult only. The results of transmission electron microscopy and double immunofluorescence staining showed that the hippocampal neuronal necrosis in the ischemic penumbra decreased whereas the immunopositivity of new neuronal-specific protein doublecortin and the percentage of Ki67/doublecortin positive cells increased in CEGI-treated rats compared with untreated rats. Our results suggest that CEGI has an effective neuroprotective effect on ischemic stroke when administered after the time window of thrombolysis. The study was approved by the Animal Ethics Committee of The Third Military Medical University, China.
Collapse
Affiliation(s)
- Jun Zhong
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
| | - Rong-Wei Li
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
- Department of Neurosurgery, Hanzhong Central Hospital, Hanzhong, Shaanxi Province, China
| | - Ju Wang
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
| | - Ying Wang
- Department of Oncology, Hanzhong Central Hospital, Hanzhong, Shaanxi Province, China
| | - Hong-Fei Ge
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
| | - Ji-Shu Xian
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Power Transmission Equipment and System Security and New Technology, School of Electrical Engineering, Chongqing University, Chongqing, China
| |
Collapse
|
27
|
Ge H, Zhang C, Yang Y, Chen W, Zhong J, Fang X, Jiang X, Tan L, Zou Y, Hu R, Chen Y, Feng H. Ambroxol Upregulates Glucocerebrosidase Expression to Promote Neural Stem Cells Differentiation Into Neurons Through Wnt/β-Catenin Pathway After Ischemic Stroke. Front Mol Neurosci 2021; 13:596039. [PMID: 33551744 PMCID: PMC7855720 DOI: 10.3389/fnmol.2020.596039] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/22/2020] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke has been becoming one of the leading causes resulting in mortality and adult long-term disability worldwide. Post-stroke pneumonia is a common complication in patients with ischemic stroke and always associated with 1-year mortality. Though ambroxol therapy often serves as a supplementary treatment for post-stroke pneumonia in ischemic stroke patients, its effect on functional recovery and potential mechanism after ischemic stroke remain elusive. In the present study, the results indicated that administration of 70 mg/kg and 100 mg/kg enhanced functional recovery by virtue of decreasing infarct volume. The potential mechanism, to some extent, was due to promoting NSCs differentiation into neurons and interfering NSCs differentiation into astrocytes through increasing GCase expression to activate Wnt/β-catenin signaling pathway in penumbra after ischemic stroke, which advanced basic knowledge of ambroxol in regulating NSCs differentiation and provided a feasible therapy for ischemic stroke treatment, even in other brain disorders in clinic.
Collapse
Affiliation(s)
- Hongfei Ge
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chao Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yang Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Weixiang Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jun Zhong
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuanyu Fang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuheng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Liang Tan
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yongjie Zou
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Neurosurgery, Hospital of People's Liberation Army, Nanchang, China
| | - Rong Hu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
28
|
Wu W, Ge H, Zhang L, Lei X, Yang Y, Fu Y, Feng H. Evaluating the Cytotoxicity of Ti3C2 MXene to Neural Stem Cells. Chem Res Toxicol 2020; 33:2953-2962. [PMID: 33253550 DOI: 10.1021/acs.chemrestox.0c00232] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Wei Wu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Orbital Disease, Third Medical Center of the Chinese PLA General Hospital, Beijing 100039, China
- Brain Research Center and State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hongfei Ge
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Long Zhang
- School of Materials Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Xuejiao Lei
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yang Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yan Fu
- The General Hospital of Western Theater Command, Chengdu 610000, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
29
|
Xue X, Zhang L, Yin X, Chen XX, Chen ZF, Wang CX, Xiang Y, Liu MY, Zhao JH. Transplantation of neural stem cells preconditioned with high‑mobility group box 1 facilitates functional recovery after spinal cord injury in rats. Mol Med Rep 2020; 22:4725-4733. [PMID: 33174002 PMCID: PMC7646886 DOI: 10.3892/mmr.2020.11565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 07/06/2020] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating disorder that often results in temporary and/or permanent functional impairment below the injured level. To date, few satisfactory therapeutic strategies are available to treat SCI. Hence, exploring novel strategies for SCI is an essential public health concern. Cell transplantation therapy, which is associated with neuroprotection, immunomodulation, axon regeneration, neuronal relay formation and myelin regeneration, provides a promising therapeutic strategy for SCI. The neuronal stem cell (NSC) preconditioning method is an emerging approach, which facilitates NSC survival and neuronal differentiation after implantation. The aim of the present study was to develop a feasible candidate for cell-based therapy following SCI in rats and to investigate the role of high mobility group box-1 (HMGB1) in NSC activation. The results of the present study showed that transplantation of NSCs, preconditioned with 1 ng/ml HMGB1, facilitated functional improvement of injured spinal cords, as indicated by Basso, Beattie and Bresnahan mean scores, mechanical hypersensitivity and cold stimulation. Meanwhile, the histological examination of hematoxylin and eosin staining indicated that engraftment of HMGB1-preconditioned NSCs resulted in decreased atrophy of the injured spinal cord. Meanwhile, the transplantation of HMGB1-preconditioned NSCs resulted in an increased number of functional Nissl bodies in neurons, as detected by Nissl staining, and an increase in the number of βIII-tubulin+ cells in the epicenter of injured spinal cords in rats with SCI. In addition, the results also demonstrated that 1 ng/ml HMGB1 promoted the differentiation of NSCs into neurons, and that the ERK signaling pathway played an important role in this process. In conclusion, the present data indicated that the preconditioning strategy with 1 ng/ml HMGB1 may present a feasible candidate for cell-based therapy following SCI in rats, which may enlarge the scope of HMGB1 in NSC activation.
Collapse
Affiliation(s)
- Xin Xue
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University (Army Medical University), Chongqing 400042, P.R. China
| | - Liang Zhang
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University (Army Medical University), Chongqing 400042, P.R. China
| | - Xiang Yin
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University (Army Medical University), Chongqing 400042, P.R. China
| | - Xing-Xing Chen
- Department of Orthopedic, No. 517 Hospital of People's Liberation Army, Xinzhou, Shanxi 030002, P.R. China
| | - Zong-Feng Chen
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University (Army Medical University), Chongqing 400042, P.R. China
| | - Chen-Xu Wang
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University (Army Medical University), Chongqing 400042, P.R. China
| | - Yu Xiang
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University (Army Medical University), Chongqing 400042, P.R. China
| | - Ming-Yong Liu
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University (Army Medical University), Chongqing 400042, P.R. China
| | - Jian-Hua Zhao
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, Third Military Medical University (Army Medical University), Chongqing 400042, P.R. China
| |
Collapse
|
30
|
Actin Alpha 2 (ACTA2) Downregulation Inhibits Neural Stem Cell Migration through Rho GTPase Activation. Stem Cells Int 2020; 2020:4764012. [PMID: 32508931 PMCID: PMC7246394 DOI: 10.1155/2020/4764012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/24/2020] [Accepted: 05/05/2020] [Indexed: 02/08/2023] Open
Abstract
Although neural stem cells (NSCs) could migrate towards lesions after central nervous system (CNS) injury, the migration ability always is restricted due to the disturbed composition and density of the adhesion ligands and extracellular matrix (ECM) gradient after CNS injury. To date, various methods have been developed to enhance NSC migration and a number of factors, which are affecting NSC migration potential, have been identified. Here, primary NSCs were cultured and the expression of actin alpha 2 (ACTA2) in NSCs was determined using reverse transcription polymerase chain reaction (RT-PCR) and immunostaining. Next, the role of ACTA2 in regulating NSC migration and the potential mechanism was explored. Our results demonstrated that ACTA2 expressed in NSCs. Meanwhile, downregulated ACTA2 using siRNA inhibited NSC migration through hindering actin filament polymerization via increasing RhoA expression and decreasing Rac1 expression. The present study might enrich the basic knowledge of ACTA2 in NSC migration and open an avenue for enhancing NSC migration potential, subsequently providing an intervention target for functional recovery after CNS injury.
Collapse
|
31
|
Zhang K, Yang Y, Ge H, Wang J, Chen X, Lei X, Zhong J, Zhang C, Xian J, Lu Y, Tan L, Feng H. Artesunate promotes the proliferation of neural stem/progenitor cells and alleviates Ischemia-reperfusion Injury through PI3K/Akt/FOXO-3a/p27 kip1 signaling pathway. Aging (Albany NY) 2020; 12:8029-8048. [PMID: 32379706 PMCID: PMC7244066 DOI: 10.18632/aging.103121] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/24/2020] [Indexed: 01/02/2023]
Abstract
Stroke is one of the leading causes of death worldwide that also result in long-term disability. Endogenous neural stem/progenitor cells (NSPCs) within subventricular (SVZ) and dentate gyrus (DG) zone, stimulated by cerebral infarction, can promote neural function recovery. However, the proliferation of eNSPCs triggered by ischemia is not enough to induce neural repair, which may contribute to the permanent disability in stroke patients. In this study, our results showed that following the treatment with artesunate (ART, 150 mg/kg), the functional recovery was significantly improved, the infarct volume was notably reduced, and the expression of Nestin, a proliferation marker of NSPCs in the infarcted cortex, was also increased. Additionally, the proliferative activity of NSPCs with or without oxygen-glucose deprivation/reperfusion was significantly promoted by ART treatment, and the therapeutic concentration was 0.8 μmol/L (without OGD/R) or 0.4 μmol/L (with OGD/R) in the in vitro model. Furthermore, the effects of ART can be abolished by the treatment of PI3K inhibitor wortmannin. The expression levels of related molecules in PI3K/Akt/FOXO-3a/p27kip1 signaling pathway (p-AKT, p-FOXO-3a, p27kip1) were examined using western blotting. The results suggested ART could inhibit the transcriptional function of FOXO-3a by inducing its phosphorylation, subsequently downregulating p27kip1 and enhancing neural stem cell proliferation in the infarcted cortex via PI3K/AKT signaling, further alleviating ischemia-reperfusion injury after ischemic stroke.
Collapse
Affiliation(s)
- Kaiyuan Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Yang Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Hongfei Ge
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Ju Wang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Xuezhu Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Xuejiao Lei
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Jun Zhong
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Chao Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Jishu Xian
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Yongling Lu
- Clinical Research Center, The Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Liang Tan
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing, China
| |
Collapse
|
32
|
Setien MB, Smith KR, Howard K, Williams K, Suhr ST, Purcell EK. Differentiation and characterization of neurons derived from rat iPSCs. J Neurosci Methods 2020; 338:108693. [DOI: 10.1016/j.jneumeth.2020.108693] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 12/26/2022]
|
33
|
Liu D, Pavathuparambil Abdul Manaph N, Al-Hawwas M, Bobrovskaya L, Xiong LL, Zhou XF. Coating Materials for Neural Stem/Progenitor Cell Culture and Differentiation. Stem Cells Dev 2020; 29:463-474. [PMID: 32106778 DOI: 10.1089/scd.2019.0288] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Neural stem/progenitor cells (NSPCs) have a potential to treat various neurological diseases, such as Parkinson's Disease, Alzheimer's Disease, and Spinal Cord Injury. However, the limitation of NSPC sources and the difficulty to maintain their stemness or to differentiate them into specific therapeutic cells are the main hurdles for clinical research and application. Thus, for obtaining a therapeutically relevant number of NSPCs in vitro, it is important to understand factors regulating their behaviors and to establish a protocol for stable NSPC proliferation and differentiation. Coating materials for cell culture, such as Matrigel, laminin, collagen, and other coating materials, can significantly affect NSPC characteristics. This article provides a review of coating materials for NSPC culturing in both two dimensions and three dimensions, and their functions in NSPC proliferation and differentiation, and presents a useful guide to select coating materials for researchers.
Collapse
Affiliation(s)
- Donghui Liu
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | | | - Mohammed Al-Hawwas
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Larisa Bobrovskaya
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Liu-Lin Xiong
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
34
|
Conejos-Sánchez I, Gallon E, Niño-Pariente A, Smith JA, De la Fuente AG, Di Canio L, Pluchino S, Franklin RJM, Vicent MJ. Polyornithine-based polyplexes to boost effective gene silencing in CNS disorders. NANOSCALE 2020; 12:6285-6299. [PMID: 31840717 DOI: 10.1039/c9nr06187h] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Gene silencing therapies have successfully suppressed the translation of target proteins, a strategy that holds great promise for the treatment of central nervous system (CNS) disorders. Advances in the current knowledge on multimolecular delivery vehicles are concentrated on overcoming the difficulties in delivery of small interfering (si)RNA to target tissues, which include anatomical accessibility, slow diffusion, safety concerns, and the requirement for specific cell uptake within the unique environment of the CNS. The present work addressed these challenges through the implementation of polyornithine derivatives in the construction of polyplexes used as non-viral siRNA delivery vectors. Physicochemical and biological characterization revealed biodegradability and biocompatibility of our polyornithine-based system and the ability to silence gene expression in primary oligodendrocyte progenitor cells (OPCs) effectively. In summary, the well-defined properties and neurological compatibility of this polypeptide-based platform highlight its potential utility in the treatment of CNS disorders.
Collapse
Affiliation(s)
- I Conejos-Sánchez
- Centro de Investigación Príncipe Felipe. Polymer Therapeutics Laboratory, C/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Harberts J, Haferkamp U, Haugg S, Fendler C, Lam D, Zierold R, Pless O, Blick RH. Interfacing human induced pluripotent stem cell-derived neurons with designed nanowire arrays as a future platform for medical applications. Biomater Sci 2020; 8:2434-2446. [DOI: 10.1039/d0bm00182a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanostructured substrates such as nanowire arrays form a powerful tool for building next-generation medical devices.
Collapse
Affiliation(s)
- Jann Harberts
- Center for Hybrid Nanostructures
- Universität Hamburg
- 22761 Hamburg
- Germany
| | | | - Stefanie Haugg
- Center for Hybrid Nanostructures
- Universität Hamburg
- 22761 Hamburg
- Germany
| | - Cornelius Fendler
- Center for Hybrid Nanostructures
- Universität Hamburg
- 22761 Hamburg
- Germany
| | - Dennis Lam
- Fraunhofer IME ScreeningPort
- 22525 Hamburg
- Germany
| | - Robert Zierold
- Center for Hybrid Nanostructures
- Universität Hamburg
- 22761 Hamburg
- Germany
| | - Ole Pless
- Fraunhofer IME ScreeningPort
- 22525 Hamburg
- Germany
| | - Robert H. Blick
- Center for Hybrid Nanostructures
- Universität Hamburg
- 22761 Hamburg
- Germany
- Material Science and Engineering
| |
Collapse
|
36
|
Cohen-Carmon D, Sorek M, Lerner V, Divya MS, Nissim-Rafinia M, Yarom Y, Meshorer E. Progerin-Induced Transcriptional Changes in Huntington's Disease Human Pluripotent Stem Cell-Derived Neurons. Mol Neurobiol 2019; 57:1768-1777. [PMID: 31834602 DOI: 10.1007/s12035-019-01839-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/14/2019] [Indexed: 01/08/2023]
Abstract
Huntington's disease (HD) is a neurodegenerative late-onset genetic disorder caused by CAG expansions in the coding region of the Huntingtin (HTT) gene, resulting in a poly-glutamine (polyQ) expanded HTT protein. Considerable efforts have been devoted for studying HD and other polyQ diseases using animal models and cell culture systems, but no treatment currently exists. Human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) offer an elegant solution for modeling human diseases. However, as embryonic or rejuvenated cells, respectively, these pluripotent stem cells (PSCs) do not recapitulate the late-onset feature of the disease. Here, we applied a robust and rapid differentiation protocol to derive electrophysiologically active striatal GABAergic neurons from human wild-type (WT) and HD ESCs and iPSCs. RNA-seq analyses revealed that HD and WT PSC-derived neurons are highly similar in their gene expression patterns. Interestingly, ectopic expression of Progerin in both WT and HD neurons exacerbated the otherwise non-significant changes in gene expression between these cells, revealing IGF1 and genes involved in neurogenesis and nervous system development as consistently altered in the HD cells. This work provides a useful tool for modeling HD in human PSCs and reveals potential molecular targets altered in HD neurons.
Collapse
Affiliation(s)
- Dorit Cohen-Carmon
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, Edmond J. Safra Campus, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Matan Sorek
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, Edmond J. Safra Campus, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel.,The Edmond and Lily Safra Center for Brain Sciences, Edmond J. Safra Campus, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Vitaly Lerner
- The Edmond and Lily Safra Center for Brain Sciences, Edmond J. Safra Campus, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel.,Department of Neurobiology, The Alexander Silberman Institute of Life Sciences, Edmond J. Safra Campus, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Mundackal S Divya
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, Edmond J. Safra Campus, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel.,Department of Pathology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, 695011, Kerala, India
| | - Malka Nissim-Rafinia
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, Edmond J. Safra Campus, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Yosef Yarom
- The Edmond and Lily Safra Center for Brain Sciences, Edmond J. Safra Campus, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel.,Department of Neurobiology, The Alexander Silberman Institute of Life Sciences, Edmond J. Safra Campus, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Eran Meshorer
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, Edmond J. Safra Campus, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel. .,The Edmond and Lily Safra Center for Brain Sciences, Edmond J. Safra Campus, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel.
| |
Collapse
|
37
|
Zhang S, Sun P, Lin K, Chan FHL, Gao Q, Lau WF, Roy VAL, Zhang H, Lai KWC, Huang Z, Yung KKL. Extracellular Nanomatrix-Induced Self-Organization of Neural Stem Cells into Miniature Substantia Nigra-Like Structures with Therapeutic Effects on Parkinsonian Rats. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901822. [PMID: 31871862 PMCID: PMC6918115 DOI: 10.1002/advs.201901822] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/29/2019] [Indexed: 05/14/2023]
Abstract
Substantia nigra (SN) is a complex and critical region of the brain wherein Parkinson's disease (PD) arises from the degeneration of dopaminergic neurons. Miniature SN-like structures (mini-SNLSs) constructed from novel combination of nanomaterials and cell technologies exhibit promise as potentially curative cell therapies for PD. In this work, a rapid self-organization of mini-SNLS, with an organizational structure and neuronal identities similar to those of the SN in vivo, is achieved by differentiating neural stem cells in vitro on biocompatible silica nanozigzags (NZs) sculptured by glancing angle deposition, without traditional chemical growth factors. The differentiated neurons exhibit electrophysiological activity in vitro. Diverse physical cues and signaling pathways that are determined by the nanomatrices and lead to the self-organization of the mini-SNLSs are clarified and elucidated. In vivo, transplantation of the neurons from a mini-SNLS results in an early and progressive amelioration of PD in rats. The sculptured medical device reported here enables the rapid and specific self-organization of region-specific and functional brain-like structures without an undesirable prognosis. This development provides promising and significant insights into the screening of potentially curative drugs and cell therapies for PD.
Collapse
Affiliation(s)
- Shiqing Zhang
- Department of BiologyHong Kong Baptist University (HKBU)Kowloon TongKowloonHong Kong SAR China
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
- HKBU Institute of Research and Continuing Education, 9FThe Industrialization Complex of Shenzhen Virtual University ParkNo. 2 Yuexing 3rd Road, South Zone, Hi‐tech Industrial Park, Nanshan DistrictShenzhen518057Guangdong ProvinceChina
| | - Peng Sun
- Department of PhysicsHKBUKowloon TongKowloonHong Kong SAR China
- Department of Materials Science and EngineeringSouthern University of Science and TechnologyShenzhen518000Guangdong ProvinceChina
| | - Kaili Lin
- Department of BiologyHong Kong Baptist University (HKBU)Kowloon TongKowloonHong Kong SAR China
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
| | - Florence Hiu Ling Chan
- Department of Biomedical EngineeringCity University of Hong Kong (CityU)Tat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Qi Gao
- Department of Biomedical EngineeringCity University of Hong Kong (CityU)Tat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Wai Fung Lau
- Department of PhysicsHKBUKowloon TongKowloonHong Kong SAR China
| | - Vellaisamy A. L. Roy
- Department of Materials Science and EngineeringCity University of Hong KongTat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Hongqi Zhang
- School of Chinese MedicineHKBUKowloon TongKowloonHong Kong SAR China
| | - King Wai Chiu Lai
- Department of Biomedical EngineeringCity University of Hong Kong (CityU)Tat Chee Avenue, Kowloon TongKowloonHong Kong SAR China
| | - Zhifeng Huang
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
- HKBU Institute of Research and Continuing Education, 9FThe Industrialization Complex of Shenzhen Virtual University ParkNo. 2 Yuexing 3rd Road, South Zone, Hi‐tech Industrial Park, Nanshan DistrictShenzhen518057Guangdong ProvinceChina
- Department of PhysicsHKBUKowloon TongKowloonHong Kong SAR China
- Institute of Advanced MaterialsState Key Laboratory of Environmental and Biological AnalysisHKBUKowloon TongKowloonHong Kong SAR China
| | - Ken Kin Lam Yung
- Department of BiologyHong Kong Baptist University (HKBU)Kowloon TongKowloonHong Kong SAR China
- Golden Meditech Center for NeuroRegeneration SciencesHKBUKowloon TongKowloonHong Kong SAR China
- HKBU Institute of Research and Continuing Education, 9FThe Industrialization Complex of Shenzhen Virtual University ParkNo. 2 Yuexing 3rd Road, South Zone, Hi‐tech Industrial Park, Nanshan DistrictShenzhen518057Guangdong ProvinceChina
- Institute of Advanced MaterialsState Key Laboratory of Environmental and Biological AnalysisHKBUKowloon TongKowloonHong Kong SAR China
| |
Collapse
|
38
|
Yang Y, Zhang K, Chen X, Wang J, Lei X, Zhong J, Xian J, Quan Y, Lu Y, Huang Q, Chen J, Ge H, Feng H. SVCT2 Promotes Neural Stem/Progenitor Cells Migration Through Activating CDC42 After Ischemic Stroke. Front Cell Neurosci 2019; 13:429. [PMID: 31607868 PMCID: PMC6761321 DOI: 10.3389/fncel.2019.00429] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/06/2019] [Indexed: 12/31/2022] Open
Abstract
Ischemic stroke is one of the most leading diseases causing death/long-term disability worldwide. Activating endogenous neural stem/progenitors cells (NSPCs), lining in the subventricular zone (SVZ) and dentate gyrus, facilitates injured brain tissue recovery in both short and long-term experimental settings. While, only a few proliferated NSPCs migrate toward the lesions to enhance endogenous repair after ischemia. Here, the results indicated that the functional recovery was evidently improved and the infarct volume was significantly reduced with ascorbic acid (AA) treatment in a dose-dependent manner from 125 to 500 mg/Kg, and the suitable therapeutic concentration was 250 mg/Kg. The possible mechanism might be due to activating sodium-vitamin C cotransporter 2 (SVCT2), which was down-regulated in SVZ after ischemia. Furthermore, immunostaining images depicted the number of migrated NSPCs from SVZ were significantly increased with 250 mg/Kg AA treatment or SVCT2 overexpression under the physiological and pathological condition in vivo. Besides, the data also represented that 250 mg/Kg AA or SVCT2 overexpression facilitated NSPCs migration via promoting F-actin assembling in the manner of up-regulating CDC42 expression using oxygen-glucose deprivation in vitro. Collectively, the present study indicates that SVCT2 promotes NSPCs migration through CDC42 activation to facilitate F-actin assembling, which enlarges the therapeutic scope of AA and the role of SVCT2 in NSPCs migration after brain injury.
Collapse
Affiliation(s)
- Yang Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Kaiyuan Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Xuezhu Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Ju Wang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Xuejiao Lei
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Jun Zhong
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Jishu Xian
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Yulian Quan
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Yongling Lu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Qianying Huang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Jingyu Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Hongfei Ge
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| |
Collapse
|
39
|
Patel BB, Clark KL, Kozik EM, Dash L, Kuhlman JA, Sakaguchi DS. Isolation and culture of primary embryonic zebrafish neural tissue. J Neurosci Methods 2019; 328:108419. [PMID: 31472190 DOI: 10.1016/j.jneumeth.2019.108419] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/23/2019] [Accepted: 08/27/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Primary cell culture is a valuable tool to utilize in parallel with in vivo studies in order to maximize our understanding of the mechanisms surrounding neurogenesis and central nervous system (CNS) regeneration and plasticity. The zebrafish is an important model for biomedical research and primary neural cells are readily obtainable from their embryonic stages viatissue dissociation. Further, transgenic reporter lines with cell type-specific expression allows for observation of distinct cell populations within the dissociated tissue. NEW METHOD Here, we define an efficient method for ex vivo quantification and characterization of neuronal and glial tissue dissociated from embryonic zebrafish. RESULTS Zebrafish brain dissociated cells have been documented to survive in culture for at least 9 days in vitro (div). Anti-HuC/D and anti-Acetylated Tubulin antibodies were used to identify neurons in culture; at 3 div approximately 48% of cells were HuC/D positive and 85% expressed serotonin, suggesting our protocol can efficiently isolate neurons from whole embryonic zebrafish brains. Live time-lapse imaging was also carried out to analyze cell migration in vitro. COMPARISON WITH EXISTING METHODS Primary cultures of zebrafish neural cells typically have low rates of survivability in vitro. We have developed a culture system that has long term cell viability, enabling direct analysis of cell-cell and cell-extracellular matrix interactions. CONCLUSIONS These results demonstrate a practical method for isolating, dissociating and culturing of embryonic zebrafish neural tissue. This approach could further be utilized to better understand zebrafish regeneration in vitro.
Collapse
Affiliation(s)
- Bhavika B Patel
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, United States; Neuroscience Program, United States
| | - Kendra L Clark
- Department of Animal Science, Iowa State University, Ames, IA 50011, United States; Genetics and Genomics Program, United States
| | - Emily M Kozik
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, United States
| | - Linkan Dash
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, United States; Genetics and Genomics Program, United States
| | - Julie A Kuhlman
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, United States.
| | - Donald S Sakaguchi
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA 50011, United States; Neuroscience Program, United States.
| |
Collapse
|
40
|
Cai Z, Gan Y, Bao C, Wu W, Wang X, Zhang Z, Zhou Q, Lin Q, Yang Y, Zhu L. Photosensitive Hydrogel Creates Favorable Biologic Niches to Promote Spinal Cord Injury Repair. Adv Healthc Mater 2019; 8:e1900013. [PMID: 31074122 DOI: 10.1002/adhm.201900013] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/21/2019] [Indexed: 12/17/2022]
Abstract
Photochemistry is considered to be a promising strategy for hydrogels to mimic the complex and dynamic properties of natural extracellular matrix. However, it is seldom applied in 3D tissue engineering and regenerative medicine due to the attenuation of light. In this study, phenyl azide photchemistry and optical fiber technology are first used to localize adhesive protein on the inner surface of the nerve guidance conduit in a 3D hydrogel scaffold. In vitro coculture assay of neural stem cells (NSCs) shows that photoimmobilization of collagen significantly improves the adhesion and survival of NSCs in the conduit, and exhibits synergistic effect with the sustainable release of growth factor. After implantation in transected spinal cord, the optimized hydrogel scaffold is found to improve the locomotion recovery of rats 12 weeks after spinal cord injury (SCI). Histological analysis suggests that the designed hydrogel scaffold provides a favorable biological niche for neuronal regeneration, thus producing directional neuron tissue and promoting the repair of SCI. This study demonstrates a promising hydrogel scaffold for SCI repair and provides the first understanding of the photoimmobilization of adhesive protein in a 3D hydrogel conduit concerning its functions on spinal cord tissue restoration.
Collapse
Affiliation(s)
- Zhengwei Cai
- Optogenetics and Synthetic Biology Interdisciplinary Research CenterState Key Laboratory of Bioreactor EngineeringEast China University of Science and Technology 130# Meilong Road Shanghai 200237 China
| | - Yibo Gan
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest HospitalThird Military Medical University (Army Medical University) 29# Gao Tan Yan Street Chongqing 400038 P. R. China
- Institute of Rocket Force MedicineState Key Laboratory of TraumaBurns and Combined InjuryThird Military Medical University (Army Medical University) 30# Gao Tan Yan Street Chongqing 400038 P. R. China
| | - Chunyan Bao
- Optogenetics and Synthetic Biology Interdisciplinary Research CenterState Key Laboratory of Bioreactor EngineeringEast China University of Science and Technology 130# Meilong Road Shanghai 200237 China
| | - Wanjiang Wu
- National Department of Neurosurgery and Key Laboratory of NeurotraumaSouthwest HospitalThird Military Medical University (Army Medical University) 29# Gao Tan Yan Street Chongqing 400038 P. R. China
| | - Xuebin Wang
- Optogenetics and Synthetic Biology Interdisciplinary Research CenterState Key Laboratory of Bioreactor EngineeringEast China University of Science and Technology 130# Meilong Road Shanghai 200237 China
| | - Zetong Zhang
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest HospitalThird Military Medical University (Army Medical University) 29# Gao Tan Yan Street Chongqing 400038 P. R. China
| | - Qiang Zhou
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest HospitalThird Military Medical University (Army Medical University) 29# Gao Tan Yan Street Chongqing 400038 P. R. China
- Bone and Trauma CenterThe Third Affiliated Hospital of Chongqing Medical University (Gener Hospital) Chongqing 401120 China
| | - Qiuning Lin
- Optogenetics and Synthetic Biology Interdisciplinary Research CenterState Key Laboratory of Bioreactor EngineeringEast China University of Science and Technology 130# Meilong Road Shanghai 200237 China
| | - Yi Yang
- Optogenetics and Synthetic Biology Interdisciplinary Research CenterState Key Laboratory of Bioreactor EngineeringEast China University of Science and Technology 130# Meilong Road Shanghai 200237 China
| | - Linyong Zhu
- Optogenetics and Synthetic Biology Interdisciplinary Research CenterState Key Laboratory of Bioreactor EngineeringEast China University of Science and Technology 130# Meilong Road Shanghai 200237 China
| |
Collapse
|
41
|
Zhong J, Lan C, Zhang C, Yang Y, Chen WX, Zhang KY, Zhao HL, Fang XY, Li HH, Tan L, Wang P, Ge HF, Hu R, Feng H. Chondroitin sulfate proteoglycan represses neural stem/progenitor cells migration via PTPσ/α-actinin4 signaling pathway. J Cell Biochem 2019; 120:11008-11021. [PMID: 30688376 DOI: 10.1002/jcb.28379] [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] [Received: 04/16/2018] [Accepted: 12/06/2018] [Indexed: 01/24/2023]
Abstract
Neural stem/progenitor cells (NSPCs) are a promising candidate for the cell-replacement therapy after central nervous system (CNS) injury. However, the short of sufficient NSPCs migration and integration into the lesions is an essential challenge for cell-based therapy after CNS injury due to the disturbance of local environmental homeostasis. Chondroitin sulfate proteoglycan (CSPG) is obviously accumulated at the lesions and destroyed local homeostasis after CNS injury. The previous study has demonstrated that the CSPG is a dominating ingredient inhibiting axonal regrowth of newly born neurons after CNS injury. NSPCs, a strain of special neural subtypes, hold the capacity of leading processes formation to regulate NSPCs migration, which has the same mechanism as axonal regrowth. Hence, it is worth investigating the effect of CSPG on NSPCs migration and its underlying mechanism. Here, different concentration of CSPG was used to evaluate its effect on NSPCs migration. The results showed that the CSPG suppressed NSPCs migration in a dose-dependent manner from 10 to 80 µg/mL with phase-contrast microscopy after 24 hours. Meanwhile, transwell assays were performed to certify the above results. Our data indicated that the 40 µg/mL CSPG obviously suppressed NSPCs migration via decreasing filopodia formation using immunofluorescence staining. Furthermore, data indicated that the 40 µg/mL CSPG upregulated protein tyrosine phosphatase receptor σ (PTPσ) expression and decreased α-actinin4 (ACTN4) expression through immunofluorescence, reverse transcription polymerase chain reaction, and Western blot assays. While the inhibitory effect was attenuated using PTPσ-specific small interfering RNA. In addition, data demonstrated that the 40 µg/mL CSPG facilitated NSPCs differentiation into glial fibrillary acidic protein-positive cells and inhibited NSPCs directing into MAP2- and MBP-positive cells. Collectively, these data demonstrated that the CSPG suppressed NSPCs migration through PTPσ/ACTN4 signaling pathway. Meanwhile, CSPG facilitated NSPCs differentiation into astrocytes and inhibited NSPCs directing into neurons and oligodendrocytes.
Collapse
Affiliation(s)
- Jun Zhong
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chuan Lan
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chao Zhang
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yang Yang
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wei-Xiang Chen
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Kai-Yuan Zhang
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Heng-Li Zhao
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuan-Yu Fang
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Huan-Huan Li
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Liang Tan
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Pan Wang
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hong-Fei Ge
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Rong Hu
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- Key Laboratory of Neurotrauma, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
42
|
Pacitti D, Privolizzi R, Bax BE. Organs to Cells and Cells to Organoids: The Evolution of in vitro Central Nervous System Modelling. Front Cell Neurosci 2019; 13:129. [PMID: 31024259 PMCID: PMC6465581 DOI: 10.3389/fncel.2019.00129] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/14/2019] [Indexed: 02/05/2023] Open
Abstract
With 100 billion neurons and 100 trillion synapses, the human brain is not just the most complex organ in the human body, but has also been described as "the most complex thing in the universe." The limited availability of human living brain tissue for the study of neurogenesis, neural processes and neurological disorders has resulted in more than a century-long strive from researchers worldwide to model the central nervous system (CNS) and dissect both its striking physiology and enigmatic pathophysiology. The invaluable knowledge gained with the use of animal models and post mortem human tissue remains limited to cross-species similarities and structural features, respectively. The advent of human induced pluripotent stem cell (hiPSC) and 3-D organoid technologies has revolutionised the approach to the study of human brain and CNS in vitro, presenting great potential for disease modelling and translational adoption in drug screening and regenerative medicine, also contributing beneficially to clinical research. We have surveyed more than 100 years of research in CNS modelling and provide in this review an historical excursus of its evolution, from early neural tissue explants and organotypic cultures, to 2-D patient-derived cell monolayers, to the latest development of 3-D cerebral organoids. We have generated a comprehensive summary of CNS modelling techniques and approaches, protocol refinements throughout the course of decades and developments in the study of specific neuropathologies. Current limitations and caveats such as clonal variation, developmental stage, validation of pluripotency and chromosomal stability, functional assessment, reproducibility, accuracy and scalability of these models are also discussed.
Collapse
Affiliation(s)
- Dario Pacitti
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, United Kingdom
- College of Medicine and Health, St Luke’s Campus, University of Exeter, Exeter, United Kingdom
| | - Riccardo Privolizzi
- Gene Transfer Technology Group, Institute for Women’s Health, University College London, London, United Kingdom
| | - Bridget E. Bax
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London, United Kingdom
| |
Collapse
|
43
|
G protein-coupled estrogen receptor 1 negatively regulates the proliferation of mouse-derived neural stem/progenitor cells via extracellular signal-regulated kinase pathway. Brain Res 2019; 1714:158-165. [PMID: 30797747 DOI: 10.1016/j.brainres.2019.02.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 12/20/2022]
Abstract
G protein-coupled estrogen receptor 1 (GPER1, also known as GPR30) has been reported to play a wide range of function in the central nervous system (CNS). However, whether GPER1 is expressed by neural stem/progenitor cells (NSPCs) and its role has not been established. Here, we found the expression of GPER1 in mouse-derived NSPCs via western blot and immunofluorescent staining. Moreover, we revealed that specific activation of GPER1 by the agonist G1 decreased the proliferation of NSPCs in a dose-dependent manner. The neurosphere formation assay and Ki67 staining further demonstrated that activation of GPER1 inhibited the proliferation of NSPCs. Additionally, the inhibitory effect of G1 on the proliferation of NSPCs could be blocked by the specific GPER1 antagonist G15. Intriguingly, ERK pathway was involved in the negative effect of GPER1 on the proliferation of NSPCs, because the phosphorylation level of ERK in NSPCs was remarkably decreased during G1 treatment. However, the antagonist G15 reversed the down-regulated level of p-ERK. Knock-down GPER1 also reversed the inhibitory effect of G1 on NSPCs proliferation. Together, our results provide the first evidence that GPER1 is expressed by NSPCs and its activation negatively modulates the proliferation of NSPCs, highlighting the importance of GPER1 in regulating NSPC behaviors.
Collapse
|
44
|
An accessible and high-throughput strategy of continuously monitoring apoptosis by fluorescent detection of caspase activation. Anal Biochem 2018; 564-565:96-101. [PMID: 30365977 DOI: 10.1016/j.ab.2018.10.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/28/2018] [Accepted: 10/21/2018] [Indexed: 01/05/2023]
Abstract
We present a real-time, high-throughput, and cost-effective method of detecting apoptosis in vitro using a previously developed reagent that detects caspase activation by fluorescence. Current methods of assessing apoptosis fail to account for the dimension of time, and thus are limited in data yielded per sample. This reagent allows real-time detection of apoptosis, but until now has been restricted to a costly automated detection system. Here, we describe apoptosis detection with the Essen Bioscience IncuCyte® Caspase-3/7 Reagent using a multimode microplate reader, a common instrument in biological laboratories, which may be used prior to or in lieu of the automated system. This modified microplate reader apoptosis assay was validated against the established automated system, and was shown to detect a strong dose-response relationship (automated system r2 = 0.9968, microplate reader r2 = 0.9924). We also propose a quick and reliable method of quantifying cell density by Hoechst 33342 nuclear staining in microplates (r2 = 0.8812 between Hoechst signal and cell density). We assert that the dimension of time should not be overlooked, and that the method presented here is an accessible strategy for many researchers due to low startup cost and precise detection of apoptosis in real time.
Collapse
|
45
|
Cai D, Fan J, Wang S, Long R, Zhou X, Liu Y. Primary biocompatibility tests of poly(lactide-co-glycolide)-(poly-L-orithine/fucoidan) core-shell nanocarriers. ROYAL SOCIETY OPEN SCIENCE 2018; 5:180320. [PMID: 30109086 PMCID: PMC6083702 DOI: 10.1098/rsos.180320] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 06/20/2018] [Indexed: 06/08/2023]
Abstract
Layer-by-layer (LbL) self-assembly is the technology used in intermolecular static electricity, hydrogen bonds, covalent bonds and other polymer interactions during film assembling. This technology has been widely studied in the drug carrier field. Given their use in drug delivery systems, the biocompatibility of these potential compounds should be addressed. In this work, the primary biocompatibility of poly(lactide-co-glycolide)-(poly-L-orithine/fucoidan) [PLGA-(PLO/fucoidan)] core-shell nanoparticles (NPs) was investigated. Atomic force microscopy revealed the PLGA-(PLO/Fucoidan)4 NPs to be spherical, with a uniform size distribution and a smooth surface, and the NPs were stable in physiological saline. The residual amount of methylene chloride was further determined by headspace gas chromatography, in which the organic solvent can be volatilized during preparation. Furthermore, cell viability, acridine orange/ethidium bromide staining, haemolysis and mouse systemic toxicity were all assessed to show that PLGA-(PLO/fucoidan)4 NPs were biocompatible with cells and mice. Therefore, these NPs are expected to have potential applications in future drug delivery systems.
Collapse
Affiliation(s)
- Duanhua Cai
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, People's Republic of China
| | - Jingqian Fan
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, People's Republic of China
| | - Shibin Wang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, People's Republic of China
- Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, People's Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, People's Republic of China
| | - Ruimin Long
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, People's Republic of China
| | - Xia Zhou
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, People's Republic of China
| | - Yuangang Liu
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, People's Republic of China
- Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, People's Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, People's Republic of China
| |
Collapse
|
46
|
Abstract
Introduction Hereditary multiple exostoses (HME) is a rare congenital pediatric disorder characterized by osteochondromas forming next to the growth plates in young patients. The osteochondromas cause multiple health problems that include skeletal deformities and chronic pain. Surgery is used to remove the most symptomatic osteochondromas but because of their large number, many are left in place, causing life-long problems and increasing the probability of malignant transformation. There is no other treatment to prevent or reduce osteochondromas formation at present. Areas covered Recent studies reviewable through PubMed are providing new insights into cellular and molecular mechanisms of osteochondroma development. The resulting data are suggesting rational and plausible new therapeutic strategies for osteochondroma prevention some of which are being tested in HME animal models and one of which is part of a just announced clinical trial. Expert Commentary This section summarizes and evaluates such strategies and points also to possible future alternatives.
Collapse
Affiliation(s)
- Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104
| |
Collapse
|
47
|
Centeno EGZ, Cimarosti H, Bithell A. 2D versus 3D human induced pluripotent stem cell-derived cultures for neurodegenerative disease modelling. Mol Neurodegener 2018; 13:27. [PMID: 29788997 PMCID: PMC5964712 DOI: 10.1186/s13024-018-0258-4] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 05/08/2018] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS), affect millions of people every year and so far, there are no therapeutic cures available. Even though animal and histological models have been of great aid in understanding disease mechanisms and identifying possible therapeutic strategies, in order to find disease-modifying solutions there is still a critical need for systems that can provide more predictive and physiologically relevant results. One possible avenue is the development of patient-derived models, e.g. by reprogramming patient somatic cells into human induced pluripotent stem cells (hiPSCs), which can then be differentiated into any cell type for modelling. These systems contain key genetic information from the donors, and therefore have enormous potential as tools in the investigation of pathological mechanisms underlying disease phenotype, and progression, as well as in drug testing platforms. hiPSCs have been widely cultured in 2D systems, but in order to mimic human brain complexity, 3D models have been proposed as a more advanced alternative. This review will focus on the use of patient-derived hiPSCs to model AD, PD, HD and ALS. In brief, we will cover the available stem cells, types of 2D and 3D culture systems, existing models for neurodegenerative diseases, obstacles to model these diseases in vitro, and current perspectives in the field.
Collapse
Affiliation(s)
- Eduarda G Z Centeno
- Department of Biotechnology, Federal University of Pelotas, Campus Capão do Leão, Pelotas, RS, 96160-000, Brazil.,Department of Pharmacology, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Helena Cimarosti
- Department of Pharmacology, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| | - Angela Bithell
- School of Pharmacy, University of Reading, Whiteknights Campus, Reading, RG6 6UB, UK.
| |
Collapse
|
48
|
Fan WL, Liu P, Wang G, Pu JG, Xue X, Zhao JH. Transplantation of hypoxic preconditioned neural stem cells benefits functional recovery via enhancing neurotrophic secretion after spinal cord injury in rats. J Cell Biochem 2018; 119:4339-4351. [PMID: 28884834 DOI: 10.1002/jcb.26397] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/30/2017] [Indexed: 12/25/2022]
Abstract
Spinal cord injury (SCI) is a debilitating, costly, and common pathological condition that affects the function of central nervous system (CNS). To date, there are few promising therapeutic strategies available for SCI. To look for a suitable therapeutic strategy, we have developed a sublethal hypoxic preconditioning procedure using Fluorescence-activated cell sorting (FACS) analysis, LDH releasing, and cell viability assays in vitro. Meanwhile, we have examined the benefits of neural stem cells (NSCs) transplantation prior to hypoxic preconditioning on functional recovery and potential mechanism via MRI screening, H&E, and Nissl staining, immunofluorescence staining and Elisa assays. Our data showed that transplantation of hypoxic prconditioned NSCs could enhance neuronal survival, especially 5-TH+ and ChAT+ neurons, in the injured spinal cord to reinforce functional benefits. The hypoxia exposure upregulated HIF-1α, neurotrophic and growth factors including neurotrophin-3 (NT-3), glial cell-derived neurotrophic factor (GDNF), and brain-derived neurotrophic factor (BDNF) in vitro and in vivo. Furthermore, functional recovery, including locomotor and hypersensitivities to mechanical and thermal stimulation assessed via behavioral and sensory tests, improved significantly in rats with engraftment of NSCs after hypoxia exposure from day 14 post-SCI, compared with the control and N-NSCs groups. In short, the approach employed in this study could result in functional recovery via upregulating neurotrophic and growth factors, which implies that hypoxic preconditioning strategy could serve as an effective and feasible strategy for cell-based therapy in the treatment of SCI in rats.
Collapse
Affiliation(s)
- Wei-Li Fan
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, The Third Military Medical University, Chongqing, China
| | - Peng Liu
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, The Third Military Medical University, Chongqing, China
| | - Guan Wang
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, The Third Military Medical University, Chongqing, China
| | - Jun-Gang Pu
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, The Third Military Medical University, Chongqing, China
| | - Xin Xue
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, The Third Military Medical University, Chongqing, China
| | - Jian-Hua Zhao
- Department of Spinal Surgery, Daping Hospital, Research Institute of Surgery, The Third Military Medical University, Chongqing, China
| |
Collapse
|
49
|
Cha KJ, Kong SY, Lee JS, Kim HW, Shin JY, La M, Han BW, Kim DS, Kim HJ. Cell density-dependent differential proliferation of neural stem cells on omnidirectional nanopore-arrayed surface. Sci Rep 2017; 7:13077. [PMID: 29026125 PMCID: PMC5638797 DOI: 10.1038/s41598-017-13372-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 09/22/2017] [Indexed: 11/09/2022] Open
Abstract
Recently, the importance of surface nanotopography in the determination of stem cell fate and behavior has been revealed. In the current study, we generated polystyrene cell-culture dishes with an omnidirectional nanopore arrayed surface (ONAS) (diameter: 200 nm, depth: 500 nm, center-to-center distance: 500 nm) and investigated the effects of nanotopography on rat neural stem cells (NSCs). NSCs cultured on ONAS proliferated better than those on the flat surface when cell density was low and showed less spontaneous differentiation during proliferation in the presence of mitogens. Interestingly, NSCs cultured on ONAS at clonal density demonstrated a propensity to generate neurospheres, whereas those on the flat surface migrated out, proliferated as individuals, and spread out to attach to the surface. However, the differential patterns of proliferation were cell density-dependent since the distinct phenomena were lost when cell density was increased. ONAS modulated cytoskeletal reorganization and inhibited formation of focal adhesion, which is generally observed in NSCs grown on flat surfaces. ONAS appeared to reinforce NSC-NSC interaction, restricted individual cell migration and prohibited NSC attachment to the nanopore surface. These data demonstrate that ONAS maintains NSCs as undifferentiated while retaining multipotency and is a better topography for culturing low density NSCs.
Collapse
Affiliation(s)
- Kyoung Je Cha
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31 Hyoja-dong Nam-gu, Pohang, 790-784, South Korea.,Ultimate Fabrication Technology Group, Korea Institute of Industrial Technology (KITECH), Techno sunhwan-ro Yuga-myeon Dalseong-gun, Deagu, 711-880, South Korea
| | - Sun-Young Kong
- Laboratory of Molecular and Stem Cell Pharmacology, College of Pharmacy, Chung-Ang University, 221 Heukseok-dong Dongjak-gu, Seoul, 156-756, South Korea
| | - Ji Soo Lee
- Laboratory of Molecular and Stem Cell Pharmacology, College of Pharmacy, Chung-Ang University, 221 Heukseok-dong Dongjak-gu, Seoul, 156-756, South Korea
| | - Hyung Woo Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31 Hyoja-dong Nam-gu, Pohang, 790-784, South Korea
| | - Jae-Yeon Shin
- Laboratory of Molecular and Stem Cell Pharmacology, College of Pharmacy, Chung-Ang University, 221 Heukseok-dong Dongjak-gu, Seoul, 156-756, South Korea
| | - Moonwoo La
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31 Hyoja-dong Nam-gu, Pohang, 790-784, South Korea.,Molds & Dies R&D Group, Korea Institute of Industrial Technology (KITECH), 156 Gaetbeol-ro, Yeonsu-gu, Incheon, 406-840, South Korea
| | - Byung Woo Han
- Department of Biochemistry, College of pharmacy, Seoul National University, San 56-1 Sillim-dong Gwanak-gu, Seoul, 151-742, South Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), San 31 Hyoja-dong Nam-gu, Pohang, 790-784, South Korea.
| | - Hyun-Jung Kim
- Laboratory of Molecular and Stem Cell Pharmacology, College of Pharmacy, Chung-Ang University, 221 Heukseok-dong Dongjak-gu, Seoul, 156-756, South Korea.
| |
Collapse
|
50
|
Yuan J, Ge H, Liu W, Zhu H, Chen Y, Zhang X, Yang Y, Yin Y, Chen W, Wu W, Yang Y, Lin J. M2 microglia promotes neurogenesis and oligodendrogenesis from neural stem/progenitor cells via the PPARγ signaling pathway. Oncotarget 2017; 8:19855-19865. [PMID: 28423639 PMCID: PMC5386728 DOI: 10.18632/oncotarget.15774] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 01/23/2017] [Indexed: 12/20/2022] Open
Abstract
Neural stem/progenitor cells (NSPCs) are an important source of cells for cell replacement therapy after nerve injury. How to induce NSPCs differentiation towards neurons and oligodendrocytes is a challenging issue in neuroscience research. In the present study, we polarized microglia into M1 and M2 phenotype, used their supernatants to induce NSPCs differentiation, and investigated the effects of different microglia phenotypes on NSPCs differentiation and their mechanisms. We discovered that, after exposure to M1 phenotype supernatant, NSPCs differentiated into fewer Tuj-1+ and Olig2+ cells, but more GFAP+ cells. Meanwhile, a significantly increased number of Tuj-1+ and Olig2+ cells and smaller number of GFAP+ cells were generated by M2 microglia supernatant-induced NSPCs differentiation. We also observed that 15d-PGJ2, an endogenous ligand of PPARγ, was elevated in M2 phenotype supernatant and could activate PPARγ expression in NSPCs, whereas use of the PPARγ inhibitor GW9662, could reduce the percentage of differentiated neurons and oligodendrocytes. Our study results confirm that M2 microglia supernatant can activate the PPARγ signaling pathway and promote neurogenesis and oligodendrogenesis from NSPCs differentiation. The present study provides a further theoretical basis for induction of NSPCs oriented differentiation.
Collapse
Affiliation(s)
- Jichao Yuan
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.,Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Hongfei Ge
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei Liu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Haitao Zhu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yaxing Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xuan Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yang Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yi Yin
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Weixiang Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wanjiang Wu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yunfeng Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jiangkai Lin
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|