1
|
Godovalova O, Proshchina A, Kharlamova A, Barabanov V, Krivova Y, Junemann O, Shahina M, Saveliev S. Heterogeneity in the formation of primary and secondary visual fields during human prenatal development. Biol Res 2024; 57:93. [PMID: 39609712 PMCID: PMC11603890 DOI: 10.1186/s40659-024-00576-0] [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: 03/05/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
The human neocortex has a huge surface area with unique cytoarchitectonics, most of which is concealed in sulci. Some cytoarchitectonic fields are associated with macroscopic landmarks. In particular, the primary visual field 17 is associated with the calcarine sulcus. During the prenatal development of the human brain, neocortical gyri and sulci undergo changes and modifications after primary formation. To explore the morphogenetic processes in visual fields during the formation of the primary (provisional) and secondary (permanent) sulci, the occipital lobe of the human fetal brain was studied using immunohistochemical methods. The distribution of various glial and neuronal markers (S-100, β-III-tubulin, NeuN, reelin) in the calcarine sulcus and parietooccipital sulcus was compared. The heterogeneity in the formation of primary and secondary visual fields was demonstrated. The study revealed that the development of the primary visual field 17, linked with the calcarine sulcus, preceded the development of a shared anlage of fields 18 and 19 linked with the parietooccipital sulcus. The functional differentiation of the primary visual field begins during the period of thalamic afferent ingrowth. This process coincides with the temporal smoothing of the calcarine sulcus, indicating a simultaneous progression of functional specialization and structural modifications. At the late fetal period, cortical plate of gyri and sulci banks showed higher NeuN-labeling than inside the sulcus in the same cytoarchitectonic field.
Collapse
Affiliation(s)
- Olga Godovalova
- Avsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Tsurupi Street, 3, Moscow, 117418, Russia.
| | - Alexandra Proshchina
- Avsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Tsurupi Street, 3, Moscow, 117418, Russia
| | - Anastasia Kharlamova
- Avsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Tsurupi Street, 3, Moscow, 117418, Russia
| | - Valeriy Barabanov
- Avsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Tsurupi Street, 3, Moscow, 117418, Russia
| | - Yuliya Krivova
- Avsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Tsurupi Street, 3, Moscow, 117418, Russia
| | - Olga Junemann
- The National Research Council Institute CNR-NANOTEC, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Marina Shahina
- Krasnopolsky Moscow Regional Research Institute of Obstetrics and Gynecology, Pokrovka Street, 22A, Moscow, 101000, Russia
| | - Sergey Saveliev
- Avsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", Tsurupi Street, 3, Moscow, 117418, Russia
| |
Collapse
|
2
|
Oh HN, Yoo D, Park S, Lee S, Kim WK. Assessment of poly(hexamethylenebicyanoguanide-hexamethylenediamine) hydrochloride-induced developmental neurotoxicity via oxidative stress mechanism: Integrative approaches with neuronal cells and zebrafish. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133146. [PMID: 38064952 DOI: 10.1016/j.jhazmat.2023.133146] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Abstract
Poly(hexamethylenebicyanoguanide-hexamethylenediamine) hydrochloride (PHMB) is a biocide with a broad spectrum of antibacterial activity. Its use as a disinfectant and preservative in consumer products results in human exposure to PHMB. Toxicity studies on PHMB mainly focus on systemic toxicity or skin irritation; however, its effects on developmental neurotoxicity (DNT) and the underlying mechanisms are poorly understood. In this study, the DNT effects of PHMB were evaluated using IMR-32 and SH-SY5Y cell lines and zebrafish. In both cell lines, PHMB concentrations ≥ 10 µM reduced neurite outgrowth, and cytotoxicity was observed at concentrations up to 40 µM. PHMB regulated expression of neurodevelopmental genes and induced reactive oxygen species (ROS) production and mitochondrial dysfunction. Treatment with N-acetylcysteine reversed the toxic effects of PHMB. Toxicity tests on zebrafish embryos showed that PHMB reduced viability and heart rate and caused irregular hatching. PHMB concentrations of 1-4 µM reduced the width of the brain and spinal cord of transgenic zebrafish and attenuated myelination processes. Furthermore, PHMB modulated expression of neurodevelopmental genes in zebrafish and induced ROS accumulation. These results suggested that PHMB exerted DNT effects in vitro and in vivo through a ROS-dependent mechanism, highlighting the risk of PHMB exposure.
Collapse
Affiliation(s)
- Ha-Na Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Donggon Yoo
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Seungmin Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Sangwoo Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Woo-Keun Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
3
|
Zhang C, Ye W, Zhao M, Long L, Xia D, Fan Z. KDM6B Negatively Regulates the Neurogenesis Potential of Apical Papilla Stem Cells via HES1. Int J Mol Sci 2023; 24:10608. [PMID: 37445785 PMCID: PMC10341966 DOI: 10.3390/ijms241310608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Stem cells from the apical papilla (SCAPs) are used to regulate the microenvironment of nerve defects. KDM6B, which functions as an H3K27me3 demethylase, is known to play a crucial role in neurogenesis. However, the mechanism by which KDM6B influences the neurogenesis potential of SCAPs remains unclear. We evaluated the expression of neural markers in SCAPs by using real-time RT-PCR and immunofluorescence staining. To assess the effectiveness of SCAP transplantation in the SCI model, we used the BBB scale to evaluate motor function. Additionally, toluidine blue staining and Immunofluorescence staining of NCAM, NEFM, β-III-tubulin, and Nestin were used to assess nerve tissue remodeling. Further analysis was conducted through Microarray analysis and ChIP assay to study the molecular mechanisms. Our results show that KDM6B inhibits the expression of NeuroD, TH, β-III tubulin, and Nestin. In vivo studies indicate that the SCAP-KDM6Bsh group is highly effective in restoring spinal cord structure and motor function in rats suffering from SCI. Our findings suggest that KDM6B directly binds to the HES1 promoter via regulating H3K27me3 and HES1 expression. In conclusion, our study can help understand the regulatory role of KDM6B in neurogenesis and provide more effective treatments for nerve injury.
Collapse
Affiliation(s)
- Chen Zhang
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China; (C.Z.); (W.Y.); (M.Z.); (L.L.)
- Department of Dental Emergency, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China;
| | - Weilong Ye
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China; (C.Z.); (W.Y.); (M.Z.); (L.L.)
| | - Mengyao Zhao
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China; (C.Z.); (W.Y.); (M.Z.); (L.L.)
| | - Lujue Long
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China; (C.Z.); (W.Y.); (M.Z.); (L.L.)
| | - Dengsheng Xia
- Department of Dental Emergency, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China;
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China; (C.Z.); (W.Y.); (M.Z.); (L.L.)
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing 100069, China
- Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing 100069, China
| |
Collapse
|
4
|
Coyoy-Salgado A, Orozco-Barrios C, Sánchez-Torres S, Olayo MG, Cruz GJ, Morales-Corona J, Olayo R, Diaz-Ruiz A, Ríos C, Alvarez-Mejia L, Mondragón-Lozano R, Morales-Guadarrama A, Alonso-García AL, Fabela-Sánchez O, Salgado-Ceballos H. Gene expression and locomotor recovery in adult rats with spinal cord injury and plasma-synthesized polypyrrole/iodine application combined with a mixed rehabilitation scheme. Front Neurol 2023; 14:1124245. [PMID: 37288064 PMCID: PMC10243140 DOI: 10.3389/fneur.2023.1124245] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/14/2023] [Indexed: 06/09/2023] Open
Abstract
Introduction Spinal cord injury (SCI) can cause paralysis, for which effective therapeutic strategies have not been developed yet. The only accepted strategy for patients is rehabilitation (RB), although this does not allow complete recovery of lost functions, which makes it necessary to combine it with strategies such as plasma-synthesized polypyrrole/iodine (PPy/I), a biopolymer with different physicochemical properties than PPy synthesized by conventional methods. After SCI in rats, PPy/I promotes functional recovery. Therefore, the purpose of this study was to increase the beneficial effects of both strategies and identify which genes activate PPy/I when applied alone or in combination with a mixed scheme of RB by swimming and enriched environment (SW/EE) in rats with SCI. Methods Microarray analysis was performed to identify mechanisms of action underlying the effects of PPy/I and PPy/I+SW/EE on motor function recovery as evaluated by the BBB scale. Results Results showed robust upregulation by PPy/I in genes related to the developmental process, biogenesis, synapse, and synaptic vesicle trafficking. In addition, PPy/I+SW/EE increased the expression of genes related to proliferation, biogenesis, cell development, morphogenesis, cell differentiation, neurogenesis, neuron development, and synapse formation processes. Immunofluorescence analysis showed the expression of β-III tubulin in all groups, a decreased expression of caspase-3 in the PPy/I group and GFAP in the PPy/I+SW/EE group (p < 0.05). Better preservation of nerve tissue was observed in PPy/I and PPy/SW/EE groups (p < 0.05). In the BBB scale, the control group scored 1.72 ± 0.41, animals with PPy/I treatment scored 4.23 ± 0.33, and those with PPy/I+SW/EE scored 9.13 ± 0.43 1 month after follow-up. Conclusion Thus, PPy/I+SW/EE could represent a therapeutic alternative for motor function recovery after SCI.
Collapse
Affiliation(s)
- Angélica Coyoy-Salgado
- Researchers for Mexico CONACyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
| | - Carlos Orozco-Barrios
- Researchers for Mexico CONACyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
| | - Stephanie Sánchez-Torres
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
- Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| | - María Guadalupe Olayo
- Instituto Nacional de Investigaciones Nucleares, Department of Physics, Axapusco, Mexico
| | - Guillermo Jesus Cruz
- Instituto Nacional de Investigaciones Nucleares, Department of Physics, Axapusco, Mexico
| | - Juan Morales-Corona
- Department of Physics, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Roberto Olayo
- Department of Physics, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | - Araceli Diaz-Ruiz
- Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez S.S.A., Department of Neurochemistry, Mexico City, Mexico
| | - Camilo Ríos
- Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez S.S.A., Department of Neurochemistry, Mexico City, Mexico
| | - Laura Alvarez-Mejia
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
- Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| | - Rodrigo Mondragón-Lozano
- Researchers for Mexico CONACyT-Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
| | - Axayacatl Morales-Guadarrama
- Electrical Engineering Department, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
- National Center for Research in Imaging and Medical Instrumentation, Universidad Autónoma Metropolitana Iztapalapa, Mexico City, Mexico
| | | | - Omar Fabela-Sánchez
- Researchers for Mexico CONACyT-Centro de Investigación en Química Aplicada, Department of Chemistry Macromolecules and Nanomaterials, Saltillo, Mexico
| | - Hermelinda Salgado-Ceballos
- Research Center of the Proyecto CAMINA A.C., Mexico City, Mexico
- Instituto Mexicano del Seguro Social, Medical Research Unit in Neurological Diseases, Specialty Hospital, National Medical Center Siglo XXI, Mexico City, Mexico
| |
Collapse
|
5
|
Yuan T, Tan M, Xu Y, Xiao Q, Wang H, Wu C, Li F, Peng L. All-in-one smart dressing for simultaneous angiogenesis and neural regeneration. J Nanobiotechnology 2023; 21:38. [PMID: 36737778 PMCID: PMC9896818 DOI: 10.1186/s12951-023-01787-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 01/15/2023] [Indexed: 02/05/2023] Open
Abstract
Wound repair, along with skin appendage regeneration, is challenged by insufficient angiogenesis and neural regeneration. Therefore, promoting both proangiogenic and neuro-regenerative therapeutic effects is essential for effective wound repair. However, most therapeutic systems apply these strategies separately or ineffectively. This study investigates the performance of an all-in-one smart dressing (ASD) that integrates angiogenic functional materials and multiple biological factors within a light crosslinked hydrogel, forming a multi-functional dressing capable of facilitating simultaneous micro-vascularization and neural regeneration. The ASD uses a zeolite-imidazolate framework 67 with anchored vanadium oxide (VO2@ZIF-67) that allows for the on-demand release of Co2+ with fluctuations in pH at the wound site to stimulate angiogenesis. It can simultaneously release CXCL12, ligustroflavone, and ginsenoside Rg1 in a sustained manner to enhance the recruitment of endogenous mesenchymal stem cells, inhibit senescence, and induce neural differentiation to achieve in situ nerve regeneration. The ASD can stimulate rapid angiogenesis and nerve regeneration within 17 days through multiple angiogenic and neuro-regenerative cues within one dressing. This study provides a proof-of-concept for integrating functional nanomaterials and multiple complementary drugs within a smart dressing for simultaneous angiogenesis and neural regeneration.
Collapse
Affiliation(s)
- Tiejun Yuan
- grid.13402.340000 0004 1759 700XCollege of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 People’s Republic of China
| | - Minhong Tan
- grid.13402.340000 0004 1759 700XCollege of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 People’s Republic of China ,grid.13402.340000 0004 1759 700XCollege of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou, 310027 People’s Republic of China
| | - Yang Xu
- grid.13402.340000 0004 1759 700XCollege of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 People’s Republic of China
| | - Qiyao Xiao
- grid.13402.340000 0004 1759 700XCollege of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 People’s Republic of China
| | - Hui Wang
- grid.13402.340000 0004 1759 700XCollege of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou, 310027 People’s Republic of China
| | - Chen Wu
- grid.13402.340000 0004 1759 700XCollege of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Zhejiang University, Hangzhou, 310027 People’s Republic of China
| | - Fulun Li
- grid.412540.60000 0001 2372 7462Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437 People’s Republic of China
| | - Lihua Peng
- grid.13402.340000 0004 1759 700XCollege of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 People’s Republic of China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, People’s Republic of China
| |
Collapse
|
6
|
Mouse Neural Stem Cell Differentiation and Human Adipose Mesenchymal Stem Cell Transdifferentiation Into Neuron- and Oligodendrocyte-like Cells With Myelination Potential. Stem Cell Rev Rep 2021; 18:732-751. [PMID: 34780018 DOI: 10.1007/s12015-021-10218-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 01/09/2023]
Abstract
Stem cell therapy is an interesting approach for neural repair, once it can improve and increase processes, like angiogenesis, neurogenesis, and synaptic plasticity. In this regard, adult neural stem cells (NSC) are studied for their mechanisms of proliferation, differentiation and functionality in neural repair. Here, we describe novel neural differentiation methods. NSC from adult mouse brains and human adipose-derived stem cells (hADSC) were isolated and characterized regarding their neural differentiation potential based on neural marker expression profiles. For both cell types, their capabilities of differentiating into neuron-, astrocyte- and oligodendrocytes-like cells (NLC, ALC and OLC, respectively) were analyzed. Our methodologies were capable of producing NLC, ALC and OLC from adult murine and human transdifferentiated NSC. NSC showed augmented gene expression of NES, TUJ1, GFAP and PDGFRA/Cnp. Following differentiation induction into NLC, OLC or ALC, specific neural phenotypes were obtained expressing MAP2, GalC/O4 or GFAP with compatible morphologies, respectively. Accordingly, immunostaining for nestin+ in NSC, GFAP+ in astrocytes and GalC/O4+ in oligodendrocytes was detected. Co-cultured NLC and OLC showed excitability in 81.3% of cells and 23.5% of neuron/oligodendrocyte marker expression overlap indicating occurrence of in vitro myelination. We show here that hADSC can be transdifferentiated into NSC and distinct neural phenotypes with the occurrence of neuron myelination in vitro, providing novel strategies for CNS regeneration therapy. Superior Part: Schematic organization of obtaining and generating hNSC from hADSC and differentiation processes and phenotypic expression of neuron, astrocyte and oligodendrocyte markers (MAP2, GFAP and O4, respectively) and stem cell marker (NES) of differentiating hNSC 14 days after induction. The nuclear staining in blue corresponds to DAPI. bar = 100 μm. Inferior part: Neural phenotype fates in diverse differentiation media. NES: nestin; GFAP: Glial fibrillary acidic protein. MAP2: Microtubule-associated protein 2. TUJ1: β-III tubulin. PDGFRA: PDGF receptor alpha. Two-way ANOVA with Bonferroni post-test with n = 3. * p < 0.05 and ** p < 0.01: (NSCiM1 NSC induction medium 1) vs differentiation media.
Collapse
|
7
|
Lomboni DJ, Steeves A, Schock S, Bonetti L, De Nardo L, Variola F. Compounded topographical and physicochemical cueing by micro-engineered chitosan substrates on rat dorsal root ganglion neurons and human mesenchymal stem cells. SOFT MATTER 2021; 17:5284-5302. [PMID: 34075927 DOI: 10.1039/d0sm02170a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Given the intertwined physicochemical effects exerted in vivo by both natural and synthetic (e.g., biomaterial) interfaces on adhering cells, the evaluation of structure-function relationships governing cellular response to micro-engineered surfaces for applications in neuronal tissue engineering requires the use of in vitro testing platforms which consist of a clinically translatable material with tunable physiochemical properties. In this work, we micro-engineered chitosan substrates with arrays of parallel channels with variable width (20 and 60 μm). A citric acid (CA)-based crosslinking approach was used to provide an additional level of synergistic cueing on adhering cells by regulating the chitosan substrate's stiffness. Morphological and physicochemical characterization was conducted to unveil the structure-function relationships which govern the activity of rat dorsal root ganglion neurons (DRGs) and human mesenchymal stem cells (hMSCs), ultimately singling out the key role of microtopography, roughness and substrate's stiffness. While substrate's stiffness predominantly affected hMSC spreading, the modulation of the channels' design affected the neuronal architecture's complexity and guided the morphological transition of hMSCs. Finally, the combined analysis of tubulin expression and cell morphology allowed us to cast new light on the predominant role of the microtopography over substrate's stiffness in the process of hMSCs neurogenic differentiation.
Collapse
Affiliation(s)
- David J Lomboni
- Department of Mechanical Engineering, University of Ottawa, K1N 6N5 Canada. and Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - Alexander Steeves
- Department of Mechanical Engineering, University of Ottawa, K1N 6N5 Canada. and Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - Sarah Schock
- Department of Cellular and Molecular Medicine, University of Ottawa, Canada and The Children's Hospital of Eastern Ontario (CHEO) Research Institute, Canada
| | - Lorenzo Bonetti
- Department of Chemistry, Materials and Chemical Engineering, "G. Natta", Politecnico di Milano, Italy
| | - Luigi De Nardo
- Department of Chemistry, Materials and Chemical Engineering, "G. Natta", Politecnico di Milano, Italy
| | - Fabio Variola
- Department of Mechanical Engineering, University of Ottawa, K1N 6N5 Canada. and Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada and Department of Cellular and Molecular Medicine, University of Ottawa, Canada and The Children's Hospital of Eastern Ontario (CHEO) Research Institute, Canada
| |
Collapse
|
8
|
Trombetta-Lima M, Assis-Ribas T, Cintra RC, Campeiro JD, Guerreiro JR, Winnischofer SMB, Nascimento ICC, Ulrich H, Hayashi MAF, Sogayar MC. Impact of Reck expression and promoter activity in neuronal in vitro differentiation. Mol Biol Rep 2021; 48:1985-1994. [PMID: 33619662 DOI: 10.1007/s11033-021-06175-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 01/20/2021] [Indexed: 02/07/2023]
Abstract
Reck (REversion-inducing Cysteine-rich protein with Kazal motifs) tumor suppressor gene encodes a multifunctional glycoprotein which inhibits the activity of several matrix metalloproteinases (MMPs), and has the ability to modulate the Notch and canonical Wnt pathways. Reck-deficient neuro-progenitor cells undergo precocious differentiation; however, modulation of Reck expression during progression of the neuronal differentiation process is yet to be characterized. In the present study, we demonstrate that Reck expression levels are increased during in vitro neuronal differentiation of PC12 pheochromocytoma cells and P19 murine teratocarcinoma cells and characterize mouse Reck promoter activity during this process. Increased Reck promoter activity was found upon induction of differentiation in PC12 cells, in accordance with its increased mRNA expression levels in mouse in vitro models. Interestingly, Reck overexpression, prior to the beginning of the differentiation protocol, led to diminished efficiency of the neuronal differentiation process. Taken together, our findings suggest that increased Reck expression at early stages of differentiation diminishes the number of neuron-like cells, which are positive for the beta-3 tubulin marker. Our data highlight the importance of Reck expression evaluation to optimize in vitro neuronal differentiation protocols.
Collapse
Affiliation(s)
- Marina Trombetta-Lima
- Núcleo de Terapia Celular e Molecular (NUCEL), Faculdade de Medicina, Universidade de São Paulo (USP), Rua Pangaré, 100 (Cidade Universitária), São Paulo, SP, 05360-130, Brazil
| | - Thais Assis-Ribas
- Núcleo de Terapia Celular e Molecular (NUCEL), Faculdade de Medicina, Universidade de São Paulo (USP), Rua Pangaré, 100 (Cidade Universitária), São Paulo, SP, 05360-130, Brazil
| | - Ricardo C Cintra
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Joana D Campeiro
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Rua 3 de Maio 100, Ed INFAR, 3º andar, São Paulo, SP, 04044-020, Brazil
| | - Juliano R Guerreiro
- Faculdade de Farmácia, Universidade Paulista (UNIP), São Paulo, SP, 05347-020, Brazil
| | - Sheila M B Winnischofer
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná (UFPR), Curitiba, PR, 81531-990, Brazil
- Departamento de Biologia Celular, Universidade Federal do Paraná (UFPR), Curitiba, PR, 81531-990, Brazil
| | - Isis C C Nascimento
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, 05508-000, Brazil
| | - Mirian A F Hayashi
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), Rua 3 de Maio 100, Ed INFAR, 3º andar, São Paulo, SP, 04044-020, Brazil.
| | - Mari C Sogayar
- Núcleo de Terapia Celular e Molecular (NUCEL), Faculdade de Medicina, Universidade de São Paulo (USP), Rua Pangaré, 100 (Cidade Universitária), São Paulo, SP, 05360-130, Brazil.
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo (USP), São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
9
|
Alexandre J, Malheiro R, Dias da Silva D, Carmo H, Carvalho F, Silva JP. The Synthetic Cannabinoids THJ-2201 and 5F-PB22 Enhance In Vitro CB 1 Receptor-Mediated Neuronal Differentiation at Biologically Relevant Concentrations. Int J Mol Sci 2020; 21:6277. [PMID: 32872617 PMCID: PMC7503567 DOI: 10.3390/ijms21176277] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/25/2020] [Accepted: 08/28/2020] [Indexed: 01/07/2023] Open
Abstract
Recreational use of synthetic cannabinoids (SCs) before and during pregnancy poses a major public health risk, due to the potential onset of neurodevelopmental disorders in the offspring. Herein, we report the assessment of the neurotoxic potential of two commonly abused SCs, THJ-2201 and 5F-PB22, particularly focusing on how they affect neuronal differentiation in vitro. Differentiation ratios, total neurite length, and neuronal marker expression were assessed in NG108-15 neuroblastoma x glioma cells exposed to the SCs at non-toxic, biologically relevant concentrations (≤1 μM), either in acute or repeated exposure settings. Both SCs enhanced differentiation ratios and total neurite length of NG108-15 cells near two-fold compared to vehicle-treated cells, in a CB1R activation-dependent way, as the CB1R blockade with a specific antagonist (SR141718) abrogated SC-induced effects. Interestingly, repeated 5F-PB22 exposure was required to reach effects similar to a single THJ-2201 dose. Cell viability and proliferation, mitochondrial membrane potential, and intracellular ATP levels were also determined. The tested SCs increased mitochondrial tetramethyl rhodamine ethyl ester (TMRE) accumulation after 24 h at biologically relevant concentrations but did not affect any of the other toxicological parameters. Overall, we report firsthand the CB1R-mediated enhancement of neurodifferentiation by 5F-PB22 and THJ-2201 at biologically relevant concentrations.
Collapse
Affiliation(s)
| | | | | | | | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.A.); (R.M.); (D.D.d.S.); (H.C.)
| | - João Pedro Silva
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (J.A.); (R.M.); (D.D.d.S.); (H.C.)
| |
Collapse
|
10
|
Murphy AR, Haynes JM, Laslett AL, Cameron NR, O'Brien CM. Three-dimensional differentiation of human pluripotent stem cell-derived neural precursor cells using tailored porous polymer scaffolds. Acta Biomater 2020; 101:102-116. [PMID: 31610339 DOI: 10.1016/j.actbio.2019.10.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/30/2019] [Accepted: 10/07/2019] [Indexed: 02/07/2023]
Abstract
This study investigates the utility of a tailored poly(ethylene glycol) diacrylate-crosslinked porous polymeric tissue engineering scaffold, with mechanical properties specifically optimised to be comparable to that of mammalian brain tissue for 3D human neural cell culture. Results obtained here demonstrate the attachment, proliferation and terminal differentiation of both human induced pluripotent stem cell- and embryonic stem cell-derived neural precursor cells (hPSC-NPCs) throughout the interconnected porous network within laminin-coated scaffolds. Phenotypic data and functional analyses are presented demonstrating that this material supports terminal in vitro neural differentiation of hPSC-NPCs to a mixed population of viable neuronal and glial cells for periods of up to 49 days. This is evidenced by the upregulation of TUBB3, MAP2, SYP and GFAP gene expression, as well as the presence of the proteins βIII-TUBULIN, NEUN, MAP2 and GFAP. Functional maturity of neural cells following 49 days 3D differentiation culture was tested via measurement of intracellular calcium. These analyses revealed spontaneously active, synchronous and rhythmic calcium flux, as well as response to the neurotransmitter glutamate. This tailored construct has potential application as an improved in vitro human neurogenesis model with utility in platform drug discovery programs. STATEMENT OF SIGNIFICANCE: The interconnected porosity of polyHIPE scaffolds exhibits the ability to support three-dimensional neural cell network formation due to limited resistance to cellular migration and re-organisation. The previously developed scaffold material displays mechanical properties similar to that of the mammalian brain. This research also employs the utility of pluripotent stem cell-derived neural cells which are of greater clinical relevance than primary neural cell lines. This scaffold material has future potential in better mimicking three-dimensional neural networks found in the human brain and may result in improved in vitro models for disease modelling and drug screening applications.
Collapse
|
11
|
Li Y, Mao X, Zhou X, Su Y, Zhou X, Shi K, Zhao S. An optimized method for neuronal differentiation of embryonic stem cells in vitro. J Neurosci Methods 2019; 330:108486. [PMID: 31706928 DOI: 10.1016/j.jneumeth.2019.108486] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 10/11/2019] [Accepted: 10/29/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Neural differentiation from embryonic stem cells (ESCs) is an excellent model for elucidating the key mechanisms involved in neurogenesis, and also provides an unlimited source of progenitors for cell-based nerve regeneration. However, the existing protocols such as small molecule substances, 3D matrix, co-culture technique and transgenic method, are complicated and difficult to operate, thus are limited by laboratory conditions. Looking for an easy-to-operate protocol with easily gained material and high induction efficiency has always been a hot issue in neuroscience research. NEW METHODS This paper established an optimized method for embryonic neurogenesis using a strategy of "combinatorial screening". In our study, the whole process of embryonic neurogenesis was divided into two phases, and the differentiation efficiency of seven experimental protocols in phase I and three protocols in phase II were systematically evaluated in A2lox and 129 ESCs. RESULTS In phase I differentiation, "2-day embryoid bodies formation + 6-day retinoic acid induction" (Phase I-protocol 3) could effectively induce the differentiation of ESCs into neural precursor cells (NPCs). Furthermore, in phase II, N2B27 medium II (Phase II-protocol 3) could better support the subsequent differentiation from NPCs into neurons. COMPARISON WITH EXISTING METHOD(S) Such a combinational method (phase I-protocol 3 and phase II-protocol 3) can realize embryonic neurogenesis with high efficiency, easy implementation and low-cost, and is suitable for promotion in most laboratories. CONCLUSIONS Through "combinatorial screening" strategy, we established an optimized method for embryonic neurogenesis in vitro, which is expected to be a powerful tool for neuroscience research.
Collapse
Affiliation(s)
- Yuan Li
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, PR China
| | - Xiang Mao
- Wuhan Centres for Disease Prevention and Control, 24# Jianghan N. Road, Wuhan, Hubei, 430015, PR China
| | - Xianyi Zhou
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, PR China
| | - Yuting Su
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, PR China
| | - Xiangyu Zhou
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, PR China
| | - Kaituo Shi
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, PR China
| | - Shasha Zhao
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430065, PR China.
| |
Collapse
|
12
|
Patel M, Lee HJ, Son S, Kim H, Kim J, Jeong B. Iron Ion-Releasing Polypeptide Thermogel for Neuronal Differentiation of Mesenchymal Stem Cells. Biomacromolecules 2019; 21:143-151. [DOI: 10.1021/acs.biomac.9b01096] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| | - Hyun Jung Lee
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| | - Seungyi Son
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| | - Heeju Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| | - Jinheung Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Korea
| |
Collapse
|
13
|
Michel N, Majumdar UB, Lannigan J, McConnell MJ. Imaging Flow Cytometry Quantifies Neural Genome Dynamics. Cytometry A 2019; 95:825-835. [PMID: 31063256 PMCID: PMC7851630 DOI: 10.1002/cyto.a.23783] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/15/2019] [Indexed: 12/18/2022]
Abstract
Somatic mosaicism is a common consequence of normal development. DNA repair is simply not perfect, and each cell's genome incurs continuous DNA damage as a consequence of transcription, replication, and other cell biological stressors. Brain somatic mosaicism is particularly noteworthy because the vast majority of an individual's neurons are with that individual for life and neural circuits give rise directly to behavioral phenotypes. Brain somatic mosaicism, now revealed and tractable due to advances in single cell 'omic approaches, has emerged as an intriguing and unexplored aspect of neuronal diversity. Furthermore, the study of DNA damage during early neurodevelopment, when the rate of mutagenesis is high, is the perfect starting point to understand the origins of brain mosaicism. Flow cytometry is a highly efficient technique to study cell cycle and intracellular proteins of interest, particularly those related to DNA damage, but it lacks the high resolution of microscopy to examine the localization of these proteins. In this study, we outline a novel single-cell approach to quantify DNA double-strand break (DNA DSB) dynamics during early human neurodevelopment by applying imaging flow cytometry (IFC) to human-induced pluripotent stem cell-derived neural progenitor cells (NPCs) undergoing neurogenesis. We establish an increase of DNA DSBs by quantifying γH2AX foci in mildly stressed NPCs using various single-cell approaches in addition to IFC including fluorescent microscopy, conventional flow cytometry, and measuring DNA DSBs with the comet assay. We demonstrate the dose-dependent sensitive detection of γH2AX foci through IFC and reveal the dynamics of DNA DSBs in proliferating and differentiating neural cells in early neurogenesis. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Nadine Michel
- Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, Neuroscience Graduate Program, Charlottesville, Virginia 22908
| | - Usnish B. Majumdar
- Department of Health System Design and Global Health, Icahn School of Medicine, 1 Gustave L. Levy Pl, New York, NY 10029
| | - Joanne Lannigan
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Flow Cytometry Core Facility, 1340 Jefferson Park Ave., Pinn Hall, Room 2011, Charlottesville, Virginia 22908-0734
| | - Michael J. McConnell
- Department of Biochemistry & Molecular Genetics, University of Virginia School of Medicine, Neuroscience Graduate Program, Charlottesville, Virginia 22908
- Department of Neuroscience, University of Virginia School of Medicine, Neuroscience Graduate Program, Charlottesville, Virginia 22908
| |
Collapse
|
14
|
Rahmani A, Nadri S, Kazemi HS, Mortazavi Y, Sojoodi M. Conductive electrospun scaffolds with electrical stimulation for neural differentiation of conjunctiva mesenchymal stem cells. Artif Organs 2019; 43:780-790. [PMID: 30674064 DOI: 10.1111/aor.13425] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/13/2019] [Accepted: 01/17/2019] [Indexed: 12/23/2022]
Abstract
An electrical stimulus is a new approach to neural differentiation of stem cells. In this work, the neural differentiation of conjunctiva mesenchymal stem cells (CJMSCs) on a new 3D conductive fibrous scaffold of silk fibroin (SF) and reduced graphene oxide (rGo) were examined. rGo (3.5% w/w) was dispersed in SF-acid formic solution (10% w/v) and conductive nanofibrous scaffold was fabricated using the electrospinning method. SEM and TEM microscopies were used for fibrous scaffold characterization. CJMSCs were cultured on the scaffold and 2 electrical impulse models (Current 1:115 V/m, 100-Hz frequency and current 2:115 v/m voltages, 0.1-Hz frequency) were applied for 7 days. Also, the effect of the fibrous scaffold and electrical impulses on cell viability and neural gene expression were examined using MTT assay and qPCR analysis. Fibrous scaffold with the 220 ± 20 nm diameter and good dispersion of graphene nanosheets at the surface of nanofibers were fabricated. The MTT result showed the viability of cells on the scaffold, with current 2 lower than current 1. qPCR analysis confirmed that the expression of β-tubulin (2.4-fold P ≤ 0.026), MAP-2 (1.48-fold; P ≤ 0.03), and nestin (1.5-fold; P ≤ 0.03) genes were higher in CJMSCs on conductive scaffold with 100-Hz frequency compared to 0.1-Hz frequency. Collectively, we proposed that SF-rGo fibrous scaffolds, as a new conductive fibrous scaffold with electrical stimulation are good strategies for neural differentiation of stem cells and the type of electrical pulses has an influence on neural differentiation and proliferation of CJMSCs.
Collapse
Affiliation(s)
- Ali Rahmani
- Department of Medical Nanotechnology, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Samad Nadri
- Department of Medical Nanotechnology, Zanjan University of Medical Sciences, Zanjan, Iran.,Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Habib Sayed Kazemi
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Yousef Mortazavi
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Medical Biotechnology, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahdi Sojoodi
- Department of Electrical and Computer Engineering, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
15
|
Hidalgo San Jose L, Stephens P, Song B, Barrow D. Microfluidic Encapsulation Supports Stem Cell Viability, Proliferation, and Neuronal Differentiation. Tissue Eng Part C Methods 2018; 24:158-170. [PMID: 29258387 PMCID: PMC5865257 DOI: 10.1089/ten.tec.2017.0368] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Stem cell encapsulation technology demonstrates much promise for the replacement of damaged tissue in several diseases, including spinal cord injury (SCI). The use of biocompatible microcapsules permits the control of stem cell fate in situ to facilitate the replacement of damaged/lost tissue. In this work, a novel customized microfluidic device was developed for the reproducible encapsulation of neural stem cells (NSCs) and dental pulp stem cells (DPSCs) within monodisperse, alginate-collagen microcapsules. Both cell types survived within the microcapsules for up to 21 days in culture. Stem cells demonstrated retention of their multipotency and neuronal differentiation properties upon selective release from the microcapsules, as demonstrated by high proliferation rates and the production of stem cell and neuronal lineage markers. When cell-laden microcapsules were transplanted into an organotypic SCI model, the microcapsules effectively retained the transplanted stem cells at the site of implantation. Implanted cells survived over a 10 day period in culture after transplantation and demonstrated commitment to a neural lineage. Our device provides a quick, effective, and aseptic method for the encapsulation of two different stem cell types (DPSCs and NSCs) within alginate-collagen microcapsules. Since stem cells were able to retain their viability and neural differentiation capacity within such microcapsules, this method provides a useful technique to study stem cell behavior within three-dimensional environments.
Collapse
Affiliation(s)
- Lorena Hidalgo San Jose
- 1 Biomedical Engineering Research Group, Cardiff School of Engineering, Cardiff University , Cardiff, United Kingdom .,2 Wound Biology Group, School of Dentistry, Cardiff Institute of Tissue Engineering and Repair, Cardiff University , Cardiff, United Kingdom
| | - Phil Stephens
- 2 Wound Biology Group, School of Dentistry, Cardiff Institute of Tissue Engineering and Repair, Cardiff University , Cardiff, United Kingdom
| | - Bing Song
- 2 Wound Biology Group, School of Dentistry, Cardiff Institute of Tissue Engineering and Repair, Cardiff University , Cardiff, United Kingdom
| | - David Barrow
- 1 Biomedical Engineering Research Group, Cardiff School of Engineering, Cardiff University , Cardiff, United Kingdom
| |
Collapse
|
16
|
Niu H, Gou R, Xu Q, Duan D. Recombinant insulin-like growth factor binding protein-4 inhibits proliferation and promotes differentiation of neural progenitor cells. Neurosci Lett 2017; 642:71-76. [DOI: 10.1016/j.neulet.2017.01.066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/16/2017] [Accepted: 01/28/2017] [Indexed: 10/20/2022]
|
17
|
Kim YY, Roubal I, Lee YS, Kim JS, Hoang M, Mathiyakom N, Kim Y. Alcohol-Induced Molecular Dysregulation in Human Embryonic Stem Cell-Derived Neural Precursor Cells. PLoS One 2016; 11:e0163812. [PMID: 27682028 PMCID: PMC5040434 DOI: 10.1371/journal.pone.0163812] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/14/2016] [Indexed: 01/08/2023] Open
Abstract
Adverse effect of alcohol on neural function has been well documented. Especially, the teratogenic effect of alcohol on neurodevelopment during embryogenesis has been demonstrated in various models, which could be a pathologic basis for fetal alcohol spectrum disorders (FASDs). While the developmental defects from alcohol abuse during gestation have been described, the specific mechanisms by which alcohol mediates these injuries have yet to be determined. Recent studies have shown that alcohol has significant effect on molecular and cellular regulatory mechanisms in embryonic stem cell (ESC) differentiation including genes involved in neural development. To test our hypothesis that alcohol induces molecular alterations during neural differentiation we have derived neural precursor cells from pluripotent human ESCs in the presence or absence of ethanol treatment. Genome-wide transcriptomic profiling identified molecular alterations induced by ethanol exposure during neural differentiation of hESCs into neural rosettes and neural precursor cell populations. The Database for Annotation, Visualization and Integrated Discovery (DAVID) functional analysis on significantly altered genes showed potential ethanol’s effect on JAK-STAT signaling pathway, neuroactive ligand-receptor interaction, Toll-like receptor (TLR) signaling pathway, cytokine-cytokine receptor interaction and regulation of autophagy. We have further quantitatively verified ethanol-induced alterations of selected candidate genes. Among verified genes we further examined the expression of P2RX3, which is associated with nociception, a peripheral pain response. We found ethanol significantly reduced the level of P2RX3 in undifferentiated hESCs, but induced the level of P2RX3 mRNA and protein in hESC-derived NPCs. Our result suggests ethanol-induced dysregulation of P2RX3 along with alterations in molecules involved in neural activity such as neuroactive ligand-receptor interaction may be a molecular event associated with alcohol-related peripheral neuropathy of an enhanced nociceptive response.
Collapse
Affiliation(s)
- Yi Young Kim
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Ivan Roubal
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Youn Soo Lee
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Jin Seok Kim
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Michael Hoang
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Nathan Mathiyakom
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
| | - Yong Kim
- Laboratory of Stem Cell & Cancer Epigenetic Research, School of Dentistry, University of California Los Angeles, 10833 Le Conte Avenue, 73–041 CHS, Los Angeles, CA, 90095, United States of America
- Center for Oral and Head/Neck Oncology Research Center, Division of Oral Biology & Medicine, UCLA School of Dentistry, 10833 Le Conte Avenue, 73–022 CHS, Los Angeles, CA, 90095, United States of America
- UCLA’s Jonsson Comprehensive Cancer Center, 8–684 Factor Building, Box 951781, Los Angeles, CA, 90095, United States of America
- UCLA Broad Stem Cell Research Center, Box 957357, Los Angeles, CA, 90095, United States of America
- * E-mail:
| |
Collapse
|
18
|
Morales-Hernández A, González-Rico FJ, Román AC, Rico-Leo E, Alvarez-Barrientos A, Sánchez L, Macia Á, Heras SR, García-Pérez JL, Merino JM, Fernández-Salguero PM. Alu retrotransposons promote differentiation of human carcinoma cells through the aryl hydrocarbon receptor. Nucleic Acids Res 2016; 44:4665-83. [PMID: 26883630 PMCID: PMC4889919 DOI: 10.1093/nar/gkw095] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/09/2016] [Indexed: 12/18/2022] Open
Abstract
Cell differentiation is a central process in development and in cancer growth and dissemination. OCT4 (POU5F1) and NANOG are essential for cell stemness and pluripotency; yet, the mechanisms that regulate their expression remain largely unknown. Repetitive elements account for almost half of the Human Genome; still, their role in gene regulation is poorly understood. Here, we show that the dioxin receptor (AHR) leads to differentiation of human carcinoma cells through the transcriptional upregulation of Alu retrotransposons, whose RNA transcripts can repress pluripotency genes. Despite the genome-wide presence of Alu elements, we provide evidences that those located at the NANOG and OCT4 promoters bind AHR, are transcribed by RNA polymerase-III and repress NANOG and OCT4 in differentiated cells. OCT4 and NANOG repression likely involves processing of Alu-derived transcripts through the miRNA machinery involving the Microprocessor and RISC. Consistently, stable AHR knockdown led to basal undifferentiation, impaired Alus transcription and blockade of OCT4 and NANOG repression. We suggest that transcripts produced from AHR-regulated Alu retrotransposons may control the expression of stemness genes OCT4 and NANOG during differentiation of carcinoma cells. The control of discrete Alu elements by specific transcription factors may have a dynamic role in genome regulation under physiological and diseased conditions.
Collapse
Affiliation(s)
- Antonio Morales-Hernández
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Avenida de Elvas s/n, 06071-Badajoz, Spain
| | - Francisco J González-Rico
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Avenida de Elvas s/n, 06071-Badajoz, Spain
| | - Angel C Román
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avenida Doctor Arce 37, 28002-Madrid, Spain
| | - Eva Rico-Leo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Avenida de Elvas s/n, 06071-Badajoz, Spain
| | - Alberto Alvarez-Barrientos
- Servicio de Técnicas Aplicadas a las Biociencias, Universidad de Extremadura, Avenida de Elvas s/n 06071-Badajoz, Spain
| | - Laura Sánchez
- GENYO. Centro de Genómica e Investigación Oncológica: Pfizer/Universidad de Granada/Junta de Andalucía, Avda. de la Ilustración 114, PTS Granada, 18016-Granada, Spain
| | - Ángela Macia
- GENYO. Centro de Genómica e Investigación Oncológica: Pfizer/Universidad de Granada/Junta de Andalucía, Avda. de la Ilustración 114, PTS Granada, 18016-Granada, Spain
| | - Sara R Heras
- GENYO. Centro de Genómica e Investigación Oncológica: Pfizer/Universidad de Granada/Junta de Andalucía, Avda. de la Ilustración 114, PTS Granada, 18016-Granada, Spain
| | - José L García-Pérez
- GENYO. Centro de Genómica e Investigación Oncológica: Pfizer/Universidad de Granada/Junta de Andalucía, Avda. de la Ilustración 114, PTS Granada, 18016-Granada, Spain
| | - Jaime M Merino
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Avenida de Elvas s/n, 06071-Badajoz, Spain
| | - Pedro M Fernández-Salguero
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, Avenida de Elvas s/n, 06071-Badajoz, Spain
| |
Collapse
|
19
|
Schulpen SHW, de Jong E, de la Fonteyne LJJ, de Klerk A, Piersma AH. Distinct gene expression responses of two anticonvulsant drugs in a novel human embryonic stem cell based neural differentiation assay protocol. Toxicol In Vitro 2014; 29:449-57. [PMID: 25524013 DOI: 10.1016/j.tiv.2014.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/14/2014] [Accepted: 12/03/2014] [Indexed: 12/23/2022]
Abstract
Hazard assessment of chemicals and pharmaceuticals is increasingly gaining from knowledge about molecular mechanisms of toxic action acquired in dedicated in vitro assays. We have developed an efficient human embryonic stem cell neural differentiation test (hESTn) that allows the study of the molecular interaction of compounds with the neural differentiation process. Within the 11-day differentiation protocol of the assay, embryonic stem cells lost their pluripotency, evidenced by the reduced expression of stem cell markers Pou5F1 and Nanog. Moreover, stem cells differentiated into neural cells, with morphologically visible neural structures together with increased expression of neural differentiation-related genes such as βIII-tubulin, Map2, Neurogin1, Mapt and Reelin. Valproic acid (VPA) and carbamazepine (CBZ) exposure during hESTn differentiation led to concentration-dependent reduced expression of βIII-tubulin, Neurogin1 and Reelin. In parallel VPA caused an increased gene expression of Map2 and Mapt which is possibly related to the neural protective effect of VPA. These findings illustrate the added value of gene expression analysis for detecting compound specific effects in hESTn. Our findings were in line with and could explain effects observed in animal studies. This study demonstrates the potential of this assay protocol for mechanistic analysis of specific compound-induced inhibition of human neural cell differentiation.
Collapse
Affiliation(s)
- Sjors H W Schulpen
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands; Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands.
| | - Esther de Jong
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands; Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands
| | - Liset J J de la Fonteyne
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands
| | - Arja de Klerk
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands
| | - Aldert H Piersma
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands; Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584 CM Utrecht, The Netherlands
| |
Collapse
|
20
|
Liu C, Sun Y, Arnold J, Lu B, Guo S. Synergistic contribution of SMAD signaling blockade and high localized cell density in the differentiation of neuroectoderm from H9 cells. Biochem Biophys Res Commun 2014; 452:895-900. [PMID: 25218470 PMCID: PMC4193974 DOI: 10.1016/j.bbrc.2014.08.137] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 08/25/2014] [Indexed: 02/08/2023]
Abstract
Directed neural differentiation of human embryonic stem cells (ESCs) enables researchers to generate diverse neuronal populations for human neural development study and cell replacement therapy. To realize this potential, it is critical to precisely understand the role of various endogenous and exogenous factors involved in neural differentiation. Cell density, one of the endogenous factors, is involved in the differentiation of human ESCs. Seeding cell density can result in variable terminal cell densities or localized cell densities (LCDs), giving rise to various outcomes of differentiation. Thus, understanding how LCD determines the differentiation potential of human ESCs is important. The aim of this study is to highlight the role of LCD in the differentiation of H9 human ESCs into neuroectoderm (NE), the primordium of the nervous system. We found the initially seeded cells form derived cells with variable LCDs and subsequently affect the NE differentiation. Using a newly established method for the quantitative examination of LCD, we demonstrated that in the presence of induction medium supplemented with or without SMAD signaling blockers, high LCD promotes the differentiation of NE. Moreover, SMAD signaling blockade promotes the differentiation of NE but not non-NE germ layers, which is dependent on high LCDs. Taken together, this study highlights the need to develop innovative strategies or techniques based on LCDs for generating neural progenies from human ESCs.
Collapse
Affiliation(s)
- Chao Liu
- Department of Histology and Embryology, Institute of Stem Cell and Tissue Engineering, Anhui Medical University, Hefei, Anhui 230032, China; Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, CA 94143, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Yaping Sun
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, CA 94143, USA
| | - Joshua Arnold
- Stem Cell Core, Gladstone Institute of Cardiovascular Disease, University of California San Francisco, San Francisco, CA 94158, USA
| | - Bingwei Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Su Guo
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, CA 94143, USA.
| |
Collapse
|