1
|
Di Lisa D, Andolfi A, Masi G, Uras G, Ferrari PF, Martinoia S, Pastorino L. Impact of perfusion on neuronal development in human derived neuronal networks. APL Bioeng 2024; 8:046102. [PMID: 39364213 PMCID: PMC11446581 DOI: 10.1063/5.0221911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
Advanced in vitro models of the brain have evolved in recent years from traditional two-dimensional (2D) ones, based on rodent derived cells, to three-dimensional (3D) ones, based on human neurons derived from induced pluripotent stem cells. To address the dynamic changes of the tissue microenvironment, bioreactors are used to control the in vitro microenvironment for viability, repeatability, and standardization. However, in neuronal tissue engineering, bioreactors have primarily been used for cell expansion purposes, while microfluidic systems have mainly been employed for culturing organoids. In this study, we explored the use of a commercial perfusion bioreactor to control the culture microenvironment of neuronal cells in both 2D and 3D cultures. Namely, neurons differentiated from human induced pluripotent stem cells (iNeurons) were cultured in 2D under different constant flow rates for 72 h. The impact of different flow rates on early-stage neuronal development and synaptogenesis was assessed by morphometric characterization and synaptic analysis. Based on these results, two involving variable flow rates were developed and applied again in 2D culture. The most effective protocol, in terms of positive impact on neuronal development, was then used for a preliminary study on the application of dynamic culturing conditions to neuronal cells in 3D. To this purpose, both iNeurons, co-cultured with astrocytes, and the human neuroblastoma cells SH-SY5Y were embedded into a hydrogel and maintained under perfusion for up to 28 days. A qualitative evaluation by immunocytochemistry and confocal microscopy was carried out to assess cell morphology and the formation of a 3D neuronal network.
Collapse
Affiliation(s)
| | - Andrea Andolfi
- DIBRIS, Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, Via Opera Pia 13, 16145 Genoa, Italy
| | - Giacomo Masi
- DIBRIS, Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, Via Opera Pia 13, 16145 Genoa, Italy
| | | | | | | | | |
Collapse
|
2
|
Pampanella L, Petrocelli G, Abruzzo PM, Zucchini C, Canaider S, Ventura C, Facchin F. Cytochalasins as Modulators of Stem Cell Differentiation. Cells 2024; 13:400. [PMID: 38474364 DOI: 10.3390/cells13050400] [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: 01/22/2024] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Regenerative medicine aims to identify new research strategies for the repair and restoration of tissues damaged by pathological or accidental events. Mesenchymal stem cells (MSCs) play a key role in regenerative medicine approaches due to their specific properties, such as the high rate of proliferation, the ability to differentiate into several cell lineages, the immunomodulatory potential, and their easy isolation with minimal ethical issues. One of the main goals of regenerative medicine is to modulate, both in vitro and in vivo, the differentiation potential of MSCs to improve their use in the repair of damaged tissues. Over the years, much evidence has been collected about the ability of cytochalasins, a large family of 60 metabolites isolated mainly from fungi, to modulate multiple properties of stem cells (SCs), such as proliferation, migration, and differentiation, by altering the organization of the cyto- and the nucleo-skeleton. In this review, we discussed the ability of two different cytochalasins, cytochalasins D and B, to influence specific SC differentiation programs modulated by several agents (chemical or physical) or intra- and extra-cellular factors, with particular attention to human MSCs (hMSCs).
Collapse
Affiliation(s)
- Luca Pampanella
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Giovannamaria Petrocelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Provvidenza Maria Abruzzo
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Cinzia Zucchini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Silvia Canaider
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Carlo Ventura
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB) c/o Eldor Lab, Via Corticella 183, 40129 Bologna, Italy
| | - Federica Facchin
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| |
Collapse
|
3
|
Paolino A, Haines EH, Bailey EJ, Black DA, Moey C, García-Moreno F, Richards LJ, Suárez R, Fenlon LR. Non-uniform temporal scaling of developmental processes in the mammalian cortex. Nat Commun 2023; 14:5950. [PMID: 37741828 PMCID: PMC10517946 DOI: 10.1038/s41467-023-41652-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023] Open
Abstract
The time that it takes the brain to develop is highly variable across animals. Although staging systems equate major developmental milestones between mammalian species, it remains unclear how distinct processes of cortical development scale within these timeframes. Here, we compare the timing of cortical development in two mammals of similar size but different developmental pace: eutherian mice and marsupial fat-tailed dunnarts. Our results reveal that the temporal relationship between cell birth and laminar specification aligns to equivalent stages between these species, but that migration and axon extension do not scale uniformly according to the developmental stages, and are relatively more advanced in dunnarts. We identify a lack of basal intermediate progenitor cells in dunnarts that likely contributes in part to this timing difference. These findings demonstrate temporal limitations and differential plasticity of cortical developmental processes between similarly sized Therians and provide insight into subtle temporal changes that may have contributed to the early diversification of the mammalian brain.
Collapse
Affiliation(s)
- Annalisa Paolino
- The University of Queensland, School of Biomedical Sciences, Brisbane, QLD 4072, Australia
- The University of Queensland, Queensland Brain Institute, Brisbane, QLD 4072, Australia
| | - Elizabeth H Haines
- The University of Queensland, School of Biomedical Sciences, Brisbane, QLD 4072, Australia
- The University of Queensland, Queensland Brain Institute, Brisbane, QLD 4072, Australia
| | - Evan J Bailey
- The University of Queensland, School of Biomedical Sciences, Brisbane, QLD 4072, Australia
- The University of Queensland, Queensland Brain Institute, Brisbane, QLD 4072, Australia
| | - Dylan A Black
- The University of Queensland, Queensland Brain Institute, Brisbane, QLD 4072, Australia
| | - Ching Moey
- The University of Queensland, Queensland Brain Institute, Brisbane, QLD 4072, Australia
| | - Fernando García-Moreno
- Achucarro Basque Center for Neuroscience, Scientific Park of the University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
- IKERBASQUE Foundation, María Díaz de Haro 3, 48013, Bilbao, Spain
| | - Linda J Richards
- The University of Queensland, School of Biomedical Sciences, Brisbane, QLD 4072, Australia
- The University of Queensland, Queensland Brain Institute, Brisbane, QLD 4072, Australia
- Washington University in St Louis School of Medicine, Department of Neuroscience, St Louis, MO, 63108, USA
| | - Rodrigo Suárez
- The University of Queensland, School of Biomedical Sciences, Brisbane, QLD 4072, Australia.
- The University of Queensland, Queensland Brain Institute, Brisbane, QLD 4072, Australia.
| | - Laura R Fenlon
- The University of Queensland, School of Biomedical Sciences, Brisbane, QLD 4072, Australia.
- The University of Queensland, Queensland Brain Institute, Brisbane, QLD 4072, Australia.
| |
Collapse
|
4
|
Di Lisa D, Muzzi L, Pepe S, Dellacasa E, Frega M, Fassio A, Martinoia S, Pastorino L. On the way back from 3D to 2D: Chitosan promotes adhesion and development of neuronal networks onto culture supports. Carbohydr Polym 2022; 297:120049. [DOI: 10.1016/j.carbpol.2022.120049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022]
|
5
|
Okada M, Kawagoe Y, Sato Y, Nozumi M, Ishikawa Y, Tamada A, Yamazaki H, Sekino Y, Kanemura Y, Shinmyo Y, Kawasaki H, Kaneko N, Sawamoto K, Fujii Y, Igarashi M. Phosphorylation of GAP-43 T172 is a molecular marker of growing axons in a wide range of mammals including primates. Mol Brain 2021; 14:66. [PMID: 33832520 PMCID: PMC8034164 DOI: 10.1186/s13041-021-00755-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
GAP-43 is a vertebrate neuron-specific protein and that is strongly related to axon growth and regeneration; thus, this protein has been utilized as a classical molecular marker of these events and growth cones. Although GAP-43 was biochemically characterized more than a quarter century ago, how this protein is related to these events is still not clear. Recently, we identified many phosphorylation sites in the growth cone membrane proteins of rodent brains. Two phosphorylation sites of GAP-43, S96 and T172, were found within the top 10 hit sites among all proteins. S96 has already been characterized (Kawasaki et al., 2018), and here, phosphorylation of T172 was characterized. In vitro (cultured neurons) and in vivo, an antibody specific to phosphorylated T172 (pT172 antibody) specifically recognized cultured growth cones and growing axons in developing mouse neurons, respectively. Immunoblotting showed that pT172 antigens were more rapidly downregulated throughout development than those of pS96 antibody. From the primary structure, this phosphorylation site was predicted to be conserved in a wide range of animals including primates. In the developing marmoset brainstem and in differentiated neurons derived from human induced pluripotent stem cells, immunoreactivity with pT172 antibody revealed patterns similar to those in mice. pT172 antibody also labeled regenerating axons following sciatic nerve injury. Taken together, the T172 residue is widely conserved in a wide range of mammals including primates, and pT172 is a new candidate molecular marker for growing axons.
Collapse
Affiliation(s)
- Masayasu Okada
- Department of Neurosurgery, Brain Research Institute, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
- Medical and Dental Hospital, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Yosuke Kawagoe
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Yuta Sato
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
- Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Motohiro Nozumi
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Yuya Ishikawa
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
- Department of Orthopedic Surgery, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Atsushi Tamada
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan
- Department of iPS Cell Applied Medicine, Faculty of Medicine, Kansai Medical University, Hirakata, Osaka, 573-1010, Japan
| | - Hiroyuki Yamazaki
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yuko Sekino
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Yonehiro Kanemura
- Division of Regenerative Medicine, Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Naoko Kaneko
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, Japan
| | - Yukihiko Fujii
- Department of Neurosurgery, Brain Research Institute, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
- Medical and Dental Hospital, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, Japan
| | - Michihiro Igarashi
- Departments of Neurochemistry and Molecular Cell Biology, School of Medicine and Graduate School of Medical/Dental Sciences, Niigata University, Niigata, 951-8510, Japan.
| |
Collapse
|
6
|
Mishra P, Cohen RI, Zhao N, Moghe PV. Fluorescence-based actin turnover dynamics of stem cells as a profiling method for stem cell functional evolution, heterogeneity and phenotypic lineage parsing. Methods 2020; 190:44-54. [PMID: 32473293 DOI: 10.1016/j.ymeth.2020.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/24/2020] [Accepted: 05/26/2020] [Indexed: 02/07/2023] Open
Abstract
Stem cells are widely explored in regenerative medicine as a source to produce diverse cell types. Despite the wide usage of stem cells like mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs), there is a lack of robust methods to rapidly discern the phenotypic and functional heterogeneity of stem cells. The organization of actin cytoskeleton has been previously used to discern divergent stem cell differentiation pathways. In this paper, we highlight the versatility of a cell profiling method for actin turnover dynamics. Actin filaments in live stem cells are labeled using SiR-actin, a cell permeable fluorogenic probe, to determine the endogenous actin turnover. Live MSC imaging after days of induction successfully demonstrated lineage specific change in actin turnover. Next, we highlighted the differences in the cellular heterogeneity of actin dynamics during adipogenic or osteogenic MSC differentiation. Next, we applied the method to differentiating iPSCs in culture, and detected a progressive slowdown in actin turnover during differentiation upon stimulation with neural or cardiac media. Finally, as a proof of concept, the actin dynamic profiling was used to isolate MSCs via flow cytometry prior to sorting into three distinct sub-populations with low, intermediate or high actin dynamics. A greater fraction of MSCs with more rapid actin dynamics demonstrated increased inclination for adipogenesis, whereas, slower actin dynamics correlated with increased osteogenesis. Together, these results show that actin turnover can serve as a versatile biomarker to not only track cellular phenotypic heterogeneity but also harvest live cells with potential for differential phenotypic fates.
Collapse
Affiliation(s)
- Prakhar Mishra
- Molecular Biosciences Graduate Program in Cell and Developmental Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Ricky I Cohen
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Nanxia Zhao
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Prabhas V Moghe
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
7
|
Mahmoodi N, Ai J, Ebrahimi‐Barough S, Hassannejad Z, Hasanzadeh E, Basiri A, Vaccaro AR, Rahimi‐Movaghar V. Microtubule stabilizer epothilone B as a motor neuron differentiation agent for human endometrial stem cells. Cell Biol Int 2020; 44:1168-1183. [DOI: 10.1002/cbin.11315] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/02/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Narges Mahmoodi
- Sina Trauma and Surgery Research Center, Sina HospitalTehran University of Medical Sciences Hasan‐Abad Square, Imam Khomeini Ave. Tehran 11365‐3876 Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineTehran University of Medical Sciences Number 88, Italy Street, Between Ghods Street and Vesal Shirazi Street Tehran 14177‐55469 Iran
| | - Somayeh Ebrahimi‐Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineTehran University of Medical Sciences Number 88, Italy Street, Between Ghods Street and Vesal Shirazi Street Tehran 14177‐55469 Iran
| | - Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Pediatric Center of ExcellenceTehran University of Medical Sciences No. 62, Dr. Gharibs Street, Keshavarz Boulevard Tehran 1419733151 Iran
| | - Elham Hasanzadeh
- Department of Tissue Engineering, School of Advanced Technologies in MedicineMazandaran University of Medical Sciences Next to Tooba Medical Building, Khazar Boulevard Sari 48471‐91971 Iran
| | - Arefeh Basiri
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineTehran University of Medical Sciences Number 88, Italy Street, Between Ghods Street and Vesal Shirazi Street Tehran 14177‐55469 Iran
| | - Alexander R. Vaccaro
- Department of Orthopedic Surgery, Rothman InstituteThomas Jefferson University 1925 Chestnut Street, 5th Floor Philadelphia Pennsylvania 19107 USA
| | - Vafa Rahimi‐Movaghar
- Sina Trauma and Surgery Research Center, Sina HospitalTehran University of Medical Sciences Hasan‐Abad Square, Imam Khomeini Ave. Tehran 11365‐3876 Iran
| |
Collapse
|
8
|
Ishizuka Y, Bramham CR. A simple DMSO-based method for cryopreservation of primary hippocampal and cortical neurons. J Neurosci Methods 2020; 333:108578. [DOI: 10.1016/j.jneumeth.2019.108578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/20/2019] [Accepted: 12/30/2019] [Indexed: 01/19/2023]
|
9
|
Bueno C, Martínez-Morga M, Martínez S. Non-proliferative neurogenesis in human periodontal ligament stem cells. Sci Rep 2019; 9:18038. [PMID: 31792338 PMCID: PMC6888846 DOI: 10.1038/s41598-019-54745-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/15/2019] [Indexed: 12/16/2022] Open
Abstract
Understanding the sequence of events from undifferentiated stem cells to neuron is not only important for the basic knowledge of stem cell biology, but also for therapeutic applications. In this study we examined the sequence of biological events during neural differentiation of human periodontal ligament stem cells (hPDLSCs). Here, we show that hPDLSCs-derived neural-like cells display a sequence of morphologic development highly similar to those reported before in primary neuronal cultures derived from rodent brains. We observed that cell proliferation is not present through neurogenesis from hPDLSCs. Futhermore, we may have discovered micronuclei movement and transient cell nuclei lobulation coincident to in vitro neurogenesis. Morphological analysis also reveals that neurogenic niches in the adult mouse brain contain cells with nuclear shapes highly similar to those observed during in vitro neurogenesis from hPDLSCs. Our results provide additional evidence that it is possible to differentiate hPDLSCs to neuron-like cells and suggest the possibility that the sequence of events from stem cell to neuron does not necessarily requires cell division from stem cell.
Collapse
Affiliation(s)
- Carlos Bueno
- Instituto de Neurociencias de Alicante (UMH-CSIC), San Juan, Alicante, 03550, Spain.
| | - Marta Martínez-Morga
- Department of Human Anatomy and Institute of Biomedical Research (IMIB), University of Murcia, Faculty of Medicine, Murcia, 30800, Spain
| | - Salvador Martínez
- Instituto de Neurociencias de Alicante (UMH-CSIC), San Juan, Alicante, 03550, Spain
| |
Collapse
|
10
|
Puspitasari A, Yamazaki H, Kawamura H, Nakano T, Takahashi A, Shirao T, Held KD. X-irradiation of developing hippocampal neurons causes changes in neuron population phenotypes, dendritic morphology and synaptic protein expression in surviving neurons at maturity. Neurosci Res 2019; 160:11-24. [PMID: 31711782 DOI: 10.1016/j.neures.2019.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/16/2019] [Accepted: 11/06/2019] [Indexed: 01/02/2023]
Abstract
The effects of X-irradiation on developing neurons and their functions are unclear. We used primary cultures of mouse hippocampal neurons to investigate the effects of X-irradiation on cell death in developing neurons by analyzing caspase-3, MAP2 and DAPI-labeled cells, and the phenotypes and function of surviving neurons, by examining GAD67-positive cells as a GABAergic marker, and the synaptic markers synapsin 1, drebrin and PSD-95 through its maturation. One-day in vitro (DIV 1) cells were exposed to 0.5 Gy or 1 Gy of X-rays. A significant increase in the percentage of activated caspase-3, a decrease in the number of MAP2/DAPI-positive cells and change in the percentage of GAD67 positive neurons, compared with sham controls, were found 6 days after 1 Gy and 13 days after 0.5 Gy of X-rays. The expression of PSD-95 and drebrin, as well as drebrin clusters, in the remaining neurons was decreased at DIV 21, in both 0.5 Gy and on 1 Gy-irradiation there was a reduced number of dendritic intersection as well. Together, our findings show that 0.5 Gy and 1 Gy of X-irradiation at DIV 1 not only causes neuronal cell death but elicits an increase in the percentage of inhibitory neurons, changes in the dendrites and decrease in expression of important synaptic proteins in the surviving neurons at maturity 3 weeks after exposure.
Collapse
Affiliation(s)
| | - Hiroyuki Yamazaki
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hidemasa Kawamura
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Takashi Nakano
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akihisa Takahashi
- Gunma University Heavy Ion Medical Center, Maebashi, Graduate School of Medicine, Gunma, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kathryn D Held
- Gunma University Initiative for Advanced Research, Maebashi, Japan; Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Miki D, Kobayashi Y, Okada T, Miyamoto T, Takei N, Sekino Y, Koganezawa N, Shirao T, Saito Y. Characterization of Functional Primary Cilia in Human Induced Pluripotent Stem Cell-Derived Neurons. Neurochem Res 2019; 44:1736-1744. [PMID: 31037609 DOI: 10.1007/s11064-019-02806-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022]
Abstract
Recent advances in human induced pluripotent stem cells (hiPSCs) offer new possibilities for biomedical research and clinical applications. Neurons differentiated from hiPSCs may be promising tools to develop novel treatment methods for various neurological diseases. However, the detailed process underlying functional maturation of hiPSC-derived neurons remains poorly understood. Here, we analyze the developmental architecture of hiPSC-derived cortical neurons, iCell GlutaNeurons, focusing on the primary cilium, a single sensory organelle that protrudes from the surface of most growth-arrested vertebrate cells. To characterize the neuronal cilia, cells were cultured for various periods and evaluated immunohistochemically by co-staining with antibodies against ciliary markers Arl13b and MAP2. Primary cilia were detected in neurons within days, and their prevalence and length increased with increasing days in culture. Treatment with the mood stabilizer lithium led to primary cilia length elongation, while treatment with the orexigenic neuropeptide melanin-concentrating hormone caused cilia length shortening in iCell GlutaNeurons. The present findings suggest that iCell GlutaNeurons develop neuronal primary cilia together with the signaling machinery for regulation of cilia length. Our approach to the primary cilium as a cellular antenna can be useful for both assessment of neuronal maturation and validation of pharmaceutical agents in hiPSC-derived neurons.
Collapse
Affiliation(s)
- Daisuke Miki
- Graduate School of Integrated Arts and Sciences, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Yuki Kobayashi
- Graduate School of Integrated Arts and Sciences, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Tomoya Okada
- Graduate School of Integrated Arts and Sciences, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan
| | - Tatuso Miyamoto
- Department of Genetics and Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Nobuyuki Takei
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Yuko Sekino
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
| | - Noriko Koganezawa
- Department of Neurobiology and Behavior, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Graduate School of Medicine, Gunma University, Maebashi, 371-8511, Japan
| | - Yumiko Saito
- Graduate School of Integrated Arts and Sciences, Hiroshima University, 1-7-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8521, Japan.
| |
Collapse
|
12
|
Masjosthusmann S, Becker D, Petzuch B, Klose J, Siebert C, Deenen R, Barenys M, Baumann J, Dach K, Tigges J, Hübenthal U, Köhrer K, Fritsche E. A transcriptome comparison of time-matched developing human, mouse and rat neural progenitor cells reveals human uniqueness. Toxicol Appl Pharmacol 2018; 354:40-55. [PMID: 29753005 DOI: 10.1016/j.taap.2018.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 12/12/2022]
Abstract
It is widely accepted that human brain development has unique features that cannot be represented by rodents. Obvious reasons are the evolutionary distance and divergent physiology. This might lead to false predictions when rodents are used for safety or pharmacological efficacy studies. For a better translation of animal-based research to the human situation, human in vitro systems might be useful. In this study, we characterize developing neural progenitor cells from prenatal human and time-matched rat and mouse brains by analyzing the changes in their transcriptome profile during neural differentiation. Moreover, we identify hub molecules that regulate neurodevelopmental processes like migration and differentiation. Consequences of modulation of three of those hubs on these processes were studied in a species-specific context. We found that although the gene expression profiles of the three species largely differ qualitatively and quantitatively, they cluster in similar GO terms like cell migration, gliogenesis, neurogenesis or development of multicellular organism. Pharmacological modulation of the identified hub molecules triggered species-specific cellular responses. This study underlines the importance of understanding species differences on the molecular level and advocates the use of human based in vitro models for pharmacological and toxicological research.
Collapse
Affiliation(s)
- Stefan Masjosthusmann
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Daniel Becker
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany
| | - Barbara Petzuch
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Jördis Klose
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Clara Siebert
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Rene Deenen
- Biological and Medical Research Centre (BMFZ), Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Duesseldorf, NRW, Germany.
| | - Marta Barenys
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Jenny Baumann
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany
| | - Katharina Dach
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany; Department of Molecular Biosciences, University of California-Davis School of Veterinary Medicine, Davis, CA 95616, United States.
| | - Julia Tigges
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Ulrike Hübenthal
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany.
| | - Karl Köhrer
- Biological and Medical Research Centre (BMFZ), Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Duesseldorf, NRW, Germany.
| | - Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, NRW, Germany; Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Duesseldorf, NRW, Germany.
| |
Collapse
|
13
|
Drebrin in Neuronal Migration and Axonal Growth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1006:141-155. [PMID: 28865019 DOI: 10.1007/978-4-431-56550-5_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
During development, production of neurons from neural stem cells, migration of neurons from their birthplace to their final location, and extension of neurites, axons, and dendrites are important for the formation of functional neuronal circuits. The actin cytoskeleton has major roles in the morphological development of neurons. In this chapter, we focused on the distribution and function of the actin-binding protein, drebrin, to elucidate the importance of drebrin-bound F-actin in neurons during early developmental stages of neurons in embryonic, postnatal, and adult brains. There are three major isoforms of drebrin in the chicken brain (E1, E2, and A) and two major isoforms in the mammalian brain (E and A). Among these drebrin isoforms, drebrin E1 and E2 in chicken and drebrin E in the mammalian brain are involved in these neuronal stages. In migrating neurons of the developing and adult brain, drebrin is localized at the base of filopodia of leading processes, to regulate neuronal migration. In axonal growth cones, drebrin is localized in the transitional zone to regulate axonal growth by inhibiting actomyosin interactions and mediating the interactions between F-actin and microtubules. For axonal collateral branching, drebrin is localized at axonal actin patches and the base of filopodia, to accelerate the transition from actin patches to filopodia and stabilize the filopodia.
Collapse
|
14
|
Koganezawa N, Hanamura K, Shirao T. Progress in applications of iPSC-derived neurons for evaluation of drugs. Nihon Yakurigaku Zasshi 2017; 149:104-109. [PMID: 28260738 DOI: 10.1254/fpj.149.104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
15
|
Amin H, Maccione A, Marinaro F, Zordan S, Nieus T, Berdondini L. Electrical Responses and Spontaneous Activity of Human iPS-Derived Neuronal Networks Characterized for 3-month Culture with 4096-Electrode Arrays. Front Neurosci 2016; 10:121. [PMID: 27065786 PMCID: PMC4811967 DOI: 10.3389/fnins.2016.00121] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/11/2016] [Indexed: 12/20/2022] Open
Abstract
The recent availability of human induced pluripotent stem cells (hiPSCs) holds great promise as a novel source of human-derived neurons for cell and tissue therapies as well as for in vitro drug screenings that might replace the use of animal models. However, there is still a considerable lack of knowledge on the functional properties of hiPSC-derived neuronal networks, thus limiting their application. Here, upon optimization of cell culture protocols, we demonstrate that both spontaneous and evoked electrical spiking activities of these networks can be characterized on-chip by taking advantage of the resolution provided by CMOS multielectrode arrays (CMOS-MEAs). These devices feature a large and closely-spaced array of 4096 simultaneously recording electrodes and multi-site on-chip electrical stimulation. Our results show that networks of human-derived neurons can respond to electrical stimulation with a physiological repertoire of spike waveforms after 3 months of cell culture, a period of time during which the network undergoes the expression of developing patterns of spontaneous spiking activity. To achieve this, we have investigated the impact on the network formation and on the emerging network-wide functional properties induced by different biochemical substrates, i.e., poly-dl-ornithine (PDLO), poly-l-ornithine (PLO), and polyethylenimine (PEI), that were used as adhesion promoters for the cell culture. Interestingly, we found that neuronal networks grown on PDLO coated substrates show significantly higher spontaneous firing activity, reliable responses to low-frequency electrical stimuli, and an appropriate level of PSD-95 that may denote a physiological neuronal maturation profile and synapse stabilization. However, our results also suggest that even 3-month culture might not be sufficient for human-derived neuronal network maturation. Taken together, our results highlight the tight relationship existing between substrate coatings and emerging network properties, i.e., spontaneous activity, responsiveness, synapse formation and maturation. Additionally, our results provide a baseline on the functional properties expressed over 3 months of network development for a commercially available line of hiPSC-derived neurons. This is a first step toward the development of functional pre-clinical assays to test pharmaceutical compounds on human-derived neuronal networks with CMOS-MEAs.
Collapse
Affiliation(s)
- Hayder Amin
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Alessandro Maccione
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Federica Marinaro
- Neurobiology of miRNA Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Stefano Zordan
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Thierry Nieus
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Luca Berdondini
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| |
Collapse
|