1
|
Han R, Luo L, Wei C, Qiao Y, Xie J, Pan X, Xing J. Stiffness-tunable biomaterials provide a good extracellular matrix environment for axon growth and regeneration. Neural Regen Res 2025; 20:1364-1376. [PMID: 39075897 PMCID: PMC11624885 DOI: 10.4103/nrr.nrr-d-23-01874] [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: 11/14/2023] [Revised: 01/31/2024] [Accepted: 03/16/2024] [Indexed: 07/31/2024] Open
Abstract
Neuronal growth, extension, branching, and formation of neural networks are markedly influenced by the extracellular matrix-a complex network composed of proteins and carbohydrates secreted by cells. In addition to providing physical support for cells, the extracellular matrix also conveys critical mechanical stiffness cues. During the development of the nervous system, extracellular matrix stiffness plays a central role in guiding neuronal growth, particularly in the context of axonal extension, which is crucial for the formation of neural networks. In neural tissue engineering, manipulation of biomaterial stiffness is a promising strategy to provide a permissive environment for the repair and regeneration of injured nervous tissue. Recent research has fine-tuned synthetic biomaterials to fabricate scaffolds that closely replicate the stiffness profiles observed in the nervous system. In this review, we highlight the molecular mechanisms by which extracellular matrix stiffness regulates axonal growth and regeneration. We highlight the progress made in the development of stiffness-tunable biomaterials to emulate in vivo extracellular matrix environments, with an emphasis on their application in neural repair and regeneration, along with a discussion of the current limitations and future prospects. The exploration and optimization of the stiffness-tunable biomaterials has the potential to markedly advance the development of neural tissue engineering.
Collapse
Affiliation(s)
- Ronglin Han
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Lanxin Luo
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Caiyan Wei
- Department of Medicinal Chemistry, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yaru Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jiming Xie
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xianchao Pan
- Department of Medicinal Chemistry, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Juan Xing
- Department of Pathophysiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
2
|
Akere MT, Zajac KK, Bretz JD, Madhavaram AR, Horton AC, Schiefer IT. Real-Time Analysis of Neuronal Cell Cultures for CNS Drug Discovery. Brain Sci 2024; 14:770. [PMID: 39199464 PMCID: PMC11352746 DOI: 10.3390/brainsci14080770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/23/2024] [Accepted: 07/27/2024] [Indexed: 09/01/2024] Open
Abstract
The ability to screen for agents that can promote the development and/or maintenance of neuronal networks creates opportunities for the discovery of novel agents for the treatment of central nervous system (CNS) disorders. Over the past 10 years, advances in robotics, artificial intelligence, and machine learning have paved the way for the improved implementation of live-cell imaging systems for drug discovery. These instruments have revolutionized our ability to quickly and accurately acquire large standardized datasets when studying complex cellular phenomena in real-time. This is particularly useful in the field of neuroscience because real-time analysis can allow efficient monitoring of the development, maturation, and conservation of neuronal networks by measuring neurite length. Unfortunately, due to the relative infancy of this type of analysis, standard practices for data acquisition and processing are lacking, and there is no standardized format for reporting the vast quantities of data generated by live-cell imaging systems. This paper reviews the current state of live-cell imaging instruments, with a focus on the most commonly used equipment (IncuCyte systems). We provide an in-depth analysis of the experimental conditions reported in publications utilizing these systems, particularly with regard to studying neurite outgrowth. This analysis sheds light on trends and patterns that will enhance the use of live-cell imaging instruments in CNS drug discovery.
Collapse
Affiliation(s)
- Millicent T. Akere
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
| | - Kelsee K. Zajac
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
| | - James D. Bretz
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
| | - Anvitha R. Madhavaram
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
| | - Austin C. Horton
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
| | - Isaac T. Schiefer
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (M.T.A.); (K.K.Z.); (J.D.B.); (A.R.M.); (A.C.H.)
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
3
|
Nepon H, Allgayer R, Julien C, Petrecca S, Kalashnikov N, Safran T, Murphy A, Dionisopolous T, Davison P, Cerruti M, Vorstenbosch J. Altered Foreign Body Response at the Posterior Surface Compared to the Anterior Surface of Human Silicone Breast Implants. ACS Biomater Sci Eng 2024; 10:3006-3016. [PMID: 38640484 DOI: 10.1021/acsbiomaterials.3c01961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Soft implantable devices are crucial to optimizing form and function for many patients. However, periprosthetic capsule fibrosis is one of the major challenges limiting the use of implants. Currently, little is understood about how spatial and temporal factors influence capsule physiology and how the local capsule environment affects the implant structure. In this work, we analyzed breast implant capsule specimens with staining, immunohistochemistry, and real-time polymerase chain reaction to investigate spatiotemporal differences in inflammation and fibrosis. We demonstrated that in comparison to the anterior capsule against the convex surface of breast implants, the posterior capsule against the flat surface of the breast implant displays several features of a dysregulated foreign body reaction including increased capsule thickness, abnormal extracellular remodeling, and infiltration of macrophages. Furthermore, the expression of pro-inflammatory cytokines increased in the posterior capsule across the lifespan of the device, but not in the anterior capsule. We also analyzed the surface oxidation of breast explant samples with XPS analysis. No significant differences in surface oxidation were identified either spatially or temporally. Collectively, our results support spatiotemporal heterogeneity in inflammation and fibrosis within the breast implant capsule. These findings presented here provide a more detailed picture of the complexity of the foreign body reaction surrounding implants destined for human use and could lead to key research avenues and clinical applications to treat periprosthetic fibrosis and improve device longevity.
Collapse
Affiliation(s)
- Hillary Nepon
- Division of Surgical and Interventional Sciences, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Raphaela Allgayer
- Department of Materials Engineering, McGill University, Wong Building, 3610 Rue University, Montreal, Quebec H3A 0C5, Canada
| | - Cedric Julien
- Research Institute of the McGill University Health Centre, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Sarah Petrecca
- Faculty of Medicine and Health Sciences, McGill University, 3605 de la Montagne, Montreal, Quebec H3G 1M1, Canada
| | - Nikita Kalashnikov
- Division of Surgical and Interventional Sciences, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
- Faculty of Medicine and Health Sciences, McGill University, 3605 de la Montagne, Montreal, Quebec H3G 1M1, Canada
| | - Tyler Safran
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Amanda Murphy
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Tassos Dionisopolous
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Peter Davison
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| | - Marta Cerruti
- Department of Materials Engineering, McGill University, Wong Building, 3610 Rue University, Montreal, Quebec H3A 0C5, Canada
| | - Joshua Vorstenbosch
- Division of Plastic & Reconstructive Surgery, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
- Research Institute of the McGill University Health Centre, McGill University, Montreal General Hospital, 1650 Cedar Avenue, Room T5-204, Montreal, Quebec H3G 1A4, Canada
| |
Collapse
|
4
|
Bryniarska-Kubiak N, Basta-Kaim A, Kubiak A. Mechanobiology of Dental Pulp Cells. Cells 2024; 13:375. [PMID: 38474339 PMCID: PMC10931140 DOI: 10.3390/cells13050375] [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: 12/23/2023] [Revised: 02/06/2024] [Accepted: 02/11/2024] [Indexed: 03/14/2024] Open
Abstract
The dental pulp is the inner part of the tooth responsible for properly functioning during its lifespan. Apart from the very big biological heterogeneity of dental cells, tooth microenvironments differ a lot in the context of mechanical properties-ranging from 5.5 kPa for dental pulp to around 100 GPa for dentin and enamel. This physical heterogeneity and complexity plays a key role in tooth physiology and in turn, is a great target for a variety of therapeutic approaches. First of all, physical mechanisms are crucial for the pain propagation process from the tooth surface to the nerves inside the dental pulp. On the other hand, the modulation of the physical environment affects the functioning of dental pulp cells and thus is important for regenerative medicine. In the present review, we describe the physiological significance of biomechanical processes in the physiology and pathology of dental pulp. Moreover, we couple those phenomena with recent advances in the fields of bioengineering and pharmacology aiming to control the functioning of dental pulp cells, reduce pain, and enhance the differentiation of dental cells into desired lineages. The reviewed literature shows great progress in the topic of bioengineering of dental pulp-although mainly in vitro. Apart from a few positions, it leaves a gap for necessary filling with studies providing the mechanisms of the mechanical control of dental pulp functioning in vivo.
Collapse
Affiliation(s)
- Natalia Bryniarska-Kubiak
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland;
- Laboratory of Stem Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa St., 30-387 Kraków, Poland
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland;
| | - Andrzej Kubiak
- Laboratory of Stem Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa St., 30-387 Kraków, Poland
| |
Collapse
|
5
|
Abstract
Recently, substrate stiffness has been involved in the physiology and pathology of the nervous system. However, the role and function of substrate stiffness remain unclear. Here, we review known effects of substrate stiffness on nerve cell morphology and function in the central and peripheral nervous systems and their involvement in pathology. We hope this review will clarify the research status of substrate stiffness in nerve cells and neurological disorder.
Collapse
Affiliation(s)
- Weijin Si
- Key Laboratory of Cognitive Science, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Jihong Gong
- Key Laboratory of Cognitive Science, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| | - Xiaofei Yang
- Key Laboratory of Cognitive Science, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, Laboratory of Membrane Ion Channels and Medicine, College of Biomedical Engineering, South-Central Minzu University, Wuhan 430074, China
| |
Collapse
|
6
|
Batzdorf CS, Morr AS, Bertalan G, Sack I, Silva RV, Infante-Duarte C. Sexual Dimorphism in Extracellular Matrix Composition and Viscoelasticity of the Healthy and Inflamed Mouse Brain. BIOLOGY 2022; 11:biology11020230. [PMID: 35205095 PMCID: PMC8869215 DOI: 10.3390/biology11020230] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/21/2022] [Accepted: 01/28/2022] [Indexed: 12/13/2022]
Abstract
Simple Summary In multiple sclerosis (MS), an autoimmune disease of the central nervous system that primarily affects women, gender differences in disease course and in brain softening have been reported. It has been shown that the molecular network found between the cells of the tissue, the extracellular matrix (ECM), influences tissue stiffness. However, it is still unclear if sex influences ECM composition. Therefore, here we investigated how brain ECM and stiffness differ between sexes in the healthy mouse, and in an MS mouse model. We applied multifrequency magnetic resonance elastography and gene expression analysis for associating in vivo brain stiffness with ECM protein content in the brain, such as collagen and laminin. We found that the cortex was softer in males than in females in both healthy and sick mice. Softening was associated with sex differences in expression levels of collagen and laminin. Our findings underscore the importance of considering sex when studying the constitution of brain tissue in health and disease, particularly when investigating the processes underlying gender differences in MS. Abstract Magnetic resonance elastography (MRE) has revealed sexual dimorphism in brain stiffness in healthy individuals and multiple sclerosis (MS) patients. In an animal model of MS, named experimental autoimmune encephalomyelitis (EAE), we have previously shown that inflammation-induced brain softening was associated with alterations of the extracellular matrix (ECM). However, it remained unclear whether the brain ECM presents sex-specific properties that can be visualized by MRE. Therefore, here we aimed at quantifying sexual dimorphism in brain viscoelasticity in association with ECM changes in healthy and inflamed brains. Multifrequency MRE was applied to the midbrain of healthy and EAE mice of both sexes to quantitatively map regional stiffness. To define differences in brain ECM composition, the gene expression of the key basement membrane components laminin (Lama4, Lama5), collagen (Col4a1, Col1a1), and fibronectin (Fn1) were investigated by RT-qPCR. We showed that the healthy male cortex expressed less Lama4, Lama5, and Col4a1, but more Fn1 (all p < 0.05) than the healthy female cortex, which was associated with 9% softer properties (p = 0.044) in that region. At peak EAE cortical softening was similar in both sexes compared to healthy tissue, with an 8% difference remaining between males and females (p = 0.006). Cortical Lama4, Lama5 and Col4a1 expression increased 2 to 3-fold in EAE in both sexes while Fn1 decreased only in males (all p < 0.05). No significant sex differences in stiffness were detected in other brain regions. In conclusion, sexual dimorphism in the ECM composition of cortical tissue in the mouse brain is reflected by in vivo stiffness measured with MRE and should be considered in future studies by sex-specific reference values.
Collapse
Affiliation(s)
- Clara Sophie Batzdorf
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Lindenberger Weg 80, 13125 Berlin, Germany; (C.S.B.); (R.V.S.)
| | - Anna Sophie Morr
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (A.S.M.); (G.B.); (I.S.)
| | - Gergely Bertalan
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (A.S.M.); (G.B.); (I.S.)
| | - Ingolf Sack
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany; (A.S.M.); (G.B.); (I.S.)
| | - Rafaela Vieira Silva
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Lindenberger Weg 80, 13125 Berlin, Germany; (C.S.B.); (R.V.S.)
- Einstein Center for Neurosciences Berlin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Carmen Infante-Duarte
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Lindenberger Weg 80, 13125 Berlin, Germany; (C.S.B.); (R.V.S.)
- Correspondence:
| |
Collapse
|
7
|
Macha P, Mayes ML, Visayas BRB, Soni V, Sammeta VR, Vasudev MC. Influence of dityrosine nanotubes on the expression of dopamine and differentiation in neural cells. J Mater Chem B 2021; 9:3900-3911. [PMID: 33928965 DOI: 10.1039/d0tb02680h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, we report the synthesis of self-assembled dityrosine nanotubes as a biologically functional scaffold and their interactions with neural cells. Quantum chemical methods were used to determine the forces involved in the self-assembly process. The physicochemical properties of the nanostructures relevant to their potential as bioactive scaffolds were characterized. The morphology, secondary structure, crystallinity, mechanical properties, and thermal characteristics of YY nanotubes were analyzed. The influence of these nanotubes as scaffolds for neural cells was studied in vitro to understand their effects on cell proliferation, morphology, and gene expression. The scanning electron microscopy and fluorescence confocal microscopy demonstrated the feasibility of nanotube scaffolds for enhanced adhesion to rat and human neural cells (PC12 and SH-SY5Y). Preliminary ELISA and qPCR analyses demonstrate the upregulation of dopamine synthesis and genes involved in dopamine expression and differentiation. The expression levels of DβH, AADC, VMAT2 and MAOA in SH-SY5Y cells cultured on the nanotube scaffolds for 7 days were elevated in comparison to the control cells.
Collapse
Affiliation(s)
- Prathyushakrishna Macha
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA.
| | - Maricris L Mayes
- Department of Chemistry and Biochemistry, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA
| | - Benjoe Rey B Visayas
- Department of Chemistry and Biochemistry, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA
| | - Vikas Soni
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA. and Department of Mechanical Engineering, George Washington University, DC 20052, USA
| | | | - Milana C Vasudev
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA.
| |
Collapse
|
8
|
Physical and Mechanical Characterization of Fibrin-Based Bioprinted Constructs Containing Drug-Releasing Microspheres for Neural Tissue Engineering Applications. Processes (Basel) 2021. [DOI: 10.3390/pr9071205] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Three-dimensional bioprinting can fabricate precisely controlled 3D tissue constructs. This process uses bioinks—specially tailored materials that support the survival of incorporated cells—to produce tissue constructs. The properties of bioinks, such as stiffness and porosity, should mimic those found in desired tissues to support specialized cell types. Previous studies by our group validated soft substrates for neuronal cultures using neural cells derived from human-induced pluripotent stem cells (hiPSCs). It is important to confirm that these bioprinted tissues possess mechanical properties similar to native neural tissues. Here, we assessed the physical and mechanical properties of bioprinted constructs generated from our novel microsphere containing bioink. We measured the elastic moduli of bioprinted constructs with and without microspheres using a modified Hertz model. The storage and loss modulus, viscosity, and shear rates were also measured. Physical properties such as microstructure, porosity, swelling, and biodegradability were also analyzed. Our results showed that the elastic modulus of constructs with microspheres was 1032 ± 59.7 Pascal (Pa), and without microspheres was 728 ± 47.6 Pa. Mechanical strength and printability were significantly enhanced with the addition of microspheres. Thus, incorporating microspheres provides mechanical reinforcement, which indicates their suitability for future applications in neural tissue engineering.
Collapse
|
9
|
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
|
10
|
Advances in 3D neuronal microphysiological systems: towards a functional nervous system on a chip. In Vitro Cell Dev Biol Anim 2021; 57:191-206. [PMID: 33438114 PMCID: PMC7802613 DOI: 10.1007/s11626-020-00532-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/16/2020] [Indexed: 12/18/2022]
Abstract
Microphysiological systems (MPS) designed to study the complexities of the peripheral and central nervous systems have made marked improvements over the years and have allowed researchers to assess in two and three dimensions the functional interconnectivity of neuronal tissues. The recent generation of brain organoids has further propelled the field into the nascent recapitulation of structural, functional, and effective connectivities which are found within the native human nervous system. Herein, we will review advances in culture methodologies, focused especially on those of human tissues, which seek to bridge the gap from 2D cultures to hierarchical and defined 3D MPS with the end goal of developing a robust nervous system-on-a-chip platform. These advances have far-reaching implications within basic science, pharmaceutical development, and translational medicine disciplines.
Collapse
|
11
|
Abstract
The brain is our most complex organ. During development, neurons extend axons, which may grow over long distances along well-defined pathways to connect to distant targets. Our current understanding of axon pathfinding is largely based on chemical signaling by attractive and repulsive guidance cues. These cues instruct motile growth cones, the leading tips of growing axons, where to turn and where to stop. However, it is not chemical signals that cause motion-motion is driven by forces. Yet our current understanding of the mechanical regulation of axon growth is very limited. In this review, I discuss the origin of the cellular forces controlling axon growth and pathfinding, and how mechanical signals encountered by growing axons may be integrated with chemical signals. This mechanochemical cross talk is an important but often overlooked aspect of cell motility that has major implications for many physiological and pathological processes involving neuronal growth.
Collapse
Affiliation(s)
- Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, United Kingdom;
| |
Collapse
|
12
|
Chao MW, Kuo HC, Tong SY, Yang YS, Chuang YC, Tseng CY. In Vitro and In Vivo Analysis of the Effects of 3,5-DMA and Its Metabolites in Neural Oxidative Stress and Neurodevelopmental Toxicity. Toxicol Sci 2020; 168:405-419. [PMID: 30590852 DOI: 10.1093/toxsci/kfy306] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
3,5-Dimethylaniline (3,5-DMA), a monocyclic aromatic amine, is widely present in a spectrum of sources including tobacco, dyes, combustion products, and suspended particulates. 3,5-DMA and its metabolites form superoxides, resulting in apoptosis or oncogenesis. Data of a direct effect of 3,5-DMA on the nervous system, especially the developing brain, are lacking. Therefore, we investigated the effects of 3,5-DMA and its metabolites on fetal neurite growth and brain development using in vitro cell cultures of primary cortical neurons to observe whether these compounds caused neuronal cytotoxicity and affected neurite structural development. With increasing concentrations of 3,5-DMA (10, 50, 100, 500, 1000 μM) and its major metabolite 5-dimethylaminophenol (3,5-DMAP) (10, 50, 100, 500, 1000 μM), reactive oxygen species (ROS), cytotoxicity, and DNA damage increased significantly in the cells and dendritic arborization decreased. The addition of 5 mM N-acetylcysteine, an ROS scavenger, reduced ROS in the cells and alleviated the neuronal damage. In vivo studies in Sprague Dawley pregnant rats suggested that exposure to 3,5-DMA (10, 30, 60, 100 mg/kg/day) subcutaneously from GD15 to GD17 led to fetal cerebral cortex thinning. BrdU labeling showed that 3,5-DMA reduced the number and generation of cortical cells. To detect the laminar position of newly generated neurons, cortex layer markers such as Satb2, Ctip2, and Tbr1 were used. 3,5-DMA perturbed the cortical layer distribution in developing fetal rats. In summary, this is the first study to provide evidence for 3,5-DMA and its metabolites causing anomalies of the fetal central nervous system development through ROS production.
Collapse
Affiliation(s)
- Ming-Wei Chao
- Department of Bioscience Technology, Chung Yuan Christian University, Zhongli District, Taoyuan 32023, Taiwan.,Center for Nanotechnology, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| | - Hui-Chuan Kuo
- Department of Pharmacy, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 33004, Taiwan
| | - Sih-Yu Tong
- Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| | - Yu-Shiu Yang
- Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| | - Yu-Chen Chuang
- Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| | - Chia-Yi Tseng
- Department of Bioscience Technology, Chung Yuan Christian University, Zhongli District, Taoyuan 32023, Taiwan.,Center for Nanotechnology, Chung Yuan Christian University, Taoyuan 32023, Taiwan.,Department of Biomedical Engineering, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| |
Collapse
|
13
|
Yamamoto H, Grob L, Sumi T, Oiwa K, Hirano-Iwata A, Wolfrum B. Ultrasoft Silicone Gel as a Biomimetic Passivation Layer in Inkjet-Printed 3D MEA Devices. ACTA ACUST UNITED AC 2019; 3:e1900130. [PMID: 32648655 DOI: 10.1002/adbi.201900130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 06/25/2019] [Indexed: 12/15/2022]
Abstract
Multielectrode arrays (MEAs) are versatile tools that are used for chronic recording and stimulation of neural cells and tissues. Driven by the recent progress in understanding of how neuronal growth and function respond to scaffold stiffness, development of MEAs with a soft cell-to-device interface has gained importance not only for in vivo but also for in vitro applications. However, the passivation layer, which constitutes the majority of the cell-device interface, is typically prepared with stiff materials. Herein, a fabrication of an MEA device with an ultrasoft passivation layer is described, which takes advantage of inkjet printing and a polydimethylsiloxane (PDMS) gel with a stiffness comparable to that of the brain. The major challenge in using the PDMS gel is that it cannot be patterned to expose the sensing area of the electrode. This issue is resolved by printing 3D micropillars at the electrode tip. Primary cortical neurons are grown on the fabricated device, and effective stimulation of the culture confirms functional cell-device coupling. The 3D MEA device with an ultrasoft interface provides a novel platform for investigating evoked activity and drug responses of living neuronal networks cultured in a biomimetic environment for both fundamental research and pharmaceutical applications.
Collapse
Affiliation(s)
- Hideaki Yamamoto
- WPI-Advanced Institute for Materials Research (WPI-AIMR), Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan.,Institute for Advanced Study, Technische Universität München, Lichtenbergstraße 2a, 85748, Garching, Germany
| | - Leroy Grob
- Munich School of BioengineeringDepartment of Electrical and Computer Engineering, Technische Universität München, Boltzmannstraße 11, 85748, Garching, Germany
| | - Takuma Sumi
- Research Institute of Electrical Communication, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan
| | - Kazuhiro Oiwa
- Advanced ICT Research Institute, National Institute of Information and Communication Technology, 588-2 Iwaoka, Iwaoka-cho, Nishi-ku, Kobe, 651-2492, Japan
| | - Ayumi Hirano-Iwata
- WPI-Advanced Institute for Materials Research (WPI-AIMR), Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan.,Research Institute of Electrical Communication, Tohoku University, 2-1-1 Katahira, Aoba-ku, Sendai, 980-8577, Japan
| | - Bernhard Wolfrum
- Munich School of BioengineeringDepartment of Electrical and Computer Engineering, Technische Universität München, Boltzmannstraße 11, 85748, Garching, Germany
| |
Collapse
|
14
|
Moxon SR, Corbett NJ, Fisher K, Potjewyd G, Domingos M, Hooper NM. Blended alginate/collagen hydrogels promote neurogenesis and neuronal maturation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109904. [PMID: 31499954 PMCID: PMC6873778 DOI: 10.1016/j.msec.2019.109904] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/23/2019] [Accepted: 06/17/2019] [Indexed: 12/30/2022]
Abstract
Brain extracellular matrix (ECM) is complex, heterogeneous and often poorly replicated in traditional 2D cell culture systems. The development of more physiologically relevant 3D cell models capable of emulating the native ECM is of paramount importance for the study of human induced pluripotent stem cell (iPSC)-derived neurons. Due to its structural similarity with hyaluronic acid, a primary component of brain ECM, alginate is a potential biomaterial for 3D cell culture systems. However, a lack of cell adhesion motifs within the chemical structure of alginate has limited its application in neural culture systems. This study presents a simple and accessible method of incorporating collagen fibrils into an alginate hydrogel by physical mixing and controlled gelation under physiological conditions and tests the hypothesis that such a substrate could influence the behaviour of human neurons in 3D culture. Regulation of the gelation process enabled the penetration of collagen fibrils throughout the hydrogel structure as demonstrated by transmission electron microscopy. Encapsulated human iPSC-derived neurons adhered to the blended hydrogel as evidenced by the increased expression of α1, α2 and β1 integrins. Furthermore, immunofluorescence microscopy revealed that encapsulated neurons formed complex neural networks and matured into branched neurons expressing synaptophysin, a key protein involved in neurotransmission, along the neurites. Mechanical tuning of the hydrogel stiffness by modulation of the alginate ionic crosslinker concentration also influenced neuron-specific gene expression. In conclusion, we have shown that by tuning the physicochemical properties of the alginate/collagen blend it is possible to create different ECM-like microenvironments where complex mechanisms underpinning the growth and development of human neurons can be simulated and systematically investigated. Alginate and collagen are blended to create a bespoke hydrogel that mimics aspects of brain ECM. Encapsulated human pluripotent stem cell derived neurons adhere to the hydrogel matrix and form 3D neural networks. Neuronal differentiation and maturation is promoted within the hydrogel matrix. Mechanical properties of the hydrogel can be easily tuned to optimise neurogenesis. The hydrogel presents a platform for studying neuronal function and dysfunction in health and disease.
Collapse
Affiliation(s)
- Samuel R Moxon
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK
| | - Nicola J Corbett
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK
| | - Kate Fisher
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK
| | - Geoffrey Potjewyd
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK; School of Mechanical, Aerospace and Civil Engineering, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
| | - Marco Domingos
- School of Mechanical, Aerospace and Civil Engineering, Faculty of Science and Engineering, The University of Manchester, Manchester M13 9PL, UK
| | - Nigel M Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK.
| |
Collapse
|
15
|
Aydoğan Y, Altay M, Ünsal O, Kaplanoğlu V, Çağır Y, Yıldız C, Beyan E, Ramadan SU. An assessment of the relationship between thyroid nodule characteristics, insulin resistance and arterial stiffness in euthyroid nodular goiter. Endocrine 2018; 62:440-447. [PMID: 30084100 DOI: 10.1007/s12020-018-1701-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/26/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVES Publications suggesting that thyroid nodule might be associated with insulin resistance and metabolic syndrome are quite interesting. There is a need for studies assessing the relationship between nodule presence and cardiovascular risk in individuals with non-functioning nodular goiter. The purpose of the present study is to reveal whether or not insulin resistance, nodule presence, and nodule stiffness affect arterial stiffness, which is a reliable and valid cardiovascular risk indicator, in individuals with euthyroid nodular goiter using the pulse wave analysis (PWA). MATERIALS AND METHODS 50 patients with euthyroid nodular goiter and 50 healthy volunteers were included in the study. All participants were examined by B-mode thyroid ultrasound, and the participants in the nodular goiter group were also examined by strain elastography (SE). The strain index of nodules was calculated according to the Rago scoring. Also, fasting plasma glucose (FPG) and insulin levels were measured, and HOMA-IR. Arterial stiffness measurements of the participants were performed using a PWA device which employs a cuff-based oscillometric method from the brachial artery. RESULTS PWV was found to be significantly higher in the euthyroid nodular goiter group (p < 0.001). PWV was found to be positively correlated with FPG and waist circumference. Fasting plasma glucose was found to be higher in the group with nodular goiter (p = 0.03). However, no difference was found between the groups in terms of HOMA-IR and insulin level. HOMA-IR was not correlated with thyroid volume, nodule volume, and nodule count. Also, HOMA-IR was not correlated with strain index value and PWA data. CONCLUSION We found that PWV was significantly higher in patients with euthyroid nodular goiter. This result suggests that these patients may be at risk for cardiovascular disease.
Collapse
Affiliation(s)
| | - Mustafa Altay
- Department of Endocrinology, Keçiören Training and Research Hospital, Ankara, Turkey
| | - Oktay Ünsal
- Department of Nefrology, Uludağ University Medical Faculty Hospital, Bursa, Turkey
| | - Veysel Kaplanoğlu
- Department of Radiology, Keçiören Training ad Research Hospital, Ankara, Turkey
| | - Yavuz Çağır
- Çankırı Çerkeş State Hospital, Çankırı, Turkey
| | - Canan Yıldız
- Aydın Didim State Hospital, Aydın, Didim, Turkey
| | - Esin Beyan
- Department of Internal Medicine, Keçiören Training and Research Hospital, Ankara, Turkey
| | - Selma Uysal Ramadan
- Deparment of Radiology, Keçiören Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
16
|
Evans EB, Brady SW, Tripathi A, Hoffman-Kim D. Schwann cell durotaxis can be guided by physiologically relevant stiffness gradients. Biomater Res 2018; 22:14. [PMID: 29780613 PMCID: PMC5948700 DOI: 10.1186/s40824-018-0124-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 04/13/2018] [Indexed: 12/21/2022] Open
Abstract
Background Successful nerve regeneration depends upon directed migration of morphologically specialized repair state Schwann cells across a nerve defect. Although several groups have studied directed migration of Schwann cells in response to chemical or topographic cues, the current understanding of how the mechanical environment influences migration remains largely understudied and incomplete. Therefore, the focus of this study was to evaluate Schwann cell migration and morphodynamics in the presence of stiffness gradients, which revealed that Schwann cells can follow extracellular gradients of increasing stiffness, in a form of directed migration termed durotaxis. Methods Polyacrylamide substrates were fabricated to mimic the range of stiffness found in peripheral nerve tissue. We assessed Schwann cell response to substrates that were either mechanically uniform or embedded with a shallow or steep stiffness gradient, respectively corresponding to the mechanical niche present during either the fluid phase or subsequent matrix phase of the peripheral nerve regeneration process. We examined cell migration (velocity and directionality) and morphology (elongation, spread area, nuclear aspect ratio, and cell process dynamics). We also characterized the surface morphology of Schwann cells by scanning electron microscopy. Results On laminin-coated polyacrylamide substrates embedded with either a shallow (∼0.04 kPa/mm) or steep (∼0.95 kPa/mm) stiffness gradient, Schwann cells displayed durotaxis, increasing both their speed and directionality along the gradient materials, fabricated with elastic moduli in the range found in peripheral nerve tissue. Uniquely and unlike cell behavior reported in other cell types, the durotactic response of Schwann cells was not dependent upon the slope of the gradient. When we examined whether durotaxis behavior was accompanied by a pro-regenerative Schwann cell phenotype, we observed altered cell morphology, including increases in spread area and the number, elongation, and branching of the cellular processes, on the steep but not the shallow gradient materials. This phenotype emerged within hours of the cells adhering to the materials and was sustained throughout the 24 hour duration of the experiment. Control experiments also showed that unlike most adherent cells, Schwann cells did not alter their morphology in response to uniform substrates of different stiffnesses. Conclusion This study is notable in its report of durotaxis of cells in response to a stiffness gradient slope, which is greater than an order of magnitude less than reported elsewhere in the literature, suggesting Schwann cells are highly sensitive detectors of mechanical heterogeneity. Altogether, this work identifies durotaxis as a new migratory modality in Schwann cells, and further shows that the presence of a steep stiffness gradient can support a pro-regenerative cell morphology. Electronic supplementary material The online version of this article (10.1186/s40824-018-0124-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elisabeth B Evans
- 1Department of Molecular Pharmacology, Physiology, Brown University, Providence, Rhode Island, 02912 USA
| | - Samantha W Brady
- 1Department of Molecular Pharmacology, Physiology, Brown University, Providence, Rhode Island, 02912 USA
| | - Anubhav Tripathi
- 1Department of Molecular Pharmacology, Physiology, Brown University, Providence, Rhode Island, 02912 USA.,2Center for Biomedical Engineering, Brown University, Providence, Rhode Island, 02912 USA
| | - Diane Hoffman-Kim
- 1Department of Molecular Pharmacology, Physiology, Brown University, Providence, Rhode Island, 02912 USA.,2Center for Biomedical Engineering, Brown University, Providence, Rhode Island, 02912 USA.,3Carney Institute for Brain Science, Brown University, Providence, Rhode Island, 02912 USA.,4Center to Advance Predictive Biology, Brown University, Providence, Rhode Island, 02912 USA
| |
Collapse
|
17
|
Sarker M, Izadifar M, Schreyer D, Chen X. Influence of ionic crosslinkers (Ca2+/Ba2+/Zn2+) on the mechanical and biological properties of 3D Bioplotted Hydrogel Scaffolds. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2018; 29:1126-1154. [DOI: 10.1080/09205063.2018.1433420] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Md. Sarker
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
| | - Mohammad Izadifar
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
- Department of Surgery, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - David Schreyer
- Department of Anatomy and Cell Biology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
18
|
Tanaka A, Fujii Y, Kasai N, Okajima T, Nakashima H. Regulation of neuritogenesis in hippocampal neurons using stiffness of extracellular microenvironment. PLoS One 2018; 13:e0191928. [PMID: 29408940 PMCID: PMC5800654 DOI: 10.1371/journal.pone.0191928] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/15/2018] [Indexed: 11/19/2022] Open
Abstract
The mechanosensitivity of neurons in the central nervous system (CNS) is an interesting issue as regards understanding neuronal development and designing compliant materials as neural interfaces between neurons and external devices for treating CNS injuries and disorders. Although neurite initiation from a cell body is known to be the first step towards forming a functional nervous network during development or regeneration, less is known about how the mechanical properties of the extracellular microenvironment affect neuritogenesis. Here, we investigated the filamentous actin (F-actin) cytoskeletal structures of neurons, which are a key factor in neuritogenesis, on gel substrates with a stiffness-controlled substrate, to reveal the relationship between substrate stiffness and neuritogenesis. We found that neuritogenesis was significantly suppressed on a gel substrate with an elastic modulus higher than the stiffness of in vivo brain. Fluorescent images of the F-actin cytoskeletal structures showed that the F-actin organization depended on the substrate stiffness. Circumferential actin meshworks and arcs were formed at the edge of the cell body on the stiff gel substrates unlike with soft substrates. The suppression of F-actin cytoskeleton formation improved neuritogenesis. The results indicate that the organization of neuronal F-actin cytoskeletons is strongly regulated by the mechanical properties of the surrounding environment, and the mechanically-induced F-actin cytoskeletons regulate neuritogenesis.
Collapse
Affiliation(s)
- Aya Tanaka
- NTT Basic Research Laboratories NTT Corporation, Atsugi, Kanagawa, Japan
- * E-mail:
| | - Yuki Fujii
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Nahoko Kasai
- NTT Basic Research Laboratories NTT Corporation, Atsugi, Kanagawa, Japan
| | - Takaharu Okajima
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hiroshi Nakashima
- NTT Basic Research Laboratories NTT Corporation, Atsugi, Kanagawa, Japan
| |
Collapse
|
19
|
Palazzolo G, Moroni M, Soloperto A, Aletti G, Naldi G, Vassalli M, Nieus T, Difato F. Fast wide-volume functional imaging of engineered in vitro brain tissues. Sci Rep 2017; 7:8499. [PMID: 28819205 PMCID: PMC5561227 DOI: 10.1038/s41598-017-08979-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022] Open
Abstract
The need for in vitro models that mimic the human brain to replace animal testing and allow high-throughput screening has driven scientists to develop new tools that reproduce tissue-like features on a chip. Three-dimensional (3D) in vitro cultures are emerging as an unmatched platform that preserves the complexity of cell-to-cell connections within a tissue, improves cell survival, and boosts neuronal differentiation. In this context, new and flexible imaging approaches are required to monitor the functional states of 3D networks. Herein, we propose an experimental model based on 3D neuronal networks in an alginate hydrogel, a tunable wide-volume imaging approach, and an efficient denoising algorithm to resolve, down to single cell resolution, the 3D activity of hundreds of neurons expressing the calcium sensor GCaMP6s. Furthermore, we implemented a 3D co-culture system mimicking the contiguous interfaces of distinct brain tissues such as the cortical-hippocampal interface. The analysis of the network activity of single and layered neuronal co-cultures revealed cell-type-specific activities and an organization of neuronal subpopulations that changed in the two culture configurations. Overall, our experimental platform represents a simple, powerful and cost-effective platform for developing and monitoring living 3D layered brain tissue on chip structures with high resolution and high throughput.
Collapse
Affiliation(s)
- G Palazzolo
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - M Moroni
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.,Center for Neuroscience and Cognitive Systems @UniTn, Istituto Italiano di Tecnologia, Rovereto, Italy.,Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - A Soloperto
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - G Aletti
- Dipartimento di Matematica, Università degli studi di Milano, Milano, Italy
| | - G Naldi
- Dipartimento di Matematica, Università degli studi di Milano, Milano, Italy
| | - M Vassalli
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy
| | - T Nieus
- Department of Biomedical and Clinical Sciences "L. Sacco", Università degli Studi di Milano, Milano, Italy.
| | - F Difato
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.
| |
Collapse
|
20
|
GOLI-MALEKABADI ZAHRA, TAFAZZOLI-SHADPOUR MOHAMMAD, SEYEDJAFARI EHSAN. EFFECTS OF SUBSTRATE DEFORMABILITY ON CELL BEHAVIORS: ELASTIC MODULUS VERSUS THICKNESS. J MECH MED BIOL 2017. [DOI: 10.1142/s0219519417500889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The deformability of the substrate stimulating cell mechanotransduction depends not only on elastic modulus but also on the thickness. Polydimethylsiloxane (PDMS) which is widely used in microfluidic chips and platforms can be fabricated in a wide range of elastic modulus and thickness. In this study, we cultured human umbilical vein endothelial cells (HUVECs) on four groups of PDMS substrates of varying thickness and elastic modulus to examine effects of these parameters on morphology, viability and proliferation of cells. Both elastic modulus and thickness affected cell behavior. In general, the thickness of substrates had relatively higher impact on endothelial morphology than elastic modulus. Elongation of HUVECs on thick substrates was more intense compared to those on thin substrates. Both lowering thickness and reducing elastic modulus of PDMS decreased the viability of HUVECs, although thickness was more influential. Decrease in substrate thickness reduced cell proliferation regardless of substrate elastic modulus. In conclusion, our results suggest that endothelial behavior depends on substrate deformability, but cells react differently to the elastic modulus and thickness of PDMS by morphology, viability and growth. Results can improve the comprehension of cell mechanotransduction.
Collapse
Affiliation(s)
- ZAHRA GOLI-MALEKABADI
- Faculty of Biomedical Engineering, Amirkabir University of Technology, 424, Hafez Avenue, Tehran, Iran
| | | | - EHSAN SEYEDJAFARI
- Department of Biotechnology, University of Tehran, 13, Shafei alley, vesal Avenue, Tehran, Iran
| |
Collapse
|
21
|
Goli-Malekabadi Z, Tafazzoli-Shadpour M, Tamayol A, Seyedjafari E. Time dependency of morphological remodeling of endothelial cells in response to substrate stiffness. ACTA ACUST UNITED AC 2017; 7:41-47. [PMID: 28546952 PMCID: PMC5439388 DOI: 10.15171/bi.2017.06] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/15/2017] [Accepted: 02/08/2017] [Indexed: 01/06/2023]
Abstract
Introduction: Substrate stiffness regulates cellular behavior as cells experience different stiffness values of tissues in the body. For example, endothelial cells (ECs) covering the inner layer of blood vessels are exposed to different stiffness values due to various pathologic and physiologic conditions. Despite numerous studies, cells by time span sense mechanical properties of the substrate, but the response is not well understood. We hypothesized that time is a major determinant influencing the behavior of cells seeded on substrates of varying stiffness. Methods: We monitored cell spreading, internal structure, 3D topography, and the viability of ECs over 24 hours of culture on polydimethylsiloxane (PDMS) substrates with two different degrees of elastic modulus. Results: Despite significant differences in cell spreading after cell seeding, cells showed a similar shape and internal structure after 24 hours of culture on both soft and stiff substrates. However, 3D topographical images confirmed existence of rich lamellipodia and filopodia around the cells cultured on stiffer PDMS substrates. Conclusion: It was concluded that the response of ECs to the substrate stiffness was time dependent with initial enhanced cellular spreading and viability on stiffer substrates. Results can provide a better comprehension of cell mechanotransduction for tissue engineering applications.
Collapse
Affiliation(s)
| | | | - Ali Tamayol
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
22
|
Wilson CL, Hayward SL, Kidambi S. Astrogliosis in a dish: substrate stiffness induces astrogliosis in primary rat astrocytes. RSC Adv 2016; 6:34447-34457. [PMID: 32742641 PMCID: PMC7394306 DOI: 10.1039/c5ra25916a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Astrogliosis due to brain injury or disease can lead to varying molecular and morphological changes in astrocytes. Magnetic resonance elastography and ultrasound have demonstrated that brain stiffness varies with age and disease state. However, there is a lack in understanding the role of varied stiffness on the progression of astrogliosis highlighting a critical need to engineer in vitro models that mimic disease stages. Such models need to incorporate the dynamic changes in the brain microenvironment including the stiffness changes. In this study we developed a polydimethyl siloxane (PDMS) based platform that modeled the physiologically relevant stiffness of brain in both a healthy (200 Pa) and diseased (8000 Pa) state to investigate the effect of stiffness on astrocyte function. We observed that astrocytes grown on soft substrates displayed a consistently more quiescent phenotype while those on stiff substrates displayed an astrogliosis-like morphology. In addition to morphological changes, astrocytes cultured on stiff substrates demonstrated significant increase in other astrogliosis hallmarks - cellular proliferation and glial fibrillary acidic protein (GFAP) protein expression. Furthermore, culturing astrocytes on a stiff surface resulted in increased reactive oxygen species (ROS) production, increased super oxide dismutase activity and decreased glutamate uptake. Our platform lends itself for study of potential therapeutic strategies for brain injury focusing on the intricate brain microenvironment-astrocytes signaling pathways.
Collapse
Affiliation(s)
- Christina L Wilson
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, 820 N 16 Street, 207 Othmer Hall, NE, 68588, USA
| | - Stephen L Hayward
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, 820 N 16 Street, 207 Othmer Hall, NE, 68588, USA
| | - Srivatsan Kidambi
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, 820 N 16 Street, 207 Othmer Hall, NE, 68588, USA
- Nebraska Center for Materials and Nanoscience, University of Nebraska-Lincoln, 855 N 16 St, Lincoln, NE, 68588, USA
- Nebraska Center for the Prevention of Obesity Diseases, University of Nebraska-Lincoln, 316C Leverton Hall, 1700 35 Street, NE, 68583, USA
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, 42nd and Emile Street, Omaha, NE, 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
23
|
Previtera ML, Sengupta A. Substrate Stiffness Regulates Proinflammatory Mediator Production through TLR4 Activity in Macrophages. PLoS One 2015; 10:e0145813. [PMID: 26710072 PMCID: PMC4692401 DOI: 10.1371/journal.pone.0145813] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 12/09/2015] [Indexed: 01/06/2023] Open
Abstract
Clinical data show that disease adversely affects tissue elasticity or stiffness. While macrophage activity plays a critical role in driving disease pathology, there are limited data available on the effects of tissue stiffness on macrophage activity. In this study, the effects of substrate stiffness on inflammatory mediator production by macrophages were investigated. Bone marrow-derived macrophages were grown on polyacrylamide gels that mimicked the stiffness of a variety of soft biological tissues. Overall, macrophages grown on soft substrates produced less proinflammatory mediators than macrophages grown on stiff substrates when the endotoxin LPS was added to media. In addition, the pathways involved in stiffness-regulated proinflammation were investigated. The TLR4 signaling pathway was examined by evaluating TLR4, p-NF-κB p65, MyD88, and p-IκBα expression as well as p-NF-κB p65 translocation. Expression and translocation of the various signaling molecules were higher in macrophages grown on stiff substrates than on soft substrates. Furthermore, TLR4 knockout experiments showed that TLR4 activity enhanced proinflammation on stiff substrates. In conclusion, these results suggest that proinflammatory mediator production initiated by TLR4 is mechanically regulated in macrophages.
Collapse
Affiliation(s)
- Michelle L. Previtera
- JFK Neuroscience Institute, JFK Medical Center, 65 James Street, Edison, New Jersey, 08820, United States of America
- Department of Neuroscience, Seton Hall University, 400 South Orange Avenue, South Orange, New Jersey, 07079, United States of America
| | - Amitabha Sengupta
- JFK Neuroscience Institute, JFK Medical Center, 65 James Street, Edison, New Jersey, 08820, United States of America
| |
Collapse
|
24
|
Ali S, Wall IB, Mason C, Pelling AE, Veraitch FS. The effect of Young's modulus on the neuronal differentiation of mouse embryonic stem cells. Acta Biomater 2015; 25:253-267. [PMID: 26159105 DOI: 10.1016/j.actbio.2015.07.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 05/27/2015] [Accepted: 07/05/2015] [Indexed: 12/15/2022]
Abstract
There is substantial evidence that cells produce a diverse response to changes in ECM stiffness depending on their identity. Our aim was to understand how stiffness impacts neuronal differentiation of embryonic stem cells (ESC's), and how this varies at three specific stages of the differentiation process. In this investigation, three effects of stiffness on cells were considered; attachment, expansion and phenotypic changes during differentiation. Stiffness was varied from 2 kPa to 18 kPa to finally 35 kPa. Attachment was found to decrease with increasing stiffness for both ESC's (with a 95% decrease on 35 kPa compared to 2 kPa) and neural precursors (with a 83% decrease on 35 kPa). The attachment of immature neurons was unaffected by stiffness. Expansion was independent of stiffness for all cell types, implying that the proliferation of cells during this differentiation process was independent of Young's modulus. Stiffness had no effect upon phenotypic changes during differentiation for mESC's and neural precursors. 2 kPa increased the proportion of cells that differentiated from immature into mature neurons. Taken together our findings imply that the impact of Young's modulus on attachment diminishes as neuronal cells become more mature. Conversely, the impact of Young's modulus on changes in phenotype increased as cells became more mature.
Collapse
|
25
|
Previtera ML, Firestein BL. Glutamate affects dendritic morphology of neurons grown on compliant substrates. Biotechnol Prog 2015; 31:1128-32. [PMID: 25827105 DOI: 10.1002/btpr.2085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 03/25/2015] [Indexed: 12/26/2022]
Abstract
Brain stiffness changes in response to injury or disease. As a secondary consequence, glutamate is released from neurons and astroglia. Two types of glutamate receptors, N-methyl-d-aspartate (NMDA) and α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, sense mechanotransduction, leading to downstream signaling in neurons. Recently, our group reported that these two receptors affect dendrite morphology in hippocampal neurons grown on compliant substrates. Blocking receptor activity has distinct effects on dendrites, depending on whether neurons are grown on soft or stiff gels. In the current study, we examine whether exposure to glutamate itself alters stiffness-mediated changes to dendrites in hippocampal neurons. We find that glutamate augments changes seen when neurons are grown on soft gels of 300 or 600 Pa, but in contrast, glutamate attenuates changes seen when neurons are grown on stiff gels of 3,000 Pa. These results suggest that there is interplay between mechanosensing and glutamate receptor activation in determining dendrite morphology in neurons.
Collapse
Affiliation(s)
- Michelle L Previtera
- Graduate Program in Molecular Biosciences, the State University of New Jersey, Piscataway, NJ, 08854-8082.,Dept. of Cell Biology and Neuroscience, the State University of New Jersey, Piscataway, NJ, 08854-8082
| | - Bonnie L Firestein
- Dept. of Cell Biology and Neuroscience, the State University of New Jersey, Piscataway, NJ, 08854-8082.,Dept. of Biomedical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ, 08854-8082
| |
Collapse
|
26
|
Marino A, Filippeschi C, Mattoli V, Mazzolai B, Ciofani G. Biomimicry at the nanoscale: current research and perspectives of two-photon polymerization. NANOSCALE 2015; 7:2841-50. [PMID: 25519056 DOI: 10.1039/c4nr06500j] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Living systems such as cells and tissues are extremely sensitive to their surrounding physico-chemical microenvironment. In the field of regenerative medicine and tissue engineering, the maintenance of culture conditions suitable for the formation of proliferation niches, for the self-renewal maintenance of stem cells, or for the promotion of a particular differentiation fate is an important issue that has been addressed using different strategies. A number of investigations suggests that a particular cell behavior can be in vitro resembled by mimicking the corresponding in vivo conditions. In this context, several biomimetic environments have been designed in order to control cell phenotypes and functions. In this review, we will analyze the most recent examples of the control of the in vitro physical micro/nano-environment by exploiting an innovative technique of high resolution 3D photolithography, the two-photon polymerization (2pp). The biomedical applications of this versatile and disruptive computer assisted design/manufacturing technology are very wide, and range from the fabrication of biomimetic and nanostructured scaffolds for tissue engineering and regenerative medicine, to the microfabrication of biomedical devices, like ossicular replacement prosthesis and microneedles.
Collapse
Affiliation(s)
- Attilio Marino
- Istituto Italiano di Tecnologia, Center for Micro-BioRobotics @SSSA, Viale Rinaldo Piaggio 34, 56025 Pontedera, Italy.
| | | | | | | | | |
Collapse
|
27
|
Béduer A, Braschler T, Peric O, Fantner GE, Mosser S, Fraering PC, Benchérif S, Mooney DJ, Renaud P. A compressible scaffold for minimally invasive delivery of large intact neuronal networks. Adv Healthc Mater 2015; 4:301-12. [PMID: 25178838 DOI: 10.1002/adhm.201400250] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/31/2014] [Indexed: 12/21/2022]
Abstract
Millimeter to centimeter-sized injectable neural scaffolds based on macroporous cryogels are presented. The polymer-scaffolds are made from alginate and carboxymethyl-cellulose by a novel simple one-pot cryosynthesis. They allow surgical sterility by means of autoclaving, and present native laminin as an attachment motive for neural adhesion and neurite development. They are designed to protect an extended, living neuronal network during compression to a small fraction of the original volume in order to enable minimally invasive delivery. The scaffolds behave as a mechanical meta-material: they are soft at the macroscopic scale, enabling injection through narrow-bore tubing and potentially good cellular scaffold integration in soft target tissues such as the brain. At the same time, the scaffold material has a high local Young modulus, allowing protection of the neuronal network during injection. Based on macroscopic and nanomechanical characterization, the generic geometrical and mechanical design rules are presented, enabling macroporous cellular scaffold injectability.
Collapse
Affiliation(s)
- Amélie Béduer
- STI-IMT-LMIS4, Station 17, EPFL; 1015 Lausanne Switzerland
| | - Thomas Braschler
- STI-IMT-LMIS4, Station 17, EPFL; 1015 Lausanne Switzerland
- School of Engineering and Applied Sciences; Harvard University; 02138 Cambridge MA USA
| | - Oliver Peric
- STI-IBI-LBNI, Station 17, EPFL; 1015 Lausanne Switzerland
| | | | | | | | - Sidi Benchérif
- School of Engineering and Applied Sciences; Harvard University; 02138 Cambridge MA USA
| | - David J. Mooney
- School of Engineering and Applied Sciences; Harvard University; 02138 Cambridge MA USA
| | | |
Collapse
|
28
|
Previtera ML, Peterman K, Shah S, Luzuriaga J. Lipid rafts direct macrophage motility in the tissue microenvironment. Ann Biomed Eng 2014; 43:896-905. [PMID: 25269613 DOI: 10.1007/s10439-014-1142-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 09/23/2014] [Indexed: 01/14/2023]
Abstract
Infiltrating leukocytes are exposed to a wide range of tissue elasticities. While we know the effects of substrate elasticity on acute inflammation via the study of neutrophil migration, we do not know its effects on leukocytes that direct chronic inflammatory events. Here, we studied morphology and motility of macrophages, the innate immune cells that orchestrate acute and chronic inflammation, on polyacrylamide hydrogels that mimicked a wide range of tissue elasticities. As expected, we found that macrophage spreading area increased as substrate elasticity increased. Unexpectedly, we found that morphology did not inversely correlate with motility. In fact, velocity of steady-state macrophages remained unaffected by substrate elasticity, while velocity of biologically stimulated macrophages was limited on stiff substrates. We also found that the lack of motility on stiff substrates was due to a lack of lipid rafts on the leading edge of the macrophages. This study implicates lipid rafts in the mechanosensory mechanism of innate immune cell infiltration.
Collapse
Affiliation(s)
- Michelle L Previtera
- JFK Neuroscience Institute, JFK Medical Center, 65 James Street, Edison, NJ, 08820, USA,
| | | | | | | |
Collapse
|
29
|
Previtera ML, Langrana NA. Preparation of DNA-crosslinked polyacrylamide hydrogels. J Vis Exp 2014. [PMID: 25226067 DOI: 10.3791/51323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Mechanobiology is an emerging scientific area that addresses the critical role of physical cues in directing cell morphology and function. For example, the effect of tissue elasticity on cell function is a major area of mechanobiology research because tissue stiffness modulates with disease, development, and injury. Static tissue-mimicking materials, or materials that cannot alter stiffness once cells are plated, are predominately used to investigate the effects of tissue stiffness on cell functions. While information gathered from static studies is valuable, these studies are not indicative of the dynamic nature of the cellular microenvironment in vivo. To better address the effects of dynamic stiffness on cell function, we developed a DNA-crosslinked polyacrylamide hydrogel system (DNA gels). Unlike other dynamic substrates, DNA gels have the ability to decrease or increase in stiffness after fabrication without stimuli. DNA gels consist of DNA crosslinks that are polymerized into a polyacrylamide backbone. Adding and removing crosslinks via delivery of single-stranded DNA allows temporal, spatial, and reversible control of gel elasticity. We have shown in previous reports that dynamic modulation of DNA gel elasticity influences fibroblast and neuron behavior. In this report and video, we provide a schematic that describes the DNA gel crosslinking mechanisms and step-by-step instructions on the preparation DNA gels.
Collapse
Affiliation(s)
| | - Noshir A Langrana
- Department of Biomedical Engineering, Rutgers University; Department of Mechanical and Aerospace Engineering, Rutgers University
| |
Collapse
|
30
|
|
31
|
Sur S, Guler MO, Webber MJ, Pashuck ET, Ito M, Stupp SI, Launey T. Synergistic regulation of cerebellar Purkinje neuron development by laminin epitopes and collagen on an artificial hybrid matrix construct. Biomater Sci 2014; 2:903-914. [PMID: 25530849 PMCID: PMC4269166 DOI: 10.1039/c3bm60228a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The extracellular matrix (ECM) creates a dynamic environment around the cells in the developing central nervous system, providing them with the necessary biochemical and biophysical signals. Although the functions of many ECM molecules in neuronal development have been individually studied in detail, the combinatorial effects of multiple ECM components are not well characterized. Here we demonstrate that the expression of collagen and laminin-1 (lam-1) are spatially and temporally correlated during embryonic and post-natal development of the cerebellum. These changes in ECM distribution correspond to specific stages of Purkinje neuron (PC) migration, somatic monolayer formation and polarization. To clarify the respective roles of these ECM molecules on PC development, we cultured cerebellar neurons on a hybrid matrix comprised of collagen and a synthetic peptide amphiphile nanofiber bearing a potent lam-1 derived bioactive IKVAV peptide epitope. By systematically varying the concentration and ratio of collagen and the laminin epitope in the matrix, we could demonstrate a synergistic relationship between these two ECM components in controlling multiple aspects of PC maturation. An optimal ratio of collagen and IKVAV in the matrix was found to promote maximal PC survival and dendrite growth, while dendrite penetration into the matrix was enhanced by a high IKVAV to collagen ratio. In addition, the laminin epitope was found to guide PC axon development. By combining our observations in vivo and in vitro, we propose a model of PC development where the synergistic effects of collagen and lam-1 play a key role in migration, polarization and morphological maturation of PCs.
Collapse
Affiliation(s)
- Shantanu Sur
- Laboratory for Memory and Learning, RIKEN Brain Science Institute, Wako-shi, 351-0198 Saitama, Japan
- School of Medical Science and Technology, IIT Kharagpur, 721302, India
- The Institute for Bionanotechnology in Medicine (IBNAM), Northwestern University, Chicago, IL 60611, USA
| | - Mustafa O. Guler
- The Institute for Bionanotechnology in Medicine (IBNAM), Northwestern University, Chicago, IL 60611, USA
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, Ankara, 06800, Turkey
| | - Matthew J. Webber
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Eugene T. Pashuck
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Masao Ito
- Laboratory for Memory and Learning, RIKEN Brain Science Institute, Wako-shi, 351-0198 Saitama, Japan
| | - Samuel I. Stupp
- The Institute for Bionanotechnology in Medicine (IBNAM), Northwestern University, Chicago, IL 60611, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Thomas Launey
- Laboratory for Memory and Learning, RIKEN Brain Science Institute, Wako-shi, 351-0198 Saitama, Japan
- Launey Research Unit for Molecular Neurocybernetics, RIKEN Brain Science Institute, Wako-shi, 351-0198 Saitama, Japan
| |
Collapse
|
32
|
da Silva CA, de Morais ECP, Costa MDM, Ribas JLC, Guiloski IC, Ramsdorf WA, Zanata SM, Cestari MM, Ribeiro CAO, Magalhães VF, Trudeau VL, de Assis HCS. Saxitoxins induce cytotoxicity, genotoxicity and oxidative stress in teleost neurons in vitro. Toxicon 2014; 86:8-15. [PMID: 24813331 DOI: 10.1016/j.toxicon.2014.04.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 04/25/2014] [Accepted: 04/29/2014] [Indexed: 11/16/2022]
Abstract
The aim of this study was establish a protocol for isolation and primary culture of neurons from tropical freshwater fish species Hoplias malabaricus for assessment of the effects of neurotoxic substances as saxitoxins (STXs). Cells from brain of H. malabaricus were treated with different concentrations of trypsin, dispase and papain for tissue dissociation. Cells type was separated by cellular gradient and basic fibroblast growth factor (bFGF) supplement nutrition media were added. The dissociated cells were plated with medium and different STXs concentrations and the toxic cellular effects such as oxidative stress, neurotoxicity, and genotoxicity and apoptosis process were evaluated. Cultures treated with bFGF showed the greatest adherence, survival and cellular development. STXs increased specific activity of glutathione peroxidase and lipoperoxidation levels, were cytotoxic and genotoxic indicated by the comet assay. Although the STXs effects due the blockage of sodium channels is reported to be reversible, the time exposure and concentration of STXs suggested cellular injuries which can lead to neuropathology. The establishment of primary neuronal culture protocol enables new applications for neurotoxicological assessments.
Collapse
Affiliation(s)
- Cesar Aparecido da Silva
- Ecology and Conservation Post-graduate Program, Federal University of Paraná, 81531-990, P.O. Box 19031, Curitiba, Paraná, Brazil
| | | | - Michele Dietrich Moura Costa
- Department of Basic Pathology, Federal University of Paraná, 81531-990, P.O. Box 19031, Curitiba, Paraná, Brazil
| | - João Luiz Coelho Ribas
- Department of Pharmacology, Federal University of Paraná, 81531-990, P.O. Box 19031, Curitiba, Paraná, Brazil
| | - Izonete Cristina Guiloski
- Department of Pharmacology, Federal University of Paraná, 81531-990, P.O. Box 19031, Curitiba, Paraná, Brazil
| | - Wanessa A Ramsdorf
- Department of Genetics, Federal University of Paraná, 81531-990, P.O. Box 19031, Curitiba, Paraná, Brazil
| | - Silvio Marques Zanata
- Department of Basic Pathology, Federal University of Paraná, 81531-990, P.O. Box 19031, Curitiba, Paraná, Brazil
| | - Marta M Cestari
- Department of Genetics, Federal University of Paraná, 81531-990, P.O. Box 19031, Curitiba, Paraná, Brazil
| | | | - Valéria F Magalhães
- Federal University of Rio de Janeiro, Institute of Biophysics Carlos Chagas Filho, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Vance L Trudeau
- Department of Biology, University of Ottawa, K1N 6N5 Ottawa, Ontario, Canada
| | - Helena C Silva de Assis
- Department of Pharmacology, Federal University of Paraná, 81531-990, P.O. Box 19031, Curitiba, Paraná, Brazil.
| |
Collapse
|
33
|
Matyash M, Despang F, Ikonomidou C, Gelinsky M. Swelling and Mechanical Properties of Alginate Hydrogels with Respect to Promotion of Neural Growth. Tissue Eng Part C Methods 2014; 20:401-11. [DOI: 10.1089/ten.tec.2013.0252] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Marina Matyash
- Center for Translational Bone, Joint and Soft Tissue Research, University Hospital and Medical Faculty, Technische Universität Dresden, Dresden, Germany
- Cellular Neuroscience, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Florian Despang
- Center for Translational Bone, Joint and Soft Tissue Research, University Hospital and Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | | | - Michael Gelinsky
- Center for Translational Bone, Joint and Soft Tissue Research, University Hospital and Medical Faculty, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
34
|
Immobilized laminin concentration gradients on electrospun fiber scaffolds for controlled neurite outgrowth. Biointerphases 2014; 9:011003. [DOI: 10.1116/1.4857295] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
35
|
Chen WH, Cheng SJ, Tzen JTC, Cheng CM, Lin YW. Probing relevant molecules in modulating the neurite outgrowth of hippocampal neurons on substrates of different stiffness. PLoS One 2013; 8:e83394. [PMID: 24386192 PMCID: PMC3875460 DOI: 10.1371/journal.pone.0083394] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 11/04/2013] [Indexed: 11/19/2022] Open
Abstract
Hippocampal neurons play a critical role in learning and memory; however, the effects of environmental mechanical forces on neurite extension and associated underlying mechanisms are largely unexplored, possibly due to difficulties in maintaining central nervous system neurons. Neuron adhesion, neurite length, and mechanotransduction are mainly influenced by the extracellular matrix (ECM), which is often associated with structural scaffolding. In this study, we investigated the relationship between substrate stiffness and hippocampal neurite outgrowth by controlling the ratios of polydimethylsiloxane (PDMS) base to curing agent to create substrates of varying stiffness. Immunostaining results demonstrated that hippocampal neurons have longer neurite elongation in 35:1 PDMS substrate compared those grown on 15:1 PDMS, indicating that soft substrates provide a more optimal stiffness for hippocampal neurons. Additionally, we discovered that pPKCα expression was higher in the 15:1 and 35:1 PDMS groups than in the poly-L-lysine-coated glass group. However, when we used a fibronectin (FN) coating, we found that pFAKy397 and pFAKy925 expression were higher in glass group than in the 15:1 or 35: 1 PDMS groups, but pPKCα and pERK1/2 expression were higher in the 35:1 PDMS group than in the glass group. These results support the hypothesis that environmental stiffness influences hippocampal neurite outgrowth and underlying signaling pathways.
Collapse
Affiliation(s)
- Wei-Hsin Chen
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Sin-Jhong Cheng
- Department of Life Science and Institute of Zoology, National Taiwan University, Taipei, Taiwan
| | - Jason T. C. Tzen
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Chao-Min Cheng
- Institute of Nanoengineering and Microsystems, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Wen Lin
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan
- Acupuncture Research Center, China Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
36
|
Biffi E, Regalia G, Menegon A, Ferrigno G, Pedrocchi A. The influence of neuronal density and maturation on network activity of hippocampal cell cultures: a methodological study. PLoS One 2013; 8:e83899. [PMID: 24386305 PMCID: PMC3873984 DOI: 10.1371/journal.pone.0083899] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 11/09/2013] [Indexed: 11/25/2022] Open
Abstract
It is known that cell density influences the maturation process of in vitro neuronal networks. Neuronal cultures plated with different cell densities differ in number of synapses per neuron and thus in single neuron synaptic transmission, which results in a density-dependent neuronal network activity. Although many authors provided detailed information about the effects of cell density on neuronal culture activity, a dedicated report of density and age influence on neuronal hippocampal culture activity has not yet been reported. Therefore, this work aims at providing reference data to researchers that set up an experimental study on hippocampal neuronal cultures, helping in planning and decoding the experiments. In this work, we analysed the effects of both neuronal density and culture age on functional attributes of maturing hippocampal cultures. We characterized the electrophysiological activity of neuronal cultures seeded at three different cell densities, recording their spontaneous electrical activity over maturation by means of MicroElectrode Arrays (MEAs). We had gather data from 86 independent hippocampal cultures to achieve solid statistic results, considering the high culture-to-culture variability. Network activity was evaluated in terms of simple spiking, burst and network burst features. We observed that electrical descriptors were characterized by a functional peak during maturation, followed by a stable phase (for sparse and medium density cultures) or by a decrease phase (for high dense neuronal cultures). Moreover, 900 cells/mm2 cultures showed characteristics suitable for long lasting experiments (e.g. chronic effect of drug treatments) while 1800 cells/mm2 cultures should be preferred for experiments that require intense electrical activity (e.g. to evaluate the effect of inhibitory molecules). Finally, cell cultures at 3600 cells/mm2 are more appropriate for experiments in which time saving is relevant (e.g. drug screenings). These results are intended to be a reference for the planning of in vitro neurophysiological and neuropharmacological experiments with MEAs.
Collapse
Affiliation(s)
- Emilia Biffi
- Neuroengineering and Medical Robotics Laboratory, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
- Advanced Light and Electron Microscopy Bio-Imaging Centre, Experimental Imaging Centre, San Raffaele Scientific Institute, Milan, Italy
- * E-mail:
| | - Giulia Regalia
- Neuroengineering and Medical Robotics Laboratory, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Andrea Menegon
- Advanced Light and Electron Microscopy Bio-Imaging Centre, Experimental Imaging Centre, San Raffaele Scientific Institute, Milan, Italy
| | - Giancarlo Ferrigno
- Neuroengineering and Medical Robotics Laboratory, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Alessandra Pedrocchi
- Neuroengineering and Medical Robotics Laboratory, Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
37
|
Previtera ML, Hui M, Verma D, Shahin AJ, Schloss R, Langrana NA. The effects of substrate elastic modulus on neural precursor cell behavior. Ann Biomed Eng 2013; 41:1193-207. [PMID: 23429962 DOI: 10.1007/s10439-013-0765-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 02/13/2013] [Indexed: 01/14/2023]
Abstract
The spinal cord has a limited capacity to self-repair. After injury, endogenous stem cells are activated and migrate, proliferate, and differentiate into glial cells. The absence of neuronal differentiation has been partly attributed to the interaction between the injured microenvironment and neural stem cells. In order to improve post-injury neuronal differentiation and/or maturation potential, cell-cell and cell-biochemical interactions have been investigated. However, little is known about the role of stem cell-matrix interactions on stem cell-mediated repair. Here, we specifically examined the effects of matrix elasticity on stem cell-mediated repair in the spinal cord, since spinal cord injury results in drastic changes in parenchyma elasticity and viscosity. Spinal cord-derived neural precursor cells (NPCs) were grown on bis-acrylamide substrates with various rigidities. NPC growth, proliferation, and differentiation were examined and optimal in the range of normal spinal cord elasticity. In conclusion, limitations in NPC growth, proliferation, and neuronal differentiation were encountered when substrate elasticity was not within normal spinal cord tissue elasticity ranges. These studies elucidate the effect injury mediated mechanical changes may have on tissue repair by stem cells. Furthermore, this information can be applied to the development of future neuroregenerative biomaterials for spinal cord repair.
Collapse
Affiliation(s)
- Michelle L Previtera
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA
| | | | | | | | | | | |
Collapse
|
38
|
Millet LJ, Gillette MU. New perspectives on neuronal development via microfluidic environments. Trends Neurosci 2012; 35:752-61. [PMID: 23031246 DOI: 10.1016/j.tins.2012.09.001] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 08/18/2012] [Accepted: 09/06/2012] [Indexed: 11/28/2022]
Abstract
Understanding the signals that guide neuronal development and direct formation of axons, dendrites, and synapses during wiring of the brain is a fundamental challenge in developmental neuroscience. Discovery of how local signals shape developing neurons has been impeded by the inability of conventional culture methods to interrogate microenvironments of complex neuronal cytoarchitectures, where different subdomains encounter distinct chemical, physical, and fluidic features. Microfabrication techniques are facilitating the creation of microenvironments tailored to neuronal structures and subdomains with unprecedented access and control. The design, fabrication, and properties of microfluidic devices offer significant advantages for addressing unresolved issues of neuronal development. These high-resolution approaches are poised to contribute new insights into mechanisms for restoring neuronal function and connectivity compromised by injury, stress, and neurodegeneration.
Collapse
Affiliation(s)
- Larry J Millet
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | |
Collapse
|
39
|
Polyelectrolyte Complex Membranes for Prevention of Post-Surgical Adhesions in Neurosurgery. Ann Biomed Eng 2012; 40:1949-60. [DOI: 10.1007/s10439-012-0564-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 03/30/2012] [Indexed: 10/28/2022]
|
40
|
Ciofani G, Genchi GG, Mattoli V. ZnO nanowire arrays as substrates for cell proliferation and differentiation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2012. [DOI: 10.1016/j.msec.2011.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
41
|
Fibroblast Morphology on Dynamic Softening of Hydrogels. Ann Biomed Eng 2011; 40:1061-72. [DOI: 10.1007/s10439-011-0483-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 11/28/2011] [Indexed: 11/26/2022]
|
42
|
Previtera ML, Langhammer CG, Langrana NA, Firestein BL. Regulation of dendrite arborization by substrate stiffness is mediated by glutamate receptors. Ann Biomed Eng 2010; 38:3733-43. [PMID: 20614247 DOI: 10.1007/s10439-010-0112-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2010] [Accepted: 06/21/2010] [Indexed: 10/19/2022]
Abstract
Brain injury or disease can initiate changes in local or global stiffness of brain tissue. While stiffness of the extracellular environment is known to affect the morphology and function of many cell types, little is known about how the dendrites of neurons respond to changes in brain stiffness. To assess how extracellular stiffness affects dendrite morphology, we took biomaterial and biomedical engineering approaches. We cultured mixed and pure hippocampal neurons on hydrogels composed of polyacrylamide (PA) of varying stiffnesses to mimic the effects of extracellular matrix stiffness on dendrite morphology. The majority of investigations of cortical and spinal cord neurons on soft hydrogels examined branching at early time points (days in vitro (DIV) 2-7), an important distinction from our study, where we include later time points that encompass the peak of branching (DIV 10-12). At DIV 12, dendrite branching was altered by stiffness for both pure and mixed neuronal cultures. Furthermore, we treated hippocampal cultures with glutamate receptor antagonists and with astrocyte-conditioned media. Blocking AMPA and NMDA receptors affected the changes in dendrite branching seen at varying rigidities. Moreover, extracellular factors secreted by astrocytes also change dendrite branching seen at varying rigidities. Thus, astrocytes and ionotropic glutamate receptors contribute to mechanosensing.
Collapse
Affiliation(s)
- Michelle L Previtera
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | | | | | | |
Collapse
|