1
|
Li C, Meng X, Li S, Wang C. Therapeutic Advances in Peripheral Nerve Injuries: Nerve-Guided Conduit and Beyond. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40195945 DOI: 10.1089/ten.teb.2024.0322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Peripheral nerve injury (PNI), a challenging neurosurgery issue, often leads to partial or complete loss of neuronal functions and even neuropathic pain. Thus far, the gold standard for treating peripheral nerve deficit remains autografts. While numerous reviews have explored PNI and regeneration, this work distinctively synthesizes recent advancements in tissue engineering-particularly four-dimensional (4D) bioprinting and exosome therapies-with an emphasis on their clinical translation. By consolidating findings spanning molecular mechanisms to therapeutic applications, this review proposes an actionable framework for advancing experimental strategies toward clinically viable solutions. Our work critically evaluates emerging innovations such as dynamically adaptive 4D-printed nerve conduits and exosome-based therapies, underscoring their potential to match conventional autografts in achieving functional restoration. Impact Statement Although several previous reviews have been made on describing with great detail the degenerative and regenerative mechanisms of the peripheral nervous systems, as well as the several existing and exploratory treatment strategies, we focus more on the latest advancements of each of those topics.
Collapse
Affiliation(s)
- Changqing Li
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xianyu Meng
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shengji Li
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjing Wang
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
2
|
Deleye L, Franchi F, Trevisani M, Loiacono F, Vercellino S, Debellis D, Liessi N, Armirotti A, Vázquez E, Valente P, Castagnola V, Benfenati F. Few-layered graphene increases the response of nociceptive neurons to irritant stimuli. NANOSCALE 2024; 16:2419-2431. [PMID: 38226500 DOI: 10.1039/d3nr03790h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The unique properties of few-layered graphene (FLG) make it interesting for a variety of applications, including biomedical applications, such as tissue engineering and drug delivery. Although different studies focus on applications in the central nervous system, its interaction with the peripheral nervous system has been so far overlooked. Here, we investigated the effects of exposure to colloidal dispersions of FLG on the sensory neurons of the rat dorsal root ganglia (DRG). We found that the FLG flakes were actively internalized by sensory neurons, accumulated in large intracellular vesicles, and possibly degraded over time, without major toxicological concerns, as neuronal viability, morphology, protein content, and basic electrical properties of DRG neurons were preserved. Interestingly, in our electrophysiological investigation under noxious stimuli, we observed an increased functional response upon FLG treatment of the nociceptive subpopulation of DRG neurons in response to irritants specific for chemoreceptors TRPV1 and TRPA1. The observed effects of FLG on DRG neurons may open-up novel opportunities for applications of these materials in specific disease models.
Collapse
Affiliation(s)
- Lieselot Deleye
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
| | - Francesca Franchi
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Martina Trevisani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- Department of Experimental Medicine, Section of Physiology, University of Genova, Genoa, 16132, Italy.
| | - Fabrizio Loiacono
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Silvia Vercellino
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Doriana Debellis
- Electron Microscopy Facility, IIT, Via Morego 30, 16163, Genoa, Italy
| | - Nara Liessi
- Analytical Chemistry Facility, IIT, via Morego, 30, 16163, Genoa, Italy
| | - Andrea Armirotti
- Analytical Chemistry Facility, IIT, via Morego, 30, 16163, Genoa, Italy
| | - Ester Vázquez
- Facultad de Ciencias Químicas, Universidad Castilla La-Mancha, Ciudad Real, 13071 Spain
| | - Pierluigi Valente
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
- Department of Experimental Medicine, Section of Physiology, University of Genova, Genoa, 16132, Italy.
| | - Valentina Castagnola
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
3
|
Piglionico SS, Varga B, Pall O, Romieu O, Gergely C, Cuisinier F, Levallois B, Panayotov IV. Biomechanical characterization of a fibrinogen-blood hydrogel for human dental pulp regeneration. Biomater Sci 2023; 11:6919-6930. [PMID: 37655620 DOI: 10.1039/d3bm00515a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
In dental practice, Regenerative Endodontic Treatment (RET) is applied as an alternative to classical endodontic treatments of immature necrotic teeth. This procedure, also known as dental pulp revitalization, relies on the formation of a blood clot inside the root canal leading to the formation of a reparative vascularized tissue similar to dental pulp, which would provide vitality to the affected tooth. Despite the benefit of this technique, it lacks reproducibility due to the fast degradation and poor mechanical properties of blood clots. This work presents a method for constructing a fibrinogen-blood hydrogel that mimics the viscoelastic properties of human dental pulp while preserving the biological properties of blood for application in RET. By varying the blood and fibrinogen concentrations, gels with different biomechanical and biological properties were obtained. Rheology and atomic force microscopy (AFM) were combined to study the viscoelastic properties. AFM was used to evaluate the elasticity of human dental pulp. The degradation and swelling rates were assessed by measuring weight changes. The biomimetic properties of the gels were demonstrated by studying the cell survival and proliferation of dental pulp cells (DPCs) for 14 days. The formation of an extracellular matrix (ECM) was assessed by multiphoton microscopy (MPM). The angiogenic potential was evaluated by an ex vivo aortic ring assay, in which the endothelial cells were observed by histological staining after migration. The results show that the Fbg-blood gel prepared with 9 mg ml-1 fibrinogen and 50% blood of the Fbg solution volume has similar elasticity to human dental pulp and adequate degradation and swelling rates. It also allows cell survival and ECM secretion and enhances endothelial cell migration and formation of neovessel-like structures.
Collapse
Affiliation(s)
- Sofia Silvia Piglionico
- LBN, Univ Montpellier, Montpellier, France.
- Centro de Investigaciones Odontológicas, National University of Cuyo, Argentina
| | - Bela Varga
- L2C, Univ Montpellier, CNRS, Montpellier, France.
| | | | | | | | | | | | | |
Collapse
|
4
|
Kong L, Gao X, Qian Y, Sun W, You Z, Fan C. Biomechanical microenvironment in peripheral nerve regeneration: from pathophysiological understanding to tissue engineering development. Am J Cancer Res 2022; 12:4993-5014. [PMID: 35836812 PMCID: PMC9274750 DOI: 10.7150/thno.74571] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/11/2022] [Indexed: 01/12/2023] Open
Abstract
Peripheral nerve injury (PNI) caused by trauma, chronic disease and other factors may lead to partial or complete loss of sensory, motor and autonomic functions, as well as neuropathic pain. Biological activities are always accompanied by mechanical stimulation, and biomechanical microenvironmental homeostasis plays a complicated role in tissue repair and regeneration. Recent studies have focused on the effects of biomechanical microenvironment on peripheral nervous system development and function maintenance, as well as neural regrowth following PNI. For example, biomechanical factors-induced cluster gene expression changes contribute to formation of peripheral nerve structure and maintenance of physiological function. In addition, extracellular matrix and cell responses to biomechanical microenvironment alterations after PNI directly trigger a series of cascades for the well-organized peripheral nerve regeneration (PNR) process, where cell adhesion molecules, cytoskeletons and mechanically gated ion channels serve as mechanosensitive units, mechanical effector including focal adhesion kinase (FAK) and yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) as mechanotransduction elements. With the rapid development of tissue engineering techniques, a substantial number of PNR strategies such as aligned nerve guidance conduits, three-dimensional topological designs and piezoelectric scaffolds emerge expected to improve the neural biomechanical microenvironment in case of PNI. These tissue engineering nerve grafts display optimized mechanical properties and outstanding mechanomodulatory effects, but a few bottlenecks restrict their application scenes. In this review, the current understanding in biomechanical microenvironment homeostasis associated with peripheral nerve function and PNR is integrated, where we proposed the importance of balances of mechanosensitive elements, cytoskeletal structures, mechanotransduction cascades, and extracellular matrix components; a wide variety of promising tissue engineering strategies based on biomechanical modulation are introduced with some suggestions and prospects for future directions.
Collapse
Affiliation(s)
- Lingchi Kong
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China.,Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xin Gao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Belt and Road Joint Laboratory of Advanced Fiber and Low-dimension Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, China
| | - Yun Qian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China.,Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.,✉ Corresponding authors: Yun Qian, E-mail: ; Wei Sun, E-mail: ; Zhengwei You, E-mail: ; Cunyi Fan, E-mail:
| | - Wei Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Belt and Road Joint Laboratory of Advanced Fiber and Low-dimension Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, China.,✉ Corresponding authors: Yun Qian, E-mail: ; Wei Sun, E-mail: ; Zhengwei You, E-mail: ; Cunyi Fan, E-mail:
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Belt and Road Joint Laboratory of Advanced Fiber and Low-dimension Materials, College of Materials Science and Engineering, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai, 201620, China.,✉ Corresponding authors: Yun Qian, E-mail: ; Wei Sun, E-mail: ; Zhengwei You, E-mail: ; Cunyi Fan, E-mail:
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China.,Youth Science and Technology Innovation Studio of Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.,✉ Corresponding authors: Yun Qian, E-mail: ; Wei Sun, E-mail: ; Zhengwei You, E-mail: ; Cunyi Fan, E-mail:
| |
Collapse
|
5
|
Procès A, Luciano M, Kalukula Y, Ris L, Gabriele S. Multiscale Mechanobiology in Brain Physiology and Diseases. Front Cell Dev Biol 2022; 10:823857. [PMID: 35419366 PMCID: PMC8996382 DOI: 10.3389/fcell.2022.823857] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/08/2022] [Indexed: 12/11/2022] Open
Abstract
Increasing evidence suggests that mechanics play a critical role in regulating brain function at different scales. Downstream integration of mechanical inputs into biochemical signals and genomic pathways causes observable and measurable effects on brain cell fate and can also lead to important pathological consequences. Despite recent advances, the mechanical forces that influence neuronal processes remain largely unexplored, and how endogenous mechanical forces are detected and transduced by brain cells into biochemical and genetic programs have received less attention. In this review, we described the composition of brain tissues and their pronounced microstructural heterogeneity. We discuss the individual role of neuronal and glial cell mechanics in brain homeostasis and diseases. We highlight how changes in the composition and mechanical properties of the extracellular matrix can modulate brain cell functions and describe key mechanisms of the mechanosensing process. We then consider the contribution of mechanobiology in the emergence of brain diseases by providing a critical review on traumatic brain injury, neurodegenerative diseases, and neuroblastoma. We show that a better understanding of the mechanobiology of brain tissues will require to manipulate the physico-chemical parameters of the cell microenvironment, and to develop three-dimensional models that can recapitulate the complexity and spatial diversity of brain tissues in a reproducible and predictable manner. Collectively, these emerging insights shed new light on the importance of mechanobiology and its implication in brain and nerve diseases.
Collapse
Affiliation(s)
- Anthony Procès
- Mechanobiology and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium.,Neurosciences Department, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Marine Luciano
- Mechanobiology and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Yohalie Kalukula
- Mechanobiology and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Laurence Ris
- Neurosciences Department, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Sylvain Gabriele
- Mechanobiology and Biomaterials group, Interfaces and Complex Fluids Laboratory, Research Institute for Biosciences, University of Mons, Mons, Belgium
| |
Collapse
|
6
|
Zhang H, Cai X, Xiang C, Han Y, Niu Q. miR-29a and the PTEN-GSK3β axis are involved in aluminum-induced damage to primary hippocampal neuronal networks. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 224:112701. [PMID: 34461321 DOI: 10.1016/j.ecoenv.2021.112701] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 06/13/2023]
Abstract
We previously reported that aluminum (Al) can cause a range of neurotoxic injuries including progressive irreversible synaptic structural damage and synaptic dysfunction, and eventually neuronal deaths. Mechanism of Al-induced electrophysiological and neuronal connectivity changes in neurons may indicate damage to the neuronal network. Here, mouse primary hippocampal neurons were cultured on micro-electrode array (MEA)- and high-content analysis (HCA)-related plates, showing that Al exposure significantly inhibited hippocampal neuronal electrical spike activity and neurite outgrowth characterized by a reduction in neurite branching and a decrease in the average total neurite length in relation to both Al dose and time of incubation. In recent years, miR-29a/ phosphatase and tensin homolog (PTEN) have been found to play pivotal roles in the morphogenesis of neurons, it has been confirmed in vitro and in vivo that the PTEN-Glycogen synthase kinase-3β (GSK-3β) axis regulates neurite outgrowth. The present study demonstrated that increases in Al exposure and dose gradually reduce miR-29a expression. Up-regulation of miR-29a in the hippocampal neurons by lentivirus transfection reversed the decrease in electrical spike activity and the reduction in both neurite branching and length induced by Al. Moreover, miR-29a suppressed the expression of PTEN and increased the level of phosphorylated Protein Kinase B (p-AKT) and p-GSK-3β which were inhibited by the Al treatment. This suggests that miR-29a is critically involved in the functional and structural neuronal damage induced by Al and is a potential target for Al neurotoxicity. Moreover, the reduction of neurite length and branching induced by Al exposure was regulated by miR-29a and its target neuronal PTEN-GSK3β signaling pathway, which also represents a possible mechanism of Al-induced the inhibition of the electrical activity. Collectively, Al-induced damage to the neuronal network occurred through miR-29a-mediated alterations of the PTEN-GSK3β signaling pathway.
Collapse
Affiliation(s)
- Huifang Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China.
| | - Xiaoya Cai
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Changxin Xiang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Yingchao Han
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China.
| |
Collapse
|
7
|
Marhuenda E, Fabre C, Zhang C, Martin-Fernandez M, Iskratsch T, Saleh A, Bauchet L, Cambedouzou J, Hugnot JP, Duffau H, Dennis JW, Cornu D, Bakalara N. Glioma stem cells invasive phenotype at optimal stiffness is driven by MGAT5 dependent mechanosensing. J Exp Clin Cancer Res 2021; 40:139. [PMID: 33894774 PMCID: PMC8067292 DOI: 10.1186/s13046-021-01925-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/23/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Glioblastomas stem-like cells (GSCs) by invading the brain parenchyma, remains after resection and radiotherapy and the tumoral microenvironment become stiffer. GSC invasion is reported as stiffness sensitive and associated with altered N-glycosylation pattern. Glycocalyx thickness modulates integrins mechanosensing, but details remain elusive and glycosylation enzymes involved are unknown. Here, we studied the association between matrix stiffness modulation, GSC migration and MGAT5 induced N-glycosylation in fibrillar 3D context. METHOD To mimic the extracellular matrix fibrillar microenvironments, we designed 3D-ex-polyacrylonitrile nanofibers scaffolds (NFS) with adjustable stiffnesses by loading multiwall carbon nanotubes (MWCNT). GSCs neurosphere were plated on NFSs, allowing GSCs migration and MGAT5 was deleted using CRISPR-Cas9. RESULTS We found that migration of GSCs was maximum at 166 kPa. Migration rate was correlated with cell shape, expression and maturation of focal adhesion (FA), Epithelial to Mesenchymal Transition (EMT) proteins and (β1,6) branched N-glycan binding, galectin-3. Mutation of MGAT5 in GSC inhibited N-glycans (β1-6) branching, suppressed the stiffness dependence of migration on 166 kPa NFS as well as the associated FA and EMT protein expression. CONCLUSION MGAT5 catalysing multibranched N-glycans is a critical regulators of stiffness induced invasion and GSCs mechanotransduction, underpinning MGAT5 as a serious target to treat cancer.
Collapse
Affiliation(s)
- Emilie Marhuenda
- Institut des Neurosciences de Montpellier (INM) U-1051, University of Montpellier, 80 rue Augustin Fliche, Hôpital Saint-Eloi, 34091, Montpellier, Cedex 5, France.
- School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS, UK.
- Institut Européen des Membranes, IEM, UMR 5635, University of Montpellier, ENSCM, CNRS, Montpellier, France.
| | - Christine Fabre
- Institut des Neurosciences de Montpellier (INM) U-1051, University of Montpellier, 80 rue Augustin Fliche, Hôpital Saint-Eloi, 34091, Montpellier, Cedex 5, France
- École nationale supérieure de chimie de Montpellier, ENSCM, 240 Avenue du Professeur Emile Jeanbrau, 34090, Montpellier, France
| | - Cunjie Zhang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Ave., Toronto, ON, M5G 1X5, Canada
- Department of Molecular Genetics, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Martà Martin-Fernandez
- Institut Charles Coulomb, UMR 5221, University of Montpellier, CNRS, Montpellier, France
| | - Thomas Iskratsch
- School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS, UK
| | - Ali Saleh
- Institut des Neurosciences de Montpellier (INM) U-1051, University of Montpellier, 80 rue Augustin Fliche, Hôpital Saint-Eloi, 34091, Montpellier, Cedex 5, France
| | - Luc Bauchet
- Institut des Neurosciences de Montpellier (INM) U-1051, University of Montpellier, 80 rue Augustin Fliche, Hôpital Saint-Eloi, 34091, Montpellier, Cedex 5, France
| | - Julien Cambedouzou
- Institut Européen des Membranes, IEM, UMR 5635, University of Montpellier, ENSCM, CNRS, Montpellier, France
- École nationale supérieure de chimie de Montpellier, ENSCM, 240 Avenue du Professeur Emile Jeanbrau, 34090, Montpellier, France
| | - Jean-Philippe Hugnot
- Institut des Neurosciences de Montpellier (INM) U-1051, University of Montpellier, 80 rue Augustin Fliche, Hôpital Saint-Eloi, 34091, Montpellier, Cedex 5, France
| | - Hugues Duffau
- Institut des Neurosciences de Montpellier (INM) U-1051, University of Montpellier, 80 rue Augustin Fliche, Hôpital Saint-Eloi, 34091, Montpellier, Cedex 5, France
| | - James W Dennis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Ave., Toronto, ON, M5G 1X5, Canada
- Department of Molecular Genetics, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - David Cornu
- Institut Européen des Membranes, IEM, UMR 5635, University of Montpellier, ENSCM, CNRS, Montpellier, France.
- École nationale supérieure de chimie de Montpellier, ENSCM, 240 Avenue du Professeur Emile Jeanbrau, 34090, Montpellier, France.
| | - Norbert Bakalara
- Institut des Neurosciences de Montpellier (INM) U-1051, University of Montpellier, 80 rue Augustin Fliche, Hôpital Saint-Eloi, 34091, Montpellier, Cedex 5, France.
- École nationale supérieure de chimie de Montpellier, ENSCM, 240 Avenue du Professeur Emile Jeanbrau, 34090, Montpellier, France.
| |
Collapse
|
8
|
Malthiery E, Chouaib B, Hernandez-Lopez AM, Martin M, Gergely C, Torres JH, Cuisinier FJ, Collart-Dutilleul PY. Effects of green light photobiomodulation on Dental Pulp Stem Cells: enhanced proliferation and improved wound healing by cytoskeleton reorganization and cell softening. Lasers Med Sci 2021; 36:437-445. [PMID: 32621128 DOI: 10.1007/s10103-020-03092-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 06/28/2020] [Indexed: 02/08/2023]
Abstract
Photobiomodulation (PBM) has been shown to improve cell proliferation and cell migration. Many cell types have been investigated, with most studies using deep penetrating red light irradiation. Considering the interest of surface biostimulation of oral mesenchymal cells after surgical wound, the present study aimed to assess green light irradiation effects on Dental Pulp Stem Cells' (DPSC) proliferation and migration. To understand the mechanisms underlying these effects, we investigated cytoskeleton organization and subsequent cell shape and stiffness. A 532-nm wavelength Nd:YAG laser (30 mW) was applied between 30 and 600 s on DPSC in vitro. Cell proliferation was analyzed at 24, 48, and 72 h after irradiation, by cell counting and enzymatic activity quantification (paranitrophenylphosphate phosphatase (pNPP) test). A wound healing assay was used to study cell migration after irradiation. Effects of PBM on cytoskeleton organization and cell shape were assessed by actin filaments staining. Elasticity changes after irradiation were quantified in terms of Young's modulus measured using Atomic Force Microscopy (AFM) force spectroscopy. Green light significantly improved DPSC proliferation with a maximal effect obtained after 300-s irradiation (energy fluence 5 J/cm2). This irradiation had a significant impact on cell migration, improving wound healing after 24 h. These results were concomitant with a decrease of cells' Young's modulus after irradiation. This cell softening was explained by actin cytoskeleton reorganization, with diminution of cell circularity and more abundant pseudopodia. This study highlights the interest of green laser PMB for the proliferation and migration of mesenchymal stem cells, with encouraging results for clinical application, especially for surgical wound healing procedures.
Collapse
Affiliation(s)
- Eve Malthiery
- LBN, University Montpellier, 545 Av Pr JL Viala, 34193 CEDEX 4, Montpellier, France
| | - Batoul Chouaib
- LBN, University Montpellier, 545 Av Pr JL Viala, 34193 CEDEX 4, Montpellier, France
| | - Ana María Hernandez-Lopez
- LBN, University Montpellier, 545 Av Pr JL Viala, 34193 CEDEX 4, Montpellier, France
- Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
| | - Marta Martin
- L2C, CNRS, University Montpellier, Montpellier, France
| | | | - Jacques-Henri Torres
- LBN, University Montpellier, 545 Av Pr JL Viala, 34193 CEDEX 4, Montpellier, France
| | - Frédéric J Cuisinier
- LBN, University Montpellier, 545 Av Pr JL Viala, 34193 CEDEX 4, Montpellier, France
| | | |
Collapse
|
9
|
Kiio TM, Park S. Nano-scientific Application of Atomic Force Microscopy in Pathology: from Molecules to Tissues. Int J Med Sci 2020; 17:844-858. [PMID: 32308537 PMCID: PMC7163363 DOI: 10.7150/ijms.41805] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/26/2020] [Indexed: 12/28/2022] Open
Abstract
The advantages of atomic force microscopy (AFM) in biological research are its high imaging resolution, sensitivity, and ability to operate in physiological conditions. Over the past decades, rigorous studies have been performed to determine the potential applications of AFM techniques in disease diagnosis and prognosis. Many pathological conditions are accompanied by alterations in the morphology, adhesion properties, mechanical compliances, and molecular composition of cells and tissues. The accurate determination of such alterations can be utilized as a diagnostic and prognostic marker. Alteration in cell morphology represents changes in cell structure and membrane proteins induced by pathologic progression of diseases. Mechanical compliances are also modulated by the active rearrangements of cytoskeleton or extracellular matrix triggered by disease pathogenesis. In addition, adhesion is a critical step in the progression of many diseases including infectious and neurodegenerative diseases. Recent advances in AFM techniques have demonstrated their ability to obtain molecular composition as well as topographic information. The quantitative characterization of molecular alteration in biological specimens in terms of disease progression provides a new avenue to understand the underlying mechanisms of disease onset and progression. In this review, we have highlighted the application of diverse AFM techniques in pathological investigations.
Collapse
Affiliation(s)
| | - Soyeun Park
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Daegu 42601, Republic of Korea
| |
Collapse
|
10
|
Miccoli B, Braeken D, Li YCE. Brain-on-a-chip Devices for Drug Screening and Disease Modeling Applications. Curr Pharm Des 2019; 24:5419-5436. [PMID: 30806304 DOI: 10.2174/1381612825666190220161254] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/12/2019] [Indexed: 01/06/2023]
Abstract
Neurodegenerative disorders are related to the progressive functional loss of the brain, often connected to emotional and physical disability and, ultimately, to death. These disorders, strongly connected to the aging process, are becoming increasingly more relevant due to the increase of life expectancy. Current pharmaceutical treatments poorly tackle these diseases, mainly acting only on their symptomology. One of the main reasons of this is the current drug development process, which is not only expensive and time-consuming but, also, still strongly relies on animal models at the preclinical stage. Organ-on-a-chip platforms have the potential to strongly impact and improve the drug screening process by recreating in vitro the functionality of human organs. Patient-derived neurons from different regions of the brain can be directly grown and differentiated on a brain-on-a-chip device where the disease development, progression and pharmacological treatments can be studied and monitored in real time. The model reliability is strongly improved by using human-derived cells, more relevant than animal models for pharmacological screening and disease monitoring. The selected cells will be then capable of proliferating and organizing themselves in the in vivo environment thanks to the device architecture, materials selection and bio-chemical functionalization. In this review, we start by presenting the fundamental strategies adopted for brain-on-a-chip devices fabrication including e.g., photolithography, micromachining and 3D printing technology. Then, we discuss the state-of-theart of brain-on-a-chip platforms including their role in the study of the functional architecture of the brain e.g., blood-brain barrier, or of the most diffuse neurodegenerative diseases like Alzheimer's and Parkinson's. At last, the current limitations and future perspectives of this approach for the development of new drugs and neurodegenerative diseases modeling will be discussed.
Collapse
Affiliation(s)
- Beatrice Miccoli
- Imec, Department of Life Sciences and Imaging, 3001 Heverlee, Belgium.,Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Dries Braeken
- Imec, Department of Life Sciences and Imaging, 3001 Heverlee, Belgium
| | - Yi-Chen Ethan Li
- Department of Chemical Engineering, Feng Chia University, Taichung City, Taiwan
| |
Collapse
|
11
|
Abdallah M, Martin M, El Tahchi MR, Balme S, Faour WH, Varga B, Cloitre T, Páll O, Cuisinier FJG, Gergely C, Bassil MJ, Bechelany M. Influence of Hydrolyzed Polyacrylamide Hydrogel Stiffness on Podocyte Morphology, Phenotype, and Mechanical Properties. ACS APPLIED MATERIALS & INTERFACES 2019; 11:32623-32632. [PMID: 31424195 DOI: 10.1021/acsami.9b09337] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Chronic kidney disease is characterized by a gradual decline in renal function that progresses toward end-stage renal disease. Podocytes are highly specialized glomerular epithelial cells which form with the glomerular basement membrane (GBM) and capillary endothelium the glomerular filtration barrier. GBM is an extracellular matrix (ECM) that acts as a mechanical support and provides biophysical signals that control normal podocytes behavior in the process of glomerular filtration. Thus, the ECM stiffness represents an essential characteristic that controls podocyte function. Hydrolyzed Polyacrylamide (PAAm) hydrogels are smart polyelectrolyte materials. Their biophysical properties can be tuned as desired to mimic the natural ECM. Therefore, these hydrogels are investigated as new ECM-like constructs to engineer a podocyte-like basement membrane that forms with cultured human podocytes a functional glomerular-like filtration barrier. Such ECM-like PAAm hydrogel construct will provide unique opportunity to reveal podocyte cell biological responses in an in vivo-like setting by controlling the physical properties of the PAAm membranes. In this work, Hydrolyzed PAAm scaffolds having different stiffness ranging between 0.6-44 kPa are prepared. The correlation between the hydrogel structural and mechanical properties and Podocyte morphology, elasticity, cytoskeleton reorganization, and podocin expression is evaluated. Results show that hydrolyzed PAAm hydrogels promote good cell adhesion and growth and are suitable materials for the development of future 3D smart scaffolds. In addition, the hydrogel properties can be easily modulated over a wide physiological range by controlling the cross-linker concentration. Finally, tuning the hydrogel properties is an effective strategy to control the cells function. This work addressed the complexity of podocytes behavior which will further enhance our knowledge to develop a kidney-on-chip model much needed in kidney function studies in both healthy and diseased states.
Collapse
Affiliation(s)
- Maya Abdallah
- Institut Européen des Membranes, ENSCM, CNRS , Université de Montpellier , Montpellier 34090 , France
- Biomaterials and Intelligent Materials Research Laboratory (LBMI) , Lebanese University , Faculty of Sciences 2, Physic Department , Jdeidet 90656 , Lebanon
| | - Marta Martin
- Laboratoire Charles Coulomb , Université de Montpellier , CNRS , Montpellier 34095 , France
| | - Mario R El Tahchi
- Biomaterials and Intelligent Materials Research Laboratory (LBMI) , Lebanese University , Faculty of Sciences 2, Physic Department , Jdeidet 90656 , Lebanon
| | - Sebastien Balme
- Institut Européen des Membranes, ENSCM, CNRS , Université de Montpellier , Montpellier 34090 , France
| | - Wissam H Faour
- Gilbert and Rose-Marie Chagoury School of Medicine , Lebanese American University , P.O. Box 36 , Byblos , Lebanon
| | - Béla Varga
- Laboratoire Charles Coulomb , Université de Montpellier , CNRS , Montpellier 34095 , France
| | - Thierry Cloitre
- Laboratoire Charles Coulomb , Université de Montpellier , CNRS , Montpellier 34095 , France
| | - Orsolya Páll
- Laboratoire de Bioingénierie et Nanosciences , Université de Montpellier , Montpellier 34090 , France
| | - Frédéric J G Cuisinier
- Laboratoire de Bioingénierie et Nanosciences , Université de Montpellier , Montpellier 34090 , France
| | - Csilla Gergely
- Laboratoire Charles Coulomb , Université de Montpellier , CNRS , Montpellier 34095 , France
| | - Maria J Bassil
- Biomaterials and Intelligent Materials Research Laboratory (LBMI) , Lebanese University , Faculty of Sciences 2, Physic Department , Jdeidet 90656 , Lebanon
| | - Mikhael Bechelany
- Institut Européen des Membranes, ENSCM, CNRS , Université de Montpellier , Montpellier 34090 , France
| |
Collapse
|
12
|
Bahri A, Martin M, Gergely C, Marchesseau S, Chevalier-Lucia D. Topographical and nanomechanical characterization of casein nanogel particles using atomic force microscopy. Food Hydrocoll 2018. [DOI: 10.1016/j.foodhyd.2018.03.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
13
|
Diniz LRL, Portella VG, da Silva Alves KS, Araújo PCDC, de Albuquerque Júnior RLC, Cavalcante de Albuquerque AA, Coelho-de-Souza AN, Leal-Cardoso JH. Electrophysiologic alterations in the excitability of the sciatic and vagus nerves during early stages of sepsis. J Pain Res 2018; 11:783-790. [PMID: 29731661 PMCID: PMC5927063 DOI: 10.2147/jpr.s144220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Nonspecific and delayed diagnosis of neurologic damage contributes to the development of neuropathies in patients with severe sepsis. The present study assessed the electrophysiologic parameters related to the excitability and conductibility of sciatic and vagus nerves during early stages of sepsis. Materials and methods Twenty-four hours after sepsis induced by cecal ligation and puncture (CLP) model, sciatic and vagus nerves of septic (CLP group) and control (sham group) rats were removed, and selected electric stimulations were applied to measure the parameters of the first and second components of the compound action potential. The first component originated from fibers with motor and sensory functions (Types Aα and Aβ fibers) with a large conduction velocity (70-120 m/s), and the second component originated from fibers (Type Aγ) with sensorial function. To evaluate the presence of sensorial alterations, the sensitivity to non-noxious mechanical stimuli was measured by using the von Frey test. Hematoxylin and eosin staining of the nerves was performed. Results We observed an increase of rheobase followed by a decrease in the first component amplitude and a higher paw withdrawal threshold in response to the application of von Frey filaments in sciatic nerves from the CLP group compared to the sham group. Differently, a decrease in rheobase and an increase in the first component amplitude of vagal C fibers from CLP group were registered. No significant morphologic alteration was observed. Conclusion Our data showed that the electrophysiologic alterations in peripheral nerves vary with the fiber type and might be identified in the first 24 h of sepsis, before clinical signs of neuromuscular disorders.
Collapse
Affiliation(s)
| | - Viviane Gomes Portella
- Department of Physiology, Superior Institute of Biomedical Sciences, State University of Ceará, Fortaleza, Brazil
| | - Kerly Shamira da Silva Alves
- Department of Physiology, Superior Institute of Biomedical Sciences, State University of Ceará, Fortaleza, Brazil
| | | | | | | | | | - José Henrique Leal-Cardoso
- Department of Physiology, Superior Institute of Biomedical Sciences, State University of Ceará, Fortaleza, Brazil
| |
Collapse
|
14
|
Myotube elasticity of an amyotrophic lateral sclerosis mouse model. Sci Rep 2018; 8:5917. [PMID: 29650983 PMCID: PMC5897453 DOI: 10.1038/s41598-018-24027-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/14/2018] [Indexed: 01/02/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that affects the motor system leading to generalized paralysis and death of patients. The understanding of early pathogenic mechanisms will help to define early diagnostics criteria that will eventually provide basis for efficient therapeutics. Early symptoms of ALS usually include muscle weakness or stiffness. Therefore, mechanical response of differentiated myotubes from primary cultures of mice, expressing the ALS-causing SOD1G93A mutation, was examined by atomic force microscopy. Simultaneous acquisition of topography and cell elasticity of ALS myotubes was performed by force mapping method, compared with healthy myotubes and supplemented with immunofluorescence and qRT-PCR studies. Wild type myotubes reveal a significant difference in elasticity between a narrow and a wide population, consistent with maturation occurring with higher actin expression relative to myosin together with larger myotube width. However, this is not true for SOD1G93A expressing myotubes, where a significant shift of thin population towards higher elastic modulus values was observed. We provide evidence that SOD1 mutant induces structural changes that occurs very early in muscle development and well before symptomatic stage of the disease. These findings could significantly contribute to the understanding of the role of skeletal muscle in ALS pathogenesis.
Collapse
|
15
|
Guo YC, Wang YX, Ge YP, Yu LJ, Guo J. Analysis of subcellular structural tension in axonal growth of neurons. Rev Neurosci 2018; 29:125-137. [DOI: 10.1515/revneuro-2017-0047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/05/2017] [Indexed: 01/08/2023]
Abstract
AbstractThe growth and regeneration of axons are the core processes of nervous system development and functional recovery. They are also related to certain physiological and pathological conditions. For decades, it has been the consensus that a new axon is formed by adding new material at the growth cone. However, using the existing technology, we have studied the structural tension of the nerve cell, which led us to hypothesize that some subcellular structural tensions contribute synergistically to axonal growth and regeneration. In this review, we classified the subcellular structural tension, osmotic pressure, microfilament and microtubule-dependent tension involved controllably in promoting axonal growth. A squeezing model was built to analyze the mechanical mechanism underlying axonal elongation, which may provide a new view of axonal growth and inspire further research.
Collapse
|
16
|
Bahri A, Martin M, Gergely C, Pugnière M, Chevalier-Lucia D, Marchesseau S. Atomic Force Microscopy Study of the Topography and Nanomechanics of Casein Micelles Captured by an Antibody. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:4720-4728. [PMID: 28481103 DOI: 10.1021/acs.langmuir.7b00311] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Casein micelles (CMs) are colloidal phospho-protein-mineral complexes naturally present in milk. This study used atomic force microscopy (AFM) in a liquid environment to evaluate the topography and nanomechanics of single native CMs immobilized by a novel capture method. The proposed immobilization method involves weak interactions with the antiphospho-Ser/Thr/Tyr monoclonal antibody covalently bound to a carboxylic acid self-assembled monolayer (SAM) on a gold surface. This capture strategy was compared to the commonly used covalent immobilization method of CMs via carbodiimide chemistry. With this conventional method, CMs remained mainly mobile during AFM measurements in liquid, disturbing the evaluation of their average size and elastic properties. Conversely, when captured by the specific antibody, they were successfully immobilized and their integrity was preserved during the AFM measurement. The characterization of both CM topography and elastic properties was carried out in a liquid ionic environment at native pH 6.6. The CMs' capture efficiency via antibody was concurrently proved by surface plasmon resonance. The calculation of casein micelles' width, height, and contact angle was carried out from the recorded 2D AFM images. CMs were characterized by a mean width of 148 ± 8 nm and a mean height of 42 ± 1 nm. Weak forces were applied to single captured CMs. The obtained force versus indentation curves were fitted using the Hertz model in order to evaluate their elastic properties. The elasticity distribution of native CMs exhibited a unimodal trend with a peak centered at 269 ± 14 kPa.
Collapse
Affiliation(s)
- Asma Bahri
- Université de Montpellier , UMR IATE, F-34095 Montpellier Cedex 05, France
| | - Marta Martin
- Laboratoire Charles Coulomb, Université de Montpellier , UMR 5221-CNRS, F-34095 Montpellier Cedex 05, France
| | - Csilla Gergely
- Laboratoire Charles Coulomb, Université de Montpellier , UMR 5221-CNRS, F-34095 Montpellier Cedex 05, France
| | - Martine Pugnière
- IRCM-CRLC Val d'Aurelle - INSERM U896, F-34298 Montpellier Cedex 5, France
| | | | - Sylvie Marchesseau
- Université de Montpellier , UMR IATE, F-34095 Montpellier Cedex 05, France
| |
Collapse
|
17
|
Heine P, Ehrlicher A, Käs J. Neuronal and metastatic cancer cells: Unlike brothers. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:3126-31. [DOI: 10.1016/j.bbamcr.2015.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/10/2015] [Accepted: 06/12/2015] [Indexed: 12/22/2022]
|
18
|
Tian J, Tu C, Liang Y, Zhou J, Ye X. Study of laser uncaging induced morphological alteration of rat cortical neurites using atomic force microscopy. J Neurosci Methods 2015; 253:151-60. [PMID: 26149288 DOI: 10.1016/j.jneumeth.2015.06.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 06/18/2015] [Accepted: 06/26/2015] [Indexed: 11/28/2022]
Abstract
Activity-dependent structural remodeling is an important aspect of neuronal plasticity. In the previous researches, neuronal structure variations resulting from external interventions were detected by the imaging instruments such as the fluorescence microscopy, the scanning/transmission electron microscopy (SEM/TEM) and the laser confocal microscopy. In this article, a new platform which combined the photochemical stimulation with atomic force microscopy (AFM) was set up to detect the activity-dependent structural remodeling. In the experiments, the cortical neurites on the glass coverslips were stimulated by locally uncaged glutamate under the ultraviolet (UV) laser pulses, and a calcium-related structural collapse of neurites (about 250 nm height decrease) was observed by an AFM. This was the first attempt to combine the laser uncaging with AFM in living cell researches. With the advantages of highly localized stimulation (<5 μm), super resolution imaging (<3.8 nm), and convenient platform building, this system was suitable for the quantitative observation of the neuron mechanical property variations and morphological alterations modified by neural activities under different photochemical stimulations, which would be helpful for studying physiological and pathological mechanisms of structural and functional changes induced by the biomolecule acting.
Collapse
Affiliation(s)
- Jian Tian
- Biosensor National Special Laboratory, Key Laboratory of BME of the Ministry of Education, Zhejiang University, Hangzhou 310027, PR China; Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, PR China.
| | - Chunlong Tu
- Biosensor National Special Laboratory, Key Laboratory of BME of the Ministry of Education, Zhejiang University, Hangzhou 310027, PR China; Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, PR China.
| | - Yitao Liang
- Biosensor National Special Laboratory, Key Laboratory of BME of the Ministry of Education, Zhejiang University, Hangzhou 310027, PR China; Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, PR China.
| | - Jian Zhou
- Biosensor National Special Laboratory, Key Laboratory of BME of the Ministry of Education, Zhejiang University, Hangzhou 310027, PR China; Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, PR China.
| | - Xuesong Ye
- Biosensor National Special Laboratory, Key Laboratory of BME of the Ministry of Education, Zhejiang University, Hangzhou 310027, PR China; Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, PR China.
| |
Collapse
|
19
|
Jembrek MJ, Šimić G, Hof PR, Šegota S. Atomic force microscopy as an advanced tool in neuroscience. Transl Neurosci 2015; 6:117-130. [PMID: 28123795 PMCID: PMC4936619 DOI: 10.1515/tnsci-2015-0011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/05/2015] [Indexed: 12/16/2022] Open
Abstract
This review highlights relevant issues about applications and improvements of atomic force microscopy (AFM) toward a better understanding of neurodegenerative changes at the molecular level with the hope of contributing to the development of effective therapeutic strategies for neurodegenerative illnesses. The basic principles of AFM are briefly discussed in terms of evaluation of experimental data, including the newest PeakForce Quantitative Nanomechanical Mapping (QNM) and the evaluation of Young’s modulus as the crucial elasticity parameter. AFM topography, revealed in imaging mode, can be used to monitor changes in live neurons over time, representing a valuable tool for high-resolution detection and monitoring of neuronal morphology. The mechanical properties of living cells can be quantified by force spectroscopy as well as by new AFM. A variety of applications are described, and their relevance for specific research areas discussed. In addition, imaging as well as non-imaging modes can provide specific information, not only about the structural and mechanical properties of neuronal membranes, but also on the cytoplasm, cell nucleus, and particularly cytoskeletal components. Moreover, new AFM is able to provide detailed insight into physical structure and biochemical interactions in both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
| | - Goran Šimić
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, 10029 New York, USA
| | - Suzana Šegota
- Division for Marine and Environmental Research, Ruđer Bošković Institute, POB 180, Zagreb, Croatia
| |
Collapse
|
20
|
Panayotov IV, Collart-Dutilleul PY, Salehi H, Martin M, Végh A, Yachouh J, Vladimirov B, Sipos P, Szalontai B, Gergely C, Cuisinier FJG. Sprayed cells and polyelectrolyte films for biomaterial functionalization: the influence of physical PLL-PGA film treatments on dental pulp cell behavior. Macromol Biosci 2014; 14:1771-1782. [PMID: 25212873 DOI: 10.1002/mabi.201400256] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 08/07/2014] [Indexed: 02/05/2023]
Abstract
Further development of biomaterials is expected as advanced therapeutic products must be compliant to good manufacturing practice regulations. A spraying method for building-up polyelectrolyte films followed by the deposition of dental pulp cells by spraying is presented. Physical treatments of UV irradiation and a drying/wetting process are applied to the system. Structural changes and elasticity modifications of the obtained coatings are revealed by atomic force microscopy and by Raman spectroscopy. This procedure results in thicker, rougher and stiffer film. The initially ordered structure composed of mainly α helices is transformed into random/β-structures. The treatment enhanced dental pulp cell adhesion and proliferation, suggesting that this system is promising for medical applications.
Collapse
Affiliation(s)
- Ivan V Panayotov
- EA4203 Laboratoire de Bio-santé et Nano-science, Université Montpellier 1, Montpellier, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Benzina O, Cloitre T, Martin M, Raoul C, Gergely C, Scamps F. Morphology and intrinsic excitability of regenerating sensory and motor neurons grown on a line micropattern. PLoS One 2014; 9:e110687. [PMID: 25329060 PMCID: PMC4203813 DOI: 10.1371/journal.pone.0110687] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 09/15/2014] [Indexed: 12/03/2022] Open
Abstract
Axonal regeneration is one of the greatest challenges in severe injuries of peripheral nerve. To provide the bridge needed for regeneration, biological or synthetic tubular nerve constructs with aligned architecture have been developed. A key point for improving axonal regeneration is assessing the effects of substrate geometry on neuronal behavior. In the present study, we used an extracellular matrix-micropatterned substrate comprising 3 µm wide lines aimed to physically mimic the in vivo longitudinal axonal growth of mice peripheral sensory and motor neurons. Adult sensory neurons or embryonic motoneurons were seeded and processed for morphological and electrical activity analyses after two days in vitro. We show that micropattern-guided sensory neurons grow one or two axons without secondary branching. Motoneurons polarity was kept on micropattern with a long axon and small dendrites. The micro-patterned substrate maintains the growth promoting effects of conditioning injury and demonstrates, for the first time, that neurite initiation and extension could be differentially regulated by conditioning injury among DRG sensory neuron subpopulations. The micro-patterned substrate impacts the excitability of sensory neurons and promotes the apparition of firing action potentials characteristic for a subclass of mechanosensitive neurons. The line pattern is quite relevant for assessing the regenerative and developmental growth of sensory and motoneurons and offers a unique model for the analysis of the impact of geometry on the expression and the activity of mechanosensitive channels in DRG sensory neurons.
Collapse
Affiliation(s)
- Ouafa Benzina
- Université Montpellier 2, Laboratoire Charles Coulomb UMR 5221, Montpellier, France
- CNRS, Laboratoire Charles Coulomb UMR 5221, Montpellier, France
| | - Thierry Cloitre
- Université Montpellier 2, Laboratoire Charles Coulomb UMR 5221, Montpellier, France
- CNRS, Laboratoire Charles Coulomb UMR 5221, Montpellier, France
| | - Marta Martin
- Université Montpellier 2, Laboratoire Charles Coulomb UMR 5221, Montpellier, France
- CNRS, Laboratoire Charles Coulomb UMR 5221, Montpellier, France
| | - Cédric Raoul
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR1051, the neuroscience institute of Montpellier, Saint Eloi hospital, Montpellier, France
- Université Montpellier 1, 2, Montpellier, France
| | - Csilla Gergely
- Université Montpellier 2, Laboratoire Charles Coulomb UMR 5221, Montpellier, France
- CNRS, Laboratoire Charles Coulomb UMR 5221, Montpellier, France
| | - Frédérique Scamps
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR1051, the neuroscience institute of Montpellier, Saint Eloi hospital, Montpellier, France
- Université Montpellier 1, 2, Montpellier, France
- * E-mail:
| |
Collapse
|
22
|
Du Y, Zhang J, Zheng Q, Li M, Liu Y, Zhang B, Liu B, Zhang H, Miao G. Heavy ion and X-ray irradiation alter the cytoskeleton and cytomechanics of cortical neurons. Neural Regen Res 2014; 9:1129-37. [PMID: 25206772 PMCID: PMC4146101 DOI: 10.4103/1673-5374.135315] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2014] [Indexed: 12/26/2022] Open
Abstract
Heavy ion beams with high linear energy transfer exhibit more beneficial physical and biological performance than conventional X-rays, thus improving the potential of this type of radiotherapy in the treatment of cancer. However, these two radiotherapy modalities both cause inevitable brain injury. The objective of this study was to evaluate the effects of heavy ion and X-ray irradiation on the cytoskeleton and cytomechanical properties of rat cortical neurons, as well as to determine the potential mechanism of neuronal injury after irradiation. Cortical neurons from 30 new-born mice were irradiated with heavy ion beams at a single dose of 2 Gy and X-rays at a single dose of 4 Gy; subsequent evaluation of their effects were carried out at 24 hours after irradiation. An immunofluorescence assay showed that after irradiation with both the heavy ion beam and X-rays, the number of primary neurons was significantly decreased, and there was evidence of apoptosis. Radiation-induced neuronal injury was more apparent after X-irradiation. Under atomic force microscopy, the neuronal membrane appeared rough and neuronal rigidity had increased. These cell changes were more apparent following exposure to X-rays. Our findings indicated that damage caused by heavy ion and X-ray irradiation resulted in the structural distortion and rearrangement of the cytoskeleton, and affected the cytomechanical properties of the cortical neurons. Moreover, this radiation injury to normal neurons was much severer after irradiation with X-rays than after heavy ion beam irradiation.
Collapse
Affiliation(s)
- Yuting Du
- School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, China ; School of Nuclear Science and Technology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Jie Zhang
- School of Nuclear Science and Technology, Lanzhou University, Lanzhou, Gansu Province, China ; School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Qian Zheng
- School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Mingxin Li
- School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yang Liu
- Department of Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu Province, China
| | - Baoping Zhang
- School of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu Province, China
| | - Bin Liu
- School of Nuclear Science and Technology, Lanzhou University, Lanzhou, Gansu Province, China ; School of Stomatology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Hong Zhang
- Department of Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu Province, China
| | - Guoying Miao
- Gansu Provincial Hospital, Lanzhou, Gansu Province, China
| |
Collapse
|
23
|
Spedden E, Staii C. Neuron biomechanics probed by atomic force microscopy. Int J Mol Sci 2013; 14:16124-40. [PMID: 23921683 PMCID: PMC3759903 DOI: 10.3390/ijms140816124] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 07/16/2013] [Accepted: 07/18/2013] [Indexed: 11/16/2022] Open
Abstract
Mechanical interactions play a key role in many processes associated with neuronal growth and development. Over the last few years there has been significant progress in our understanding of the role played by the substrate stiffness in neuronal growth, of the cell-substrate adhesion forces, of the generation of traction forces during axonal elongation, and of the relationships between the neuron soma elastic properties and its health. The particular capabilities of the Atomic Force Microscope (AFM), such as high spatial resolution, high degree of control over the magnitude and orientation of the applied forces, minimal sample damage, and the ability to image and interact with cells in physiologically relevant conditions make this technique particularly suitable for measuring mechanical properties of living neuronal cells. This article reviews recent advances on using the AFM for studying neuronal biomechanics, provides an overview about the state-of-the-art measurements, and suggests directions for future applications.
Collapse
Affiliation(s)
- Elise Spedden
- Department of Physics and Astronomy and Center for Nanoscopic Physics, Tufts University, 4 Colby Street, Medford, MA 02155, USA; E-Mail:
| | - Cristian Staii
- Department of Physics and Astronomy and Center for Nanoscopic Physics, Tufts University, 4 Colby Street, Medford, MA 02155, USA; E-Mail:
| |
Collapse
|