1
|
Sun Q, Mu X, Gao Q, Wang J, Hu M, Liu H. Influences of physical stimulations on the migration and differentiation of Schwann cells involved in peripheral nerve repair. Cell Adh Migr 2025; 19:2450311. [PMID: 39817348 PMCID: PMC11740713 DOI: 10.1080/19336918.2025.2450311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025] Open
Abstract
Peripheral nerve injury repair has always been a research concern of scientists. At the tissue level, axonal regeneration has become a research spotlight in peripheral nerve repair. Through transplantation of autologous nerve grafts or other emerging biomaterials functional recovery after facial nerve injury is not ideal in clinical scenarios. Great strides have been made to improve facial nerve repair at the micro-cellular level. Physical stimulation techniques can trigger Schwann cells (SCs) to migrate and differentiate into cells required for peripheral nerve repair. Classified by the sources of physical stimulations, SCs repair peripheral nerves through galvanotaxis, magnetotaxis and durotaxis. This article summarized the activation, directional migration and differentiation of SCs induced by physical stimulations, thus providing new ideas for the research of peripheral nerve repair.
Collapse
Affiliation(s)
- Qingyan Sun
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Chinese People’s Liberation Army (PLA) Medical School, Beijing, China
| | - Xiaodan Mu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Chinese People’s Liberation Army (PLA) Medical School, Beijing, China
- Department of Stomatology of Air Force Hospital in the Southern Theater, Guangzhou, Guangdong Province, China
| | - Qi Gao
- Department of Stomatology of Air Force Hospital in the Southern Theater, Guangzhou, Guangdong Province, China
| | - Juncheng Wang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Min Hu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Huawei Liu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
2
|
Xu Y, Rentuya N, Yu T, Yan J, Zhang H, Zhang Y, Zhang H, Sun J, Liu J. Tuina promotes nerve myelin regeneration in SNI rats through Piezo1/YAP/TAZ pathway. J Orthop Surg Res 2025; 20:454. [PMID: 40350445 PMCID: PMC12067706 DOI: 10.1186/s13018-025-05794-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
PURPOSE The changes in the mechanical environment of local nerves after peripheral nerve injury (PNI) can cause a series of mechanical electrochemical signal reactions that affect the process of nerve regeneration and functional recovery. Piezo1/YAP/TAZ is an important transduction pathway that affects myelin regeneration. Our previous studies showed that Tuina could treat PNI in a variety of ways, including promoting nerve repair. However, whether Tuina as a kind of benign mechanical stimulation could promote nerve repair by changing the neuromechanical environment and causing changes in the mechanical electrochemical signal transduction pathway Piezo1/YAP/TAZ is unknown. METHODS The rats were divided into 4 groups, Sham group, sciatic nerve injury (SNI) group, Tuina group and Tuina + GsMTx4 group, with 6 rats in each group. We established an SNI model. Sciatic nerves at the mid-thigh level were exposed and crushed using a pair of non-serrated forceps for 5 s and the damage points about 2 mm. We used a Tuina manipulation emulator designed by our team to intervent. According to the "Three-Manipulation and Three-Acupoint": the emulator was used to perform the Dian, Bo, and Rou methods on Yinmen (BL37), Chengshan (BL57) and Yanglingquan (GB34) sequentially on the affected side. Each Tuina method was applied for 1 min on each acupoint respectively. Tuina treatment was administered once daily for 20 days. And we observed Somatic Functional Index (SFI), Mechanical Withdrawal Threshold (MWT), electrophysiological test and Shear wave elastography (SWE) examination in each group. Toluidine blue staining was performed to observe nerve fibers. The expression of Piezo1, Yes-associated protein (YAP), transcriptional coactivator with PDZ-binding motif (TAZ), Myelin basic protein (MBP), Neurofilament 200 (NF200), S100 calcium-binding protein β(S100β) and Ca2+ were detected using Immunofluorescence (IF), Western Blot (WB), Real-Time Quantitative PCR (RT-PCR) and Calcium Assay Kit. RESULTS Tuina improved the SFI, MWT, and compound action potential (CMAP) changes after SNI. The SWE results showed that Tuina reduced Emax and Smax. Piezo1, Ca2+ expression were reduced, YAP, TAZ, MBP, NF200, S100β expression were enhanced by Tuina. CONCLUSION The activation of Schwann cells (SCs) and the regeneration of injured nerve myelin post-Tuina intervention are associated with alterations in the Piezo1/YAP/TAZ signaling pathway within SCs, induced by the mechanical forces generated through Tuina.
Collapse
Affiliation(s)
- Yue Xu
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Na Rentuya
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Tianyuan Yu
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People's Republic of China.
| | - Jiawang Yan
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Hongzheng Zhang
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Yingqi Zhang
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Hanyu Zhang
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Jiawei Sun
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Jiayue Liu
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| |
Collapse
|
3
|
Li Z, Su T, Yang Y, Zhao H. Construction of Multicellular Neural Tissue Using Three-Dimensional Printing Technology: Cell Interaction. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40256794 DOI: 10.1089/ten.teb.2024.0323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
The study of the human nervous system remains challenging due to its inherent complexity and difficulty in obtaining original samples. Three-dimensional (3D) bioprinting is a rapidly evolving technology in the field of tissue engineering that has made significant contributions to several disciplines, including neuroscience. In order to more accurately reflect the intricate multicellular milieu of the in vivo environment, an increasing number of studies have commenced experimentation with the coprinting of diverse cell types. This article provides an overview of technical details and the application of 3D bioprinting with multiple cell types in the field of neuroscience, focusing on the challenges of coprinting and the research conducted based on multicellular printing. This review discusses cell interactions in coprinting systems, stem cell applications, the construction of brain-like organoids, the establishment of disease models, and the potential for integrating 3D bioprinting with other 3D culture techniques.
Collapse
Affiliation(s)
- Zhixiang Li
- Tissue Engineering Laboratory, School of Biology, Food, and Environment, Hefei University, Hefei, PR China
| | - Tong Su
- Tissue Engineering Laboratory, School of Biology, Food, and Environment, Hefei University, Hefei, PR China
| | - Yujie Yang
- Tissue Engineering Laboratory, School of Biology, Food, and Environment, Hefei University, Hefei, PR China
| | - Huan Zhao
- Tissue Engineering Laboratory, School of Biology, Food, and Environment, Hefei University, Hefei, PR China
| |
Collapse
|
4
|
Zeng Z, Chen E, Xue J. Emerging roles of mechanically activated ion channels in autoimmune disease. Autoimmun Rev 2025; 24:103813. [PMID: 40194731 DOI: 10.1016/j.autrev.2025.103813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
Mechanically activated (MA) ion channels have rapidly gained prominence as vital conduits bridging aberrant mechanical cues in tissues with the dysregulated immune responses at the core of autoimmune diseases. Once regarded as peripheral players in inflammation, these channels, exemplified by PIEZO1, TRPV4, and specific K2P family members, now play a central role in modulating T-cell effector functions, B- cell activation and the activity of macrophages and dendritic cells. Their gating is intimately tied to physical distortions such as increased tissue stiffness, osmotic imbalances, or fluid shear, triggering a cascade of ionic fluxes that elevate proinflammatory signaling and drive tissue-destructive loops. Recognition of these channels as central mediators of mechanical stress-induced inflammation responses in autoimmune pathogenesis is rapidly expanding. In parallel, the emerging therapeutic strategies aim to restrain overactive mechanosensors or selectively harness them in affected tissues. Small molecules, peptide blockers, and gene-targeting approaches show preclinical promise, although off-target effects and the broader homeostatic roles of these channels warrant caution. This review explores how integrating mechanobiological concepts with established immunological paradigms enables a more detailed understanding of autoimmune pathogenesis. By elucidating how mechanical forces potentiate or dampen pathological immunity, we propose innovative strategies that exploit mechanosensitivity to recalibrate immune responses across a spectrum of autoimmune conditions.
Collapse
Affiliation(s)
- Zhiru Zeng
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Engeng Chen
- Department of Zhejiang Provincial Key Laboratory of Biotherapy, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Jing Xue
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
5
|
Loret C, Scherrer C, Rovini A, Lesage E, Richard L, Danigo A, Sturtz F, Favreau F, Faye PA, Lia AS. Addressing myelination disorders: Novel strategies using human 3D peripheral nerve model. Brain Res Bull 2025; 222:111252. [PMID: 39938756 DOI: 10.1016/j.brainresbull.2025.111252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
Peripheral myelination disorders encompass a variety of disorders that affect myelin sheaths in the peripheral nervous system. The Charcot-Marie-Tooth disease (CMT), the most common inherited peripheral neuropathy, is one of the most prevalent among them. CMT stems from a wide range of genetic causes that disrupt the nerve conduction, leading to progressive muscle weakness and atrophy, sensory loss, and motor function impairment. Historically, the study of these disorders has relied heavily on animal studies, owing to the challenges in accessing human cells. However, the advent of human induced pluripotent stem cell (hiPSC)-derived neuronal cells has addressed these limitations in the realm of peripheral myelination disorders. Despite this, obtaining myelin in these models remains an expensive, time-consuming, and material-intensive process. This study presents a novel, cost-effective method utilizing hiPSC-derived Schwann cells and motor neurons in a three-dimensional culture system. Our method successfully enabled the acquisition of myelin in a control clone within just four weeks, as confirmed by electron microscopy. Furthermore, the utility of these approaches was validated by studying CMT4C, also named AR-CMTde-SH3TC2, the most common recessive demyelinating form of CMT. This revealed defects in Schwann cell support to motor neuron neurite outgrowth and impaired myelination in disease-specific hiPSC-derived lines. This approach offers valuable insights into the pathogenesis of peripheral myelination disorders and provides a platform for testing potential therapeutic strategies.
Collapse
Affiliation(s)
- Camille Loret
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France.
| | - Camille Scherrer
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France
| | - Amandine Rovini
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France
| | - Esther Lesage
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France
| | - Laurence Richard
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Service de Neurologie, Limoges F-87000, France
| | - Aurore Danigo
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Service de Neurologie, Limoges F-87000, France
| | - Franck Sturtz
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Department of Biochemistry and Molecular Genetics, Limoges F-87000, France
| | - Frédéric Favreau
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Department of Biochemistry and Molecular Genetics, Limoges F-87000, France.
| | - Pierre-Antoine Faye
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Department of Biochemistry and Molecular Genetics, Limoges F-87000, France
| | - Anne-Sophie Lia
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Department of Biochemistry and Molecular Genetics, Limoges F-87000, France; CHU Limoges, Department of Bioinformatics, Limoges F-87000, France
| |
Collapse
|
6
|
Rambo M, Agarwala I, Vanek C, Xiao Y, Brown E, Mills KL. Schwann Cells Deficient in Neurofibromin Lack Sensitivity to Their Biomechanical Microenvironment. Genes Chromosomes Cancer 2025; 64:e70036. [PMID: 39996425 DOI: 10.1002/gcc.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND AND AIMS Plexiform neurofibromas (PNFs) are benign tumors of the peripheral nervous system that affect approximately 30% of people with neurofibromatosis type 1 (NF1). Schwann cells (SCs), the tumor progenitor cells, respond to and use biomechanical signals like tissue stiffness and mechanical loads in their maintenance and repair functions in healthy tissues. PNFs are described as having altered biomechanics, and we hypothesize this plays a role in PNF development. As a first step in studying the role that altered biomechanics may play in the development of PNFs, we aimed to determine how PNF SCs alter in their response to various biomechanical signals as compared to healthy SCs. METHODS We examined the behavior of healthy and PNF SCs in three different tissue-mimicking biomechanical models. First, we examined their spreading behavior on extracellular matrix (ECM) protein-coated polyacrylamide hydrogels of varying stiffness in the healthy and pathological range. Second, we investigated their collective migration with respect to substrate stiffness and ECM protein-coating. Finally, we generated multicellular spheroid tissue models using healthy and PNF SCs and measured their mechanical properties as a function of spheroid size. RESULTS We found that PNF SCs are differently sensitive to substrate stiffness in a physiological range compared to healthy SCs, lack sensitivity to ECM protein coating when collectively migrating, and lack sensitivity to environmental deficiencies in oxygen and nutrient supplies when in spheroid culture. INTERPRETATION We propose that PNF SC altered biomechanics likely play a role in tumor initiation and progression, and that further biomechanical-based investigations of NF1 tumor growth are needed.
Collapse
Affiliation(s)
- Micah Rambo
- Biomedical Engineering Department, Rensselaer Polytechnic Institute, Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Isheka Agarwala
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
- Mechanical, Aerospace, and Nuclear Engineering Department, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Camdyn Vanek
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
- Mechanical, Aerospace, and Nuclear Engineering Department, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Yuxin Xiao
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
- Mechanical, Aerospace, and Nuclear Engineering Department, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Emma Brown
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
- Mechanical, Aerospace, and Nuclear Engineering Department, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - K L Mills
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
- Mechanical, Aerospace, and Nuclear Engineering Department, Rensselaer Polytechnic Institute, Troy, New York, USA
| |
Collapse
|
7
|
Montgomery A, Westphal J, Bryan AE, Harris GM. Dynamically changing extracellular matrix stiffness drives Schwann cell phenotype. Matrix Biol Plus 2025; 25:100167. [PMID: 39868413 PMCID: PMC11754676 DOI: 10.1016/j.mbplus.2024.100167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025] Open
Abstract
Schwann cells (SCs) hold key roles in axonal function and maintenance in the peripheral nervous system (PNS) and are a critical component to the regeneration process following trauma. Following PNS trauma, SCs respond to both physical and chemical signals to modify phenotype and assist in the regeneration of damaged axons and extracellular matrix (ECM). There is currently a lack of knowledge regarding the SC response to dynamic, temporal changes in the ECM brought on by swelling and the development of scar tissue as part of the body's wound-healing process. Thus, this work seeks to utilize a biocompatible, mechanically tunable biomaterial to mimic changes in the microenvironment following injury and over time. Previously, we have reported that ECM cues such as ligand type and substrate stiffness impact SC phenotype and plasticity, which was demonstrated by SCs on mechanically stable biomaterials. However, to better realize SC potential for plasticity following traumatic injury, a UV-tunable polydimethylsiloxane (PDMS) substrate with dynamically changing stiffness was utilized to mimic changes over time in the microenvironment. The dynamic biomaterial showed an increase in stress fibers, greater YAP expression, and fluctuations in c-Jun production in SCs in comparison to stiff and soft static controls. Utilizing biomaterials to better understand the role between temporal mechanical dynamics and SC phenotype holds a very high potential for developing future PNS therapies.
Collapse
Affiliation(s)
- Alyssa Montgomery
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jennifer Westphal
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Andrew E. Bryan
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Greg M. Harris
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Cincinnati VA Medical Center, Cincinnati, OH 45220, USA
| |
Collapse
|
8
|
Aluru N, Chapman DP, W Becker K, Van Mooy BAS, Karchner SI, Stegeman JJ, Hahn ME. Developmental exposure of zebrafish to saxitoxin causes altered expression of genes associated with axonal growth. Neurotoxicology 2024; 105:303-312. [PMID: 39571800 PMCID: PMC11645194 DOI: 10.1016/j.neuro.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/19/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Saxitoxin (STX) is a potent neurotoxin naturally produced by dinoflagellates and cyanobacteria. STX inhibits voltage-gated sodium channels (VGSCs), affecting the propagation of action potentials. Consumption of seafood contaminated with STX is responsible for paralytic shellfish poisoning (PSP). Humans are among the species most sensitive to PSP; neurological symptoms of exposure range from tingling of the extremities to severe paralysis. The objective of this study was to determine the effects of STX exposure on developmental processes during early embryogenesis. This study was designed to test the hypothesis that early developmental exposure to STX would disrupt key processes, particularly those related to neural development. Zebrafish embryos were exposed to STX (24 or 48 pg) or vehicle (0.3 mM HCl) at 6 h post fertilization (hpf) via microinjection. There was no overt toxicity but starting at 36 hpf there was a temporary lack of pigmentation in STX-injected embryos, which resolved by 72 hpf. Using high performance liquid chromatography, we found that STX was retained in embryos up to 72 hpf in a dose-dependent manner. Temporal transcriptional profiling of embryos exposed to 48 pg STX per embryo revealed no differentially expressed genes (DEGs) at 24 hpf, but at 36 and 48 hpf, there were 3547 and 3356 DEGs, respectively. KEGG pathway analysis revealed significant enrichment of genes related to focal adhesion, adherens junction and regulation of actin cytoskeleton, suggesting that cell-cell and cell-extracellular matrix interactions were affected by STX. Genes affected are critical for axonal growth and the development of functional neural networks. We confirmed these findings by visualizing axonal defects in transgenic zebrafish with fluorescently labeled sensory neurons. In addition, our gene expression results suggest that STX exposure affects both canonical and noncanonical functions of VGSCs. Given the fundamental role of VGSCs in both physiology and development, these findings offer valuable insights into effects of exposure to neurotoxins.
Collapse
Affiliation(s)
- Neelakanteswar Aluru
- Biology Department and Center for Oceans and Human Health,Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA.
| | - Daniel P Chapman
- Biology Department and Center for Oceans and Human Health,Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA; Eckerd College, 4200 54th Ave S, St. Petersburg, FL 33711, USA; Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington DC 20057, USA
| | - Kevin W Becker
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA; GEOMAR Helmholtz Centre for Ocean Research Kiel, Wischhofstr. 1-3, Kiel 24148, Germany
| | - Benjamin A S Van Mooy
- Department of Marine Chemistry and Geochemistry, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA
| | - Sibel I Karchner
- Biology Department and Center for Oceans and Human Health,Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA
| | - John J Stegeman
- Biology Department and Center for Oceans and Human Health,Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA
| | - Mark E Hahn
- Biology Department and Center for Oceans and Human Health,Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA
| |
Collapse
|
9
|
Prieto-López L, Pereiro X, Vecino E. The mechanics of the retina: Müller glia role on retinal extracellular matrix and modelling. Front Med (Lausanne) 2024; 11:1393057. [PMID: 39296899 PMCID: PMC11410058 DOI: 10.3389/fmed.2024.1393057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/13/2024] [Indexed: 09/21/2024] Open
Abstract
The retina is a highly heterogeneous tissue, both cell-wise but also regarding its extracellular matrix (ECM). The stiffness of the ECM is pivotal in retinal development and maturation and has also been associated with the onset and/or progression of numerous retinal pathologies, such as glaucoma, proliferative vitreoretinopathy (PVR), age-related macular degeneration (AMD), epiretinal membrane (ERM) formation or uveitis. Nonetheless, much remains unknown about the biomechanical milieu of the retina, and specifically the role that Müller glia play as principal mechanosensors and major producers of ECM constituents. So far, new approaches need to be developed to further the knowledge in the field of retinal mechanobiology for ECM-target applications to arise. In this review, we focus on the involvement of Müller glia in shaping and altering the retinal ECM under both physiological and pathological conditions and look into various biomaterial options to more accurately replicate the impact of matrix stiffness in vitro.
Collapse
Affiliation(s)
- Laura Prieto-López
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
| | - Xandra Pereiro
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| | - Elena Vecino
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| |
Collapse
|
10
|
Cao W, Zhang Y, Li L, Liu B, Ding J, Chen X. Physical cues of scaffolds promote peripheral nerve regeneration. APPLIED PHYSICS REVIEWS 2024; 11. [DOI: 10.1063/5.0189181] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
The effective treatment of long-gap peripheral nerve injury (PNI) remains a challenge in clinical settings. The autograft, the gold standard for the long-gap PNI therapy, has several limitations, including a limited supply of donor nerve, size mismatch between the donor and recipient sites, functional loss at the donor site, neuroma formation, and the requirement for two operations. With the increasing abundance of biocompatible materials with adjustable structures and properties, tissue engineering provides a promising avenue for bridging peripheral nerve gaps and addressing the above issues of autograft. The physical cues provided by tissue engineering scaffolds, essential for regulating the neural cell fate and microenvironments, have received considerable research attention. This review elaborates on three major physical cues of tissue engineering scaffolds for peripheral nerve regeneration: topological structure, mechanical support, and electrical stimulation. These three aspects are analogs to Lego bricks, wherein different combinations result in diverse functions. Innovative and more effective bricks, along with multi-level and all-around integration, are expected to provide new advances in tissue engineering for peripheral nerve generation.
Collapse
Affiliation(s)
- Wanqing Cao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences 1 , 5625 Renmin Street, Changchun 130022, People's Republic of China
- School of Applied Chemistry and Engineering, University of Science and Technology of China 2 , 96 Jinzhai Road, Hefei 230026, People's Republic of China
| | - Ye Zhang
- National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Center, Collaborative Innovation Center of Advanced Microstructures, College of Engineering and Applied Sciences, Nanjing University 3 , 163 Xianlin Avenue, Nanjing 210023, People's Republic of China
| | - Luhe Li
- National Laboratory of Solid State Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Chemistry and Biomedicine Innovation Center, Collaborative Innovation Center of Advanced Microstructures, College of Engineering and Applied Sciences, Nanjing University 3 , 163 Xianlin Avenue, Nanjing 210023, People's Republic of China
| | - Bin Liu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University 4 , 1 Xinmin Street, Changchun 130061, People's Republic of China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences 1 , 5625 Renmin Street, Changchun 130022, People's Republic of China
- School of Applied Chemistry and Engineering, University of Science and Technology of China 2 , 96 Jinzhai Road, Hefei 230026, People's Republic of China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences 1 , 5625 Renmin Street, Changchun 130022, People's Republic of China
- School of Applied Chemistry and Engineering, University of Science and Technology of China 2 , 96 Jinzhai Road, Hefei 230026, People's Republic of China
| |
Collapse
|
11
|
Muratori L, Crosio A, Ronchi G, Molinaro D, Tos P, Lovati AB, Raimondo S. Exploring an innovative decellularization protocol for porcine nerve grafts: a translational approach to peripheral nerve repair. Front Neuroanat 2024; 18:1380520. [PMID: 38567289 PMCID: PMC10985228 DOI: 10.3389/fnana.2024.1380520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Peripheral nerves are frequently affected by lesions caused by traumatic or iatrogenic damages, resulting in loss of motor and sensory function, crucial in orthopedic outcomes and with a significant impact on patients' quality of life. Many strategies have been proposed over years to repair nerve injuries with substance loss, to achieve musculoskeletal reinnervation and functional recovery. Allograft have been tested as an alternative to the gold standard, the autograft technique, but nerves from donors frequently cause immunogenic response. For this reason, several studies are focusing to find the best way to decellularize nerves preserving either the extracellular matrix, either the basal lamina, as the key elements used by Schwann cells and axons during the regenerative process. Methods This study focuses on a novel decellularization protocol for porcine nerves, aimed at reducing immunogenicity while preserving essential elements like the extracellular matrix and basal lamina, vital for nerve regeneration. To investigate the efficacy of the decellularization protocol to remove immunogenic cellular components of the nerve tissue and to preserve the basal lamina and extracellular matrix, morphological analysis was performed through Masson's Trichrome staining, immunofluorescence, high resolution light microscopy and transmission electron microscopy. Decellularized porcine nerve graft were then employed in vivo to repair a rat median nerve lesion. Morphological analysis was also used to study the ability of the porcine decellularized graft to support the nerve regeneration. Results and Discussion The decellularization method was effective in preparing porcine superficial peroneal nerves for grafting as evidenced by the removal of immunogenic components and preservation of the ECM. Morphological analysis demonstrated that four weeks after injury, regenerating fibers colonized the graft suggesting a promising use to repair severe nerve lesions. The idea of using a porcine nerve graft arises from a translational perspective. This approach offers a promising direction in the orthopedic field for nerve repair, especially in severe cases where conventional methods are limited.
Collapse
Affiliation(s)
- Luisa Muratori
- Department of Clinical and Biological Sciences, Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Turin, Italy
| | - Alessandro Crosio
- Department of Clinical and Biological Sciences, Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Turin, Italy
- UOC Traumatology-Reconstructive Microsurgery, Department of Orthopedics and Traumatology, CTO Hospital, Turin, Italy
| | - Giulia Ronchi
- Department of Clinical and Biological Sciences, Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Turin, Italy
| | - Debora Molinaro
- Department of Clinical and Biological Sciences, Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Turin, Italy
| | - Pierluigi Tos
- Reconstructive Microsurgery and Hand Surgery Unit, ASST Pini-CTO, Milan, Italy
| | - Arianna B. Lovati
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences, Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Turin, Turin, Italy
| |
Collapse
|
12
|
Nakamura F. The Role of Mechanotransduction in Contact Inhibition of Locomotion and Proliferation. Int J Mol Sci 2024; 25:2135. [PMID: 38396812 PMCID: PMC10889191 DOI: 10.3390/ijms25042135] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Contact inhibition (CI) represents a crucial tumor-suppressive mechanism responsible for controlling the unbridled growth of cells, thus preventing the formation of cancerous tissues. CI can be further categorized into two distinct yet interrelated components: CI of locomotion (CIL) and CI of proliferation (CIP). These two components of CI have historically been viewed as separate processes, but emerging research suggests that they may be regulated by both distinct and shared pathways. Specifically, recent studies have indicated that both CIP and CIL utilize mechanotransduction pathways, a process that involves cells sensing and responding to mechanical forces. This review article describes the role of mechanotransduction in CI, shedding light on how mechanical forces regulate CIL and CIP. Emphasis is placed on filamin A (FLNA)-mediated mechanotransduction, elucidating how FLNA senses mechanical forces and translates them into crucial biochemical signals that regulate cell locomotion and proliferation. In addition to FLNA, trans-acting factors (TAFs), which are proteins or regulatory RNAs capable of directly or indirectly binding to specific DNA sequences in distant genes to regulate gene expression, emerge as sensitive players in both the mechanotransduction and signaling pathways of CI. This article presents methods for identifying these TAF proteins and profiling the associated changes in chromatin structure, offering valuable insights into CI and other biological functions mediated by mechanotransduction. Finally, it addresses unanswered research questions in these fields and delineates their possible future directions.
Collapse
Affiliation(s)
- Fumihiko Nakamura
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, China
| |
Collapse
|
13
|
Pillai EK, Franze K. Mechanics in the nervous system: From development to disease. Neuron 2024; 112:342-361. [PMID: 37967561 DOI: 10.1016/j.neuron.2023.10.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 11/17/2023]
Abstract
Physical forces are ubiquitous in biological processes across scales and diverse contexts. This review highlights the significance of mechanical forces in nervous system development, homeostasis, and disease. We provide an overview of mechanical signals present in the nervous system and delve into mechanotransduction mechanisms translating these mechanical cues into biochemical signals. During development, mechanical cues regulate a plethora of processes, including cell proliferation, differentiation, migration, network formation, and cortex folding. Forces then continue exerting their influence on physiological processes, such as neuronal activity, glial cell function, and the interplay between these different cell types. Notably, changes in tissue mechanics manifest in neurodegenerative diseases and brain tumors, potentially offering new diagnostic and therapeutic target opportunities. Understanding the role of cellular forces and tissue mechanics in nervous system physiology and pathology adds a new facet to neurobiology, shedding new light on many processes that remain incompletely understood.
Collapse
Affiliation(s)
- Eva K Pillai
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany; Developmental Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany.
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK; Institute of Medical Physics and Microtissue Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Henkestraße 91, 91052 Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, Kussmaulallee 1, 91054 Erlangen, Germany.
| |
Collapse
|
14
|
Hong J, Kirkland JM, Acheta J, Marziali LN, Beck B, Jeanette H, Bhatia U, Davis G, Herron J, Roué C, Abi-Ghanem C, Feltri ML, Zuloaga K, Bechler ME, Poitelon Y, Belin S. YAP and TAZ regulate remyelination in the central nervous system. Glia 2024; 72:156-166. [PMID: 37724047 PMCID: PMC10659087 DOI: 10.1002/glia.24467] [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: 10/21/2022] [Revised: 08/19/2023] [Accepted: 08/26/2023] [Indexed: 09/20/2023]
Abstract
Myelinating cells are sensitive to mechanical stimuli from their extracellular matrix. Ablation of YAP and TAZ mechanotransducers in Schwann cells abolishes the axon-Schwann cell recognition, myelination, and remyelination in the peripheral nervous system. It was unknown if YAP and TAZ are also required for myelination and remyelination in the central nervous system. Here we define the importance of oligodendrocyte (OL) YAP and TAZ in vivo, by specific deletion in oligodendroglial cells in adult OLs during myelin repair. Blocking YAP and TAZ expression in OL lineage cells did not affect animal viability or any major defects on OL maturation and myelination. However, using a mouse model of demyelination/remyelination, we demonstrate that YAP and TAZ modulate the capacity of OLs to remyelinate axons, particularly during the early stage of the repair process, when OL proliferation is most important. These results indicate that YAP and TAZ signaling is necessary for effective remyelination of the mouse brain.
Collapse
Affiliation(s)
- Jiayue Hong
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Jules M Kirkland
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Jenica Acheta
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Leandro N Marziali
- Institute for Myelin and Glia Exploration, Dept. Biochemistry, University at Buffalo, Buffalo, NY, 14203, USA
| | - Brianna Beck
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Haley Jeanette
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Urja Bhatia
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Grace Davis
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Jacob Herron
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Clémence Roué
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Charly Abi-Ghanem
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - M Laura Feltri
- Institute for Myelin and Glia Exploration, Dept. Biochemistry, University at Buffalo, Buffalo, NY, 14203, USA
- Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Kristen Zuloaga
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Marie E Bechler
- Department of Cell and Developmental Biology, and Department of Neuroscience and Physiology State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Yannick Poitelon
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| | - Sophie Belin
- Albany Medical College, Department of Neuroscience and Experimental Therapeutics, Albany, NY, 12208, USA
| |
Collapse
|
15
|
Hromada C, Szwarc-Hofbauer D, Quyen Nguyen M, Tomasch J, Purtscher M, Hercher D, Teuschl-Woller AH. Strain-induced bands of Büngner formation promotes axon growth in 3D tissue-engineered constructs. J Tissue Eng 2024; 15:20417314231220396. [PMID: 38249993 PMCID: PMC10798132 DOI: 10.1177/20417314231220396] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/28/2023] [Indexed: 01/23/2024] Open
Abstract
Treatment of peripheral nerve lesions remains a major challenge due to poor functional recovery; hence, ongoing research efforts strive to enhance peripheral nerve repair. In this study, we aimed to establish three-dimensional tissue-engineered bands of Büngner constructs by subjecting Schwann cells (SCs) embedded in fibrin hydrogels to mechanical stimulation. We show for the first time that the application of strain induces (i) longitudinal alignment of SCs resembling bands of Büngner, and (ii) the expression of a pronounced repair SC phenotype as evidenced by upregulation of BDNF, NGF, and p75NTR. Furthermore, we show that mechanically aligned SCs provide physical guidance for migrating axons over several millimeters in vitro in a co-culture model with rat dorsal root ganglion explants. Consequently, these constructs hold great therapeutic potential for transplantation into patients and might also provide a physiologically relevant in vitro peripheral nerve model for drug screening or investigation of pathologic or regenerative processes.
Collapse
Affiliation(s)
- Carina Hromada
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Dorota Szwarc-Hofbauer
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Mai Quyen Nguyen
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
| | - Janine Tomasch
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Michaela Purtscher
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - David Hercher
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
| | - Andreas Herbert Teuschl-Woller
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
16
|
Carnicer-Lombarte A, Barone DG, Wronowski F, Malliaras GG, Fawcett JW, Franze K. Regenerative capacity of neural tissue scales with changes in tissue mechanics post injury. Biomaterials 2023; 303:122393. [PMID: 37977006 DOI: 10.1016/j.biomaterials.2023.122393] [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: 12/19/2022] [Revised: 10/23/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Spinal cord injuries have devastating consequences for humans, as mammalian neurons of the central nervous system (CNS) cannot regenerate. In the peripheral nervous system (PNS), however, neurons may regenerate to restore lost function following injury. While mammalian CNS tissue softens after injury, how PNS tissue mechanics changes in response to mechanical trauma is currently poorly understood. Here we characterised mechanical rat nerve tissue properties before and after in vivo crush and transection injuries using atomic force microscopy-based indentation measurements. Unlike CNS tissue, PNS tissue significantly stiffened after both types of tissue damage. This nerve tissue stiffening strongly correlated with an increase in collagen I levels. Schwann cells, which crucially support PNS regeneration, became more motile and proliferative on stiffer substrates in vitro, suggesting that changes in tissue stiffness may play a key role in facilitating or impeding nervous system regeneration.
Collapse
Affiliation(s)
- Alejandro Carnicer-Lombarte
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK; Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, CB3 0FA, UK.
| | - Damiano G Barone
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Filip Wronowski
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, CB3 0FA, UK
| | - George G Malliaras
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, CB3 0FA, UK
| | - James W Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0PY, UK; Centre for Reconstructive Neuroscience, Institute for Experimental Medicine CAS, Prague, Czech Republic
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, UK; Institute of Medical Physics and Micro-Tissue Engineering, Friedrich-Alexander Universität Erlangen-Nürnberg, 91052, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91054, Erlangen, Germany.
| |
Collapse
|
17
|
Thede AT, Tang JD, Cocker CE, Harold LJ, Amelung CD, Kittel AR, Taylor PA, Lampe KJ. Effects of Cell-Adhesive Ligand Presentation on Pentapeptide Supramolecular Assembly and Gelation: Simulations and Experiments. Cells Tissues Organs 2023; 212:468-483. [PMID: 37751723 DOI: 10.1159/000534280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 09/21/2023] [Indexed: 09/28/2023] Open
Abstract
The extracellular matrix (ECM) is a complex, hierarchical material containing structural and bioactive components. This complexity makes decoupling the effects of biomechanical properties and cell-matrix interactions difficult, especially when studying cellular processes in a 3D environment. Matrix mechanics and cell adhesion are both known regulators of specific cellular processes such as stem cell proliferation and differentiation. However, more information is required about how such variables impact various neural lineages that could, upon transplantation, therapeutically improve neural function after a central nervous system injury or disease. Rapidly Assembling Pentapeptides for Injectable Delivery (RAPID) hydrogels are one biomaterial approach to meet these goals, consisting of a family of peptide sequences that assemble into physical hydrogels in physiological media. In this study, we studied our previously reported supramolecularly-assembling RAPID hydrogels functionalized with the ECM-derived cell-adhesive peptide ligands RGD, IKVAV, and YIGSR. Using molecular dynamics simulations and experimental rheology, we demonstrated that these integrin-binding ligands at physiological concentrations (3-12 mm) did not impact the assembly of the KYFIL peptide system. In simulations, molecular measures of assembly such as hydrogen bonding and pi-pi interactions appeared unaffected by cell-adhesion sequence or concentration. Visualizations of clustering and analysis of solvent-accessible surface area indicated that the integrin-binding domains remained exposed. KYFIL or AYFIL hydrogels containing 3 mm of integrin-binding domains resulted in mechanical properties consistent with their non-functionalized equivalents. This strategy of doping RAPID gels with cell-adhesion sequences allows for the precise tuning of peptide ligand concentration, independent of the rheological properties. The controllability of the RAPID hydrogel system provides an opportunity to investigate the effect of integrin-binding interactions on encapsulated neural cells to discern how hydrogel microenvironment impacts growth, maturation, or differentiation.
Collapse
Affiliation(s)
- Andrew T Thede
- University of Virginia Biomedical Engineering, Charlottesville, Virginia, USA
| | - James D Tang
- University of Virginia Chemical Engineering, Charlottesville, Virginia, USA
| | - Clare E Cocker
- University of Virginia Chemical Engineering, Charlottesville, Virginia, USA
| | - Liza J Harold
- University of Virginia Biomedical Engineering, Charlottesville, Virginia, USA
| | - Connor D Amelung
- University of Virginia Biomedical Engineering, Charlottesville, Virginia, USA
| | - Anna R Kittel
- University of Virginia Biomedical Engineering, Charlottesville, Virginia, USA
| | - Phillip A Taylor
- University of Virginia Chemical Engineering, Charlottesville, Virginia, USA
| | | |
Collapse
|
18
|
Guan Y, Ren Z, Yang B, Xu W, Wu W, Li X, Zhang T, Li D, Chen S, Bai J, Song X, Jia Z, Xiong X, He S, Li C, Meng F, Wu T, Zhang J, Liu X, Meng H, Peng J, Wang Y. Dual-bionic regenerative microenvironment for peripheral nerve repair. Bioact Mater 2023; 26:370-386. [PMID: 36942011 PMCID: PMC10024190 DOI: 10.1016/j.bioactmat.2023.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 03/18/2023] Open
Abstract
Autologous nerve grafting serves is considered the gold standard treatment for peripheral nerve defects; however, limited availability and donor area destruction restrict its widespread clinical application. Although the performance of allogeneic decellularized nerve implants has been explored, challenges such as insufficient human donors have been a major drawback to its clinical use. Tissue-engineered neural regeneration materials have been developed over the years, and researchers have explored strategies to mimic the peripheral neural microenvironment during the design of nerve catheter grafts, namely the extracellular matrix (ECM), which includes mechanical, physical, and biochemical signals that support nerve regeneration. In this study, polycaprolactone/silk fibroin (PCL/SF)-aligned electrospun material was modified with ECM derived from human umbilical cord mesenchymal stem cells (hUMSCs), and a dual-bionic nerve regeneration material was successfully fabricated. The results indicated that the developed biomimetic material had excellent biological properties, providing sufficient anchorage for Schwann cells and subsequent axon regeneration and angiogenesis processes. Moreover, the dual-bionic material exerted a similar effect to that of autologous nerve transplantation in bridging peripheral nerve defects in rats. In conclusion, this study provides a new concept for designing neural regeneration materials, and the prepared dual-bionic repair materials have excellent auxiliary regenerative ability and further preclinical testing is warranted to evaluate its clinical application potential.
Collapse
Affiliation(s)
- Yanjun Guan
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province, 226007, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Zhiqi Ren
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Boyao Yang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Wenjing Xu
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Wenjun Wu
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Xiangling Li
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Tieyuan Zhang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Dongdong Li
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Shengfeng Chen
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Jun Bai
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Xiangyu Song
- Hebei North University, Zhangjiakou, 075051, PR China
| | - Zhibo Jia
- Hebei North University, Zhangjiakou, 075051, PR China
| | - Xing Xiong
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Songlin He
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- School of Medicine, Nankai University, Tianjin, 300071, PR China
| | - Chaochao Li
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Fanqi Meng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Tong Wu
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Jian Zhang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Xiuzhi Liu
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Haoye Meng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Jiang Peng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province, 226007, PR China
- Corresponding author. Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China.
| | - Yu Wang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province, 226007, PR China
- Corresponding author. Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China.
| |
Collapse
|
19
|
Pawelec KM, Hix JML, Shapiro EM. Functional attachment of primary neurons and glia on radiopaque implantable biomaterials for nerve repair. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 52:102692. [PMID: 37328139 PMCID: PMC10527527 DOI: 10.1016/j.nano.2023.102692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/05/2023] [Accepted: 05/24/2023] [Indexed: 06/18/2023]
Abstract
Repairing peripheral nerve injuries remains a challenge, even with use of auxiliary implantable biomaterial conduits. After implantation the location or function of polymeric devices cannot be assessed via clinical imaging modalities. Adding nanoparticle contrast agents into polymers can introduce radiopacity enabling imaging using computed tomography. Radiopacity must be balanced with changes in material properties impacting device function. In this study radiopaque composites were made from polycaprolactone and poly(lactide-co-glycolide) 50:50 and 85:15 with 0-40 wt% tantalum oxide (TaOx) nanoparticles. To achieve radiopacity, ≥5 wt% TaOx was required, with ≥20 wt% TaOx reducing mechanical properties and causing nanoscale surface roughness. Composite films facilitated nerve regeneration in an in vitro co-culture of adult glia and neurons, measured by markers for myelination. The ability of radiopaque films to support regeneration was driven by the properties of the polymer, with 5-20 wt% TaOx balancing imaging functionality with biological response and proving that in situ monitoring is feasible.
Collapse
Affiliation(s)
- Kendell M Pawelec
- Michigan State University, Dept Radiology, East Lansing, MI 48823, United States of America.
| | - Jeremy M L Hix
- Michigan State University, Dept Radiology, East Lansing, MI 48823, United States of America; Michigan State University, Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI 48823, United States of America
| | - Erik M Shapiro
- Michigan State University, Dept Radiology, East Lansing, MI 48823, United States of America.
| |
Collapse
|
20
|
Siddiqui AM, Thiele F, Stewart RN, Rangnick S, Weiss GJ, Chen BK, Silvernail JL, Strickland T, Nesbitt JJ, Lim K, Schwarzbauer JE, Schwartz J, Yaszemski MJ, Windebank AJ, Madigan NN. Open-Spaced Ridged Hydrogel Scaffolds Containing TiO 2-Self-Assembled Monolayer of Phosphonates Promote Regeneration and Recovery Following Spinal Cord Injury. Int J Mol Sci 2023; 24:10250. [PMID: 37373396 DOI: 10.3390/ijms241210250] [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: 03/06/2023] [Revised: 05/31/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The spinal cord has a poor ability to regenerate after an injury, which may be due to cell loss, cyst formation, inflammation, and scarring. A promising approach to treating a spinal cord injury (SCI) is the use of biomaterials. We have developed a novel hydrogel scaffold fabricated from oligo(poly(ethylene glycol) fumarate) (OPF) as a 0.08 mm thick sheet containing polymer ridges and a cell-attractive surface on the other side. When the cells are cultured on OPF via chemical patterning, the cells attach, align, and deposit ECM along the direction of the pattern. Animals implanted with the rolled scaffold sheets had greater hindlimb recovery compared to that of the multichannel scaffold control, which is likely due to the greater number of axons growing across it. The immune cell number (microglia or hemopoietic cells: 50-120 cells/mm2 in all conditions), scarring (5-10% in all conditions), and ECM deposits (Laminin or Fibronectin: approximately 10-20% in all conditions) were equal in all conditions. Overall, the results suggest that the scaffold sheets promote axon outgrowth that can be guided across the scaffold, thereby promoting hindlimb recovery. This study provides a hydrogel scaffold construct that can be used in vitro for cell characterization or in vivo for future neuroprosthetics, devices, or cell and ECM delivery.
Collapse
Affiliation(s)
- Ahad M Siddiqui
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Frederic Thiele
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Program in Human Medicine, Paracelsus Medical Private University, 5020 Salzburg, Austria
| | - Rachel N Stewart
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Simone Rangnick
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Program in Human Medicine, Paracelsus Medical Private University, 5020 Salzburg, Austria
| | - Georgina J Weiss
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Program in Human Medicine, Paracelsus Medical Private University, 90419 Nuremberg, Germany
| | - Bingkun K Chen
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Tammy Strickland
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, H91 TK33 Galway, Ireland
| | | | - Kelly Lim
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Jeffrey Schwartz
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | | | | | | |
Collapse
|
21
|
Weickenmeier J. Exploring the multiphysics of the brain during development, aging, and in neurological diseases. BRAIN MULTIPHYSICS 2023; 4:100068. [PMID: 37476409 PMCID: PMC10358452 DOI: 10.1016/j.brain.2023.100068] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023] Open
Abstract
The human brain remains an endless source of wonder and represents an intruiging scientific frontier. Multiphysics approaches naturally lend themselves to combine our extensive knowledge about the neurobiology of aging and diseases with mechanics to better capture the multiscale behavior of the brain. Our group uses experimental methods, medical image analysis, and constitutive modeling to develop better disease models with the long-term goal to improve diagnosis, treatment, and ultimately enable prevention of many prevalent age- and disease-related brain changes. In the present perspective, we outline on-going work related to neurodevelopment, aging, and neurodegenerative disease.
Collapse
Affiliation(s)
- Johannes Weickenmeier
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States of America
| |
Collapse
|
22
|
Millesi F, Mero S, Semmler L, Rad A, Stadlmayr S, Borger A, Supper P, Haertinger M, Ploszczanski L, Windberger U, Weiss T, Naghilou A, Radtke C. Systematic Comparison of Commercial Hydrogels Revealed That a Synergy of Laminin and Strain-Stiffening Promotes Directed Migration of Neural Cells. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12678-12695. [PMID: 36876876 PMCID: PMC10020957 DOI: 10.1021/acsami.2c20040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/23/2023] [Indexed: 05/19/2023]
Abstract
Hydrogels have shown potential in replacing damaged nerve tissue, but the ideal hydrogel is yet to be found. In this study, various commercially available hydrogels were compared. Schwann cells, fibroblasts, and dorsal root ganglia neurons were seeded on the hydrogels, and their morphology, viability, proliferation, and migration were examined. Additionally, detailed analyses of the gels' rheological properties and topography were conducted. Our results demonstrate vast differences on cell elongation and directed migration on the hydrogels. Laminin was identified as the driver behind cell elongation and in combination with a porous, fibrous, and strain-stiffening matrix structure responsible for oriented cell motility. This study improves our understanding of cell-matrix interactions and thereby facilitates tailored fabrication of hydrogels in the future.
Collapse
Affiliation(s)
- Flavia Millesi
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Sascha Mero
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Lorenz Semmler
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Anda Rad
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Sarah Stadlmayr
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Anton Borger
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Paul Supper
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Maximilian Haertinger
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Leon Ploszczanski
- Institute
for Physics and Materials Science, University
of Natural Resources and Life Sciences, Vienna 1190, Austria
| | - Ursula Windberger
- Decentralized
Biomedical Facilities, Core Unit Laboratory Animal Breeding and Husbandry, Medical University Vienna, Vienna 1090, Austria
| | - Tamara Weiss
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Aida Naghilou
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
- Department
of Physical Chemistry, University of Vienna, Vienna 1090, Austria
| | - Christine Radtke
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
- Department
of Plastic, Reconstructive
and Aesthetic Surgery, Medical University
of Vienna, Vienna 1090, Austria
| |
Collapse
|
23
|
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
|
24
|
Selcen I, Prentice E, Casaccia P. The epigenetic landscape of oligodendrocyte lineage cells. Ann N Y Acad Sci 2023; 1522:24-41. [PMID: 36740586 PMCID: PMC10085863 DOI: 10.1111/nyas.14959] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The epigenetic landscape of oligodendrocyte lineage cells refers to the cell-specific modifications of DNA, chromatin, and RNA that define a unique gene expression pattern of functionally specialized cells. Here, we focus on the epigenetic changes occurring as progenitors differentiate into myelin-forming cells and respond to the local environment. First, modifications of DNA, RNA, nucleosomal histones, key principles of chromatin organization, topologically associating domains, and local remodeling will be reviewed. Then, the relationship between epigenetic modulators and RNA processing will be explored. Finally, the reciprocal relationship between the epigenome as a determinant of the mechanical properties of cell nuclei and the target of mechanotransduction will be discussed. The overall goal is to provide an interpretative key on how epigenetic changes may account for the heterogeneity of the transcriptional profiles identified in this lineage.
Collapse
Affiliation(s)
- Ipek Selcen
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, New York, New York, USA.,Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of The City University of New York, New York, New York, USA
| | - Emily Prentice
- Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of The City University of New York, New York, New York, USA.,Graduate Program in Biology, The Graduate Center of The City University of New York, New York, New York, USA
| | - Patrizia Casaccia
- Graduate Program in Biochemistry, The Graduate Center of The City University of New York, New York, New York, USA.,Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of The City University of New York, New York, New York, USA.,Graduate Program in Biology, The Graduate Center of The City University of New York, New York, New York, USA
| |
Collapse
|
25
|
Pawelec KM, Hix JM, Shapiro EM. Radiopaque Implantable Biomaterials for Nerve Repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.05.522860. [PMID: 36711915 PMCID: PMC9881907 DOI: 10.1101/2023.01.05.522860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Repairing peripheral nerve injuries remains a clinical challenge. To enhance nerve regeneration and functional recovery, the use of auxiliary implantable biomaterial conduits has become widespread. After implantation, there is currently no way to assess the location or function of polymeric biomedical devices, as they cannot be easily differentiated from surrounding tissue using clinical imaging modalities. Adding nanoparticle contrast agents into polymer matrices can introduce radiopacity and enable imaging using computed tomography (CT), but radiopacity must be balanced with changes in material properties that impact device function and biological response. In this study radiopacity was introduced to porous films of polycaprolactone (PCL) and poly(lactide-co-glycolide) (PLGA) 50:50 and 85:15 with 0-40wt% biocompatible tantalum oxide (TaO x ) nanoparticles. To achieve radiopacity, at least 5wt% TaO x was required, with ≥ 20wt% TaO x leading to reduced mechanical properties and increased nano-scale surface roughness of films. As polymers used for peripheral nerve injury devices, films facilitated nerve regeneration in an in vitro co-culture model of glia (Schwann cells) and dorsal root ganglion neurons (DRG), measured by expression markers for myelination. The ability of radiopaque films to support nerve regeneration was determined by the properties of the polymer matrix, with a range of 5-20wt% TaO x balancing both imaging functionality with biological response and proving that in situ monitoring of nerve repair devices is feasible.
Collapse
Affiliation(s)
| | - Jeremy Ml Hix
- Michigan State University, Dept Radiology, East Lansing, MI 48823
- Michigan State University, Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI 48823
| | - Erik M Shapiro
- Michigan State University, Dept Radiology, East Lansing, MI 48823
| |
Collapse
|
26
|
Castillo Ransanz L, Van Altena PFJ, Heine VM, Accardo A. Engineered cell culture microenvironments for mechanobiology studies of brain neural cells. Front Bioeng Biotechnol 2022; 10:1096054. [PMID: 36588937 PMCID: PMC9794772 DOI: 10.3389/fbioe.2022.1096054] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
The biomechanical properties of the brain microenvironment, which is composed of different neural cell types, the extracellular matrix, and blood vessels, are critical for normal brain development and neural functioning. Stiffness, viscoelasticity and spatial organization of brain tissue modulate proliferation, migration, differentiation, and cell function. However, the mechanical aspects of the neural microenvironment are largely ignored in current cell culture systems. Considering the high promises of human induced pluripotent stem cell- (iPSC-) based models for disease modelling and new treatment development, and in light of the physiological relevance of neuromechanobiological features, applications of in vitro engineered neuronal microenvironments should be explored thoroughly to develop more representative in vitro brain models. In this context, recently developed biomaterials in combination with micro- and nanofabrication techniques 1) allow investigating how mechanical properties affect neural cell development and functioning; 2) enable optimal cell microenvironment engineering strategies to advance neural cell models; and 3) provide a quantitative tool to assess changes in the neuromechanobiological properties of the brain microenvironment induced by pathology. In this review, we discuss the biological and engineering aspects involved in studying neuromechanobiology within scaffold-free and scaffold-based 2D and 3D iPSC-based brain models and approaches employing primary lineages (neural/glial), cell lines and other stem cells. Finally, we discuss future experimental directions of engineered microenvironments in neuroscience.
Collapse
Affiliation(s)
- Lucía Castillo Ransanz
- Department of Child and Adolescence Psychiatry, Amsterdam Neuroscience, Emma Children’s Hospital, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Pieter F. J. Van Altena
- Department of Precision and Microsystems Engineering, Delft University of Technology, Delft, Netherlands
| | - Vivi M. Heine
- Department of Child and Adolescence Psychiatry, Amsterdam Neuroscience, Emma Children’s Hospital, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Department of Complex Trait Genetics, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Angelo Accardo
- Department of Precision and Microsystems Engineering, Delft University of Technology, Delft, Netherlands
| |
Collapse
|
27
|
Acheta J, Bhatia U, Haley J, Hong J, Rich K, Close R, Bechler ME, Belin S, Poitelon Y. Piezo channels contribute to the regulation of myelination in Schwann cells. Glia 2022; 70:2276-2289. [PMID: 35903933 PMCID: PMC10638658 DOI: 10.1002/glia.24251] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 12/19/2022]
Abstract
Peripheral nerves and Schwann cells have to sustain constant mechanical constraints, caused by developmental growth as well as stretches associated with movements of the limbs and mechanical compressions from daily activities. In Schwann cells, signaling molecules sensitive to stiffness or stretch of the extracellular matrix, such as YAP/TAZ, have been shown to be critical for Schwann cell development and peripheral nerve regeneration. YAP/TAZ have also been suggested to contribute to tumorigenesis, neuropathic pain, and inherited disorders. Yet, the role of mechanosensitive ion channels in myelinating Schwann cells is vastly unexplored. Here we comprehensively assessed the expression of mechanosensitive ion channels in Schwann cells and identified that PIEZO1 and PIEZO2 are among the most abundant mechanosensitive ion channels expressed by Schwann cells. Using classic genetic ablation studies, we show that PIEZO1 is a transient inhibitor of radial and longitudinal myelination in Schwann cells. Contrastingly, we show that PIEZO2 may be required for myelin formation, as the absence of PIEZO2 in Schwann cells delays myelin formation. We found an epistatic relationship between PIEZO1 and PIEZO2, at both the morphological and molecular levels. Finally, we show that PIEZO1 channels affect the regulation of YAP/TAZ activation in Schwann cells. Overall, we present here the first demonstration that PIEZO1 and PIEZO2 contribute to mechanosensation in Schwann cells as well myelin development in the peripheral nervous system.
Collapse
Affiliation(s)
- Jenica Acheta
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Urja Bhatia
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Jeanette Haley
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Jiayue Hong
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Kyle Rich
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Rachel Close
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Marie E. Bechler
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| |
Collapse
|
28
|
Hörner SJ, Couturier N, Gueiber DC, Hafner M, Rudolf R. Development and In Vitro Differentiation of Schwann Cells. Cells 2022; 11:3753. [PMID: 36497014 PMCID: PMC9739763 DOI: 10.3390/cells11233753] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Schwann cells are glial cells of the peripheral nervous system. They exist in several subtypes and perform a variety of functions in nerves. Their derivation and culture in vitro are interesting for applications ranging from disease modeling to tissue engineering. Since primary human Schwann cells are challenging to obtain in large quantities, in vitro differentiation from other cell types presents an alternative. Here, we first review the current knowledge on the developmental signaling mechanisms that determine neural crest and Schwann cell differentiation in vivo. Next, an overview of studies on the in vitro differentiation of Schwann cells from multipotent stem cell sources is provided. The molecules frequently used in those protocols and their involvement in the relevant signaling pathways are put into context and discussed. Focusing on hiPSC- and hESC-based studies, different protocols are described and compared, regarding cell sources, differentiation methods, characterization of cells, and protocol efficiency. A brief insight into developments regarding the culture and differentiation of Schwann cells in 3D is given. In summary, this contribution provides an overview of the current resources and methods for the differentiation of Schwann cells, it supports the comparison and refinement of protocols and aids the choice of suitable methods for specific applications.
Collapse
Affiliation(s)
- Sarah Janice Hörner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Nathalie Couturier
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Daniele Caroline Gueiber
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Department of Electronics Engineering, Federal University of Technology Paraná, Ponta Grossa 84017-220, Brazil
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| |
Collapse
|
29
|
Reed CB, Feltri ML, Wilson ER. Peripheral glia diversity. J Anat 2022; 241:1219-1234. [PMID: 34131911 PMCID: PMC8671569 DOI: 10.1111/joa.13484] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Recent years have seen an evolving appreciation for the role of glial cells in the nervous system. As we move away from the typical neurocentric view of neuroscience, the complexity and variability of central nervous system glia is emerging, far beyond the three main subtypes: astrocytes, oligodendrocytes, and microglia. Yet the diversity of the glia found in the peripheral nervous system remains rarely discussed. In this review, we discuss the developmental origin, morphology, and function of the different populations of glia found in the peripheral nervous system, including: myelinating Schwann cells, Remak Schwann cells, repair Schwann cells, satellite glia, boundary cap-derived glia, perineurial glia, terminal Schwann cells, glia found in the skin, olfactory ensheathing cells, and enteric glia. The morphological and functional heterogeneity of glia found in the periphery reflects the diverse roles the nervous system performs throughout the body. Further, it highlights a complexity that should be appreciated and considered when it comes to a complete understanding of the peripheral nervous system in health and disease.
Collapse
Affiliation(s)
- Chelsey B. Reed
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of NeurologyJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| | - M. Laura Feltri
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of NeurologyJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
- Department of BiochemistryJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| | - Emma R. Wilson
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of BiochemistryJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| |
Collapse
|
30
|
Klymenko A, Lutz D. Melatonin signalling in Schwann cells during neuroregeneration. Front Cell Dev Biol 2022; 10:999322. [PMID: 36299487 PMCID: PMC9589221 DOI: 10.3389/fcell.2022.999322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
It has widely been thought that in the process of nerve regeneration Schwann cells populate the injury site with myelinating, non–myelinating, phagocytic, repair, and mesenchyme–like phenotypes. It is now clear that the Schwann cells modify their shape and basal lamina as to accommodate re–growing axons, at the same time clear myelin debris generated upon injury, and regulate expression of extracellular matrix proteins at and around the lesion site. Such a remarkable plasticity may follow an intrinsic functional rhythm or a systemic circadian clock matching the demands of accurate timing and precision of signalling cascades in the regenerating nervous system. Schwann cells react to changes in the external circadian clock clues and to the Zeitgeber hormone melatonin by altering their plasticity. This raises the question of whether melatonin regulates Schwann cell activity during neurorepair and if circadian control and rhythmicity of Schwann cell functions are vital aspects of neuroregeneration. Here, we have focused on different schools of thought and emerging concepts of melatonin–mediated signalling in Schwann cells underlying peripheral nerve regeneration and discuss circadian rhythmicity as a possible component of neurorepair.
Collapse
|
31
|
Smith CS, Orkwis JA, Bryan AE, Xu Z, Harris GM. The impact of physical, biochemical, and electrical signaling on Schwann cell plasticity. Eur J Cell Biol 2022; 101:151277. [PMID: 36265214 DOI: 10.1016/j.ejcb.2022.151277] [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: 09/02/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 12/14/2022] Open
Abstract
Peripheral nervous system (PNS) injuries are an ongoing health care concern. While autografts and allografts are regarded as the current clinical standard for traumatic injury, there are inherent limitations that suggest alternative remedies should be considered for therapeutic purposes. In recent years, nerve guidance conduits (NGCs) have become increasingly popular as surgical repair devices, with a multitude of various natural and synthetic biomaterials offering potential to enhance the design of conduits or supplant existing technologies entirely. From a cellular perspective, it has become increasingly evident that Schwann cells (SCs), the primary glia of the PNS, are a predominant factor mediating nerve regeneration. Thus, the development of severe nerve trauma therapies requires a deep understanding of how SCs interact with their environment, and how SC microenvironmental cues may be engineered to enhance regeneration. Here we review the most recent advancements in biomaterials development and cell stimulation strategies, with a specific focus on how the microenvironment influences the behavior of SCs and can potentially lead to functional repair. We focus on microenvironmental cues that modulate SC morphology, proliferation, migration, and differentiation to alternative phenotypes. Promotion of regenerative phenotypic responses in SCs and other non-neuronal cells that can augment the regenerative capacity of multiple biomaterials is considered along with innovations and technologies for traumatic injury.
Collapse
Affiliation(s)
- Corinne S Smith
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jacob A Orkwis
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Andrew E Bryan
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Zhenyuan Xu
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Greg M Harris
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
32
|
van Schaik PEM, Zuhorn IS, Baron W. Targeting Fibronectin to Overcome Remyelination Failure in Multiple Sclerosis: The Need for Brain- and Lesion-Targeted Drug Delivery. Int J Mol Sci 2022; 23:8418. [PMID: 35955549 PMCID: PMC9368816 DOI: 10.3390/ijms23158418] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS) is a neuroinflammatory and neurodegenerative disease with unknown etiology that can be characterized by the presence of demyelinated lesions. Prevailing treatment protocols in MS rely on the modulation of the inflammatory process but do not impact disease progression. Remyelination is an essential factor for both axonal survival and functional neurological recovery but is often insufficient. The extracellular matrix protein fibronectin contributes to the inhibitory environment created in MS lesions and likely plays a causative role in remyelination failure. The presence of the blood-brain barrier (BBB) hinders the delivery of remyelination therapeutics to lesions. Therefore, therapeutic interventions to normalize the pathogenic MS lesion environment need to be able to cross the BBB. In this review, we outline the multifaceted roles of fibronectin in MS pathogenesis and discuss promising therapeutic targets and agents to overcome fibronectin-mediated inhibition of remyelination. In addition, to pave the way for clinical use, we reflect on opportunities to deliver MS therapeutics to lesions through the utilization of nanomedicine and discuss strategies to deliver fibronectin-directed therapeutics across the BBB. The use of well-designed nanocarriers with appropriate surface functionalization to cross the BBB and target the lesion sites is recommended.
Collapse
Affiliation(s)
- Pauline E. M. van Schaik
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| | - Inge S. Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Wia Baron
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| |
Collapse
|
33
|
Velasco-Estevez M, Koch N, Klejbor I, Caratis F, Rutkowska A. Mechanoreceptor Piezo1 Is Downregulated in Multiple Sclerosis Brain and Is Involved in the Maturation and Migration of Oligodendrocytes in vitro. Front Cell Neurosci 2022; 16:914985. [PMID: 35722613 PMCID: PMC9204635 DOI: 10.3389/fncel.2022.914985] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Mechanical properties of the brain such as intracranial pressure or stiffness of the matrix play an important role in the brain’s normal physiology and pathophysiology. The physical properties are sensed by the cells through mechanoreceptors and translated into ion currents which activate multiple biochemical cascades allowing the cells to adapt and respond to changes in their microenvironment. Piezo1 is one of the first identified mechanoreceptors. It modulates various central nervous system functions such as axonal growth or activation of astrocytes. Piezo1 signaling was also shown to play a role in the pathophysiology of Alzheimer’s disease. Here, we explore the expression of the mechanoreceptor Piezo1 in human MO3.13 oligodendrocytes and human MS/non-MS patients’ brains and investigate its putative effects on oligodendrocyte proliferation, maturation, and migration. We found that Piezo1 is expressed in human oligodendrocytes and oligodendrocyte progenitor cells in the human brain and that its inhibition with GsMTx4 leads to an increment in proliferation and migration of MO3.13 oligodendrocytes. Activation of Piezo1 with Yoda-1 induced opposite effects. Further, we observed that expression of Piezo1 decreased with MO3.13 maturation in vitro. Differences in expression were also observed between healthy and multiple sclerosis brains. Remarkably, the data showed significantly lower expression of Piezo1 in the white matter in multiple sclerosis brains compared to its expression in the white matter in healthy controls. There were no differences in Piezo1 expression between the white matter plaque and healthy-appearing white matter in the multiple sclerosis brain. Taken together, we here show that Piezo1-induced signaling can be used to modulate oligodendrocyte function and that it may be an important player in the pathophysiology of multiple sclerosis.
Collapse
Affiliation(s)
- Maria Velasco-Estevez
- H12O-CNIO Hematological Malignancies Group, Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Nina Koch
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Ilona Klejbor
- Department of Anatomy, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Fionä Caratis
- Department of Anatomy and Neurobiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Rutkowska
- Department of Anatomy and Neurobiology, Medical University of Gdańsk, Gdańsk, Poland
- *Correspondence: Aleksandra Rutkowska,
| |
Collapse
|
34
|
Wang R, Zhou R, Chen Z, Gao S, Zhou F. The Glial Cells Respond to Spinal Cord Injury. Front Neurol 2022; 13:844497. [PMID: 35599739 PMCID: PMC9120539 DOI: 10.3389/fneur.2022.844497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/08/2022] [Indexed: 12/24/2022] Open
Abstract
It is been over 100 years since glial cells were discovered by Virchow. Since then, a great deal of research was carried out to specify these further roles and properties of glial cells in central nervous system (CNS). As it is well-known that glial cells, such as astrocytes, microglia, oligodendrocytes (OLs), and oligodendrocyte progenitor cells (OPCs) play an important role in supporting and enabling the effective nervous system function in CNS. After spinal cord injury (SCI), these glial cells play different roles in SCI and repair. In this review, we will discuss in detail about the role of glial cells in the healthy CNS and how they respond to SCI.
Collapse
|
35
|
Abstract
The successful transplantation of stem cells has the potential to transform regenerative medicine approaches and open promising avenues to repair, replace, and regenerate diseased, damaged, or aged tissues. However, pre-/post-transplantation issues of poor cell survival, retention, cell fate regulation, and insufficient integration with host tissues constitute significant challenges. The success of stem cell transplantation depends upon the coordinated sequence of stem cell renewal, specific lineage differentiation, assembly, and maintenance of long-term function. Advances in biomaterials can improve pre-/post-transplantation outcomes by integrating biophysiochemical cues and emulating tissue microenvironments. This review highlights leading biomaterials-based approaches for enhancing stem cell transplantation.
Collapse
Affiliation(s)
- Bhushan N Kharbikar
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Priya Mohindra
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA 94158, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA 94158, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; School of Engineering, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
36
|
Park HJ, Tsai E, Huang D, Weaver M, Frick L, Alcantara A, Moran JJ, Patzig J, Melendez-Vasquez CV, Crabtree GR, Feltri M, Svaren J, Casaccia P. ACTL6a coordinates axonal caliber recognition and myelination in the peripheral nerve. iScience 2022; 25:104132. [PMID: 35434551 PMCID: PMC9010646 DOI: 10.1016/j.isci.2022.104132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/29/2022] [Accepted: 03/17/2022] [Indexed: 11/12/2022] Open
Abstract
Cells elaborate transcriptional programs in response to external signals. In the peripheral nerves, Schwann cells (SC) sort axons of given caliber and start the process of wrapping their membrane around them. We identify Actin-like protein 6a (ACTL6a), part of SWI/SNF chromatin remodeling complex, as critical for the integration of axonal caliber recognition with the transcriptional program of myelination. Nuclear levels of ACTL6A in SC are increased by contact with large caliber axons or nanofibers, and result in the eviction of repressive histone marks to facilitate myelination. Without Actl6a the SC are unable to coordinate caliber recognition and myelin production. Peripheral nerves in knockout mice display defective radial sorting, hypo-myelination of large caliber axons, and redundant myelin around small caliber axons, resulting in a clinical motor phenotype. Overall, this suggests that ACTL6A is a key component of the machinery integrating external signals for proper myelination of the peripheral nerve.
Collapse
Affiliation(s)
- Hye-Jin Park
- Advanced Science Research Center (ASRC) at The Graduate Center of the City University of New York (CUNY), New York, NY 10031, USA
| | - Eric Tsai
- Advanced Science Research Center (ASRC) at The Graduate Center of the City University of New York (CUNY), New York, NY 10031, USA
- Graduate Program in Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dennis Huang
- Advanced Science Research Center (ASRC) at The Graduate Center of the City University of New York (CUNY), New York, NY 10031, USA
- Graduate Program in Biology, Graduate Center of CUNY, New York, NY 10016, USA
| | - Michael Weaver
- Hunter James Kelly Research Institute, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Luciana Frick
- Hunter James Kelly Research Institute, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Ace Alcantara
- Graduate Program in Biology, Graduate Center of CUNY, New York, NY 10016, USA
- Hunter College, Department of Biological Sciences, New York, NY 10065, USA
| | - John J. Moran
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin, Madison, WI 53705, USA
| | - Julia Patzig
- Advanced Science Research Center (ASRC) at The Graduate Center of the City University of New York (CUNY), New York, NY 10031, USA
| | - Carmen V. Melendez-Vasquez
- Graduate Program in Biology, Graduate Center of CUNY, New York, NY 10016, USA
- Hunter College, Department of Biological Sciences, New York, NY 10065, USA
| | - Gerald R. Crabtree
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - M.L. Feltri
- Hunter James Kelly Research Institute, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - John Svaren
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin, Madison, WI 53705, USA
| | - Patrizia Casaccia
- Advanced Science Research Center (ASRC) at The Graduate Center of the City University of New York (CUNY), New York, NY 10031, USA
- Graduate Program in Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate Program in Biology, Graduate Center of CUNY, New York, NY 10016, USA
| |
Collapse
|
37
|
Carvalho E, Morais M, Ferreira H, Silva M, Guimarães S, Pêgo A. A paradigm shift: Bioengineering meets mechanobiology towards overcoming remyelination failure. Biomaterials 2022; 283:121427. [DOI: 10.1016/j.biomaterials.2022.121427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 01/31/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022]
|
38
|
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
|
39
|
Marangon D, Caporale N, Boccazzi M, Abbracchio MP, Testa G, Lecca D. Novel in vitro Experimental Approaches to Study Myelination and Remyelination in the Central Nervous System. Front Cell Neurosci 2021; 15:748849. [PMID: 34720882 PMCID: PMC8551863 DOI: 10.3389/fncel.2021.748849] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
Myelin is the lipidic insulating structure enwrapping axons and allowing fast saltatory nerve conduction. In the central nervous system, myelin sheath is the result of the complex packaging of multilamellar extensions of oligodendrocyte (OL) membranes. Before reaching myelinating capabilities, OLs undergo a very precise program of differentiation and maturation that starts from OL precursor cells (OPCs). In the last 20 years, the biology of OPCs and their behavior under pathological conditions have been studied through several experimental models. When co-cultured with neurons, OPCs undergo terminal maturation and produce myelin tracts around axons, allowing to investigate myelination in response to exogenous stimuli in a very simple in vitro system. On the other hand, in vivo models more closely reproducing some of the features of human pathophysiology enabled to assess the consequences of demyelination and the molecular mechanisms of remyelination, and they are often used to validate the effect of pharmacological agents. However, they are very complex, and not suitable for large scale drug discovery screening. Recent advances in cell reprogramming, biophysics and bioengineering have allowed impressive improvements in the methodological approaches to study brain physiology and myelination. Rat and mouse OPCs can be replaced by human OPCs obtained by induced pluripotent stem cells (iPSCs) derived from healthy or diseased individuals, thus offering unprecedented possibilities for personalized disease modeling and treatment. OPCs and neural cells can be also artificially assembled, using 3D-printed culture chambers and biomaterial scaffolds, which allow modeling cell-to-cell interactions in a highly controlled manner. Interestingly, scaffold stiffness can be adopted to reproduce the mechanosensory properties assumed by tissues in physiological or pathological conditions. Moreover, the recent development of iPSC-derived 3D brain cultures, called organoids, has made it possible to study key aspects of embryonic brain development, such as neuronal differentiation, maturation and network formation in temporal dynamics that are inaccessible to traditional in vitro cultures. Despite the huge potential of organoids, their application to myelination studies is still in its infancy. In this review, we shall summarize the novel most relevant experimental approaches and their implications for the identification of remyelinating agents for human diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nicolò Caporale
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Human Technopole, Milan, Italy
| | - Marta Boccazzi
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Maria P. Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Testa
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Human Technopole, Milan, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
40
|
Low-intensity extracorporeal shock wave therapy promotes recovery of sciatic nerve injury and the role of mechanical sensitive YAP/TAZ signaling pathway for nerve regeneration. Chin Med J (Engl) 2021; 134:2710-2720. [PMID: 34845995 PMCID: PMC8631414 DOI: 10.1097/cm9.0000000000001431] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Background: Histological and functional recovery after peripheral nerve injury (PNI) is of significant clinical value as delayed surgical repair and longer distances to innervate terminal organs may account for poor outcomes. Low-intensity extracorporeal shock wave therapy (LiESWT) has already been proven to be beneficial for injured tissue recovery on various pathological conditions. The objective of this study was to explore the potential effect and mechanism of LiESWT on PNI recovery. Methods: In this project, we explored LiESWT's role using an animal model of sciatic nerve injury (SNI). Shockwave was delivered to the region of the SNI site with a special probe at 3 Hz, 500 shocks each time, and 3 times a week for 3 weeks. Rat Schwann cells (SCs) and rat perineurial fibroblasts (PNFs) cells, the two main compositional cell types in peripheral nerve tissue, were cultured in vitro, and LiESWT was applied through the cultured dish to the adherent cells. Tissues and cell cultures were harvested at corresponding time points for a reverse transcription-polymerase chain reaction, Western blotting, and immunofluorescence staining. Multiple groups were compared by using one-way analysis of variance followed by the Tukey-Kramer test for post hoc comparisons. Results: LiESWT treatment promoted the functional recovery of lower extremities with SNI. More nerve fibers and myelin sheath were found after LiESWT treatment associated with local upregulation of mechanical sensitive yes-associated protein (YAP)/transcriptional co-activator with a PDZ-binding domain (TAZ) signaling pathway. In vitro results showed that SCs were more sensitive to LiESWT than PNFs. LiESWT promoted SCs activation with more expression of p75 (a SCs dedifferentiation marker) and Ki67 (a SCs proliferation marker). The SCs activation process was dependent on the intact YAP/TAZ signaling pathway as knockdown of TAZ by TAZ small interfering RNA significantly attenuated this process. Conclusion: The LiESWT mechanical signal perception and YAP/TAZ upregulation in SCs might be one of the underlying mechanisms for SCs activation and injured nerve axon regeneration.
Collapse
|
41
|
Linka K, Reiter N, Würges J, Schicht M, Bräuer L, Cyron CJ, Paulsen F, Budday S. Unraveling the Local Relation Between Tissue Composition and Human Brain Mechanics Through Machine Learning. Front Bioeng Biotechnol 2021; 9:704738. [PMID: 34485258 PMCID: PMC8415910 DOI: 10.3389/fbioe.2021.704738] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/28/2021] [Indexed: 11/13/2022] Open
Abstract
The regional mechanical properties of brain tissue are not only key in the context of brain injury and its vulnerability towards mechanical loads, but also affect the behavior and functionality of brain cells. Due to the extremely soft nature of brain tissue, its mechanical characterization is challenging. The response to loading depends on length and time scales and is characterized by nonlinearity, compression-tension asymmetry, conditioning, and stress relaxation. In addition, the regional heterogeneity-both in mechanics and microstructure-complicates the comprehensive understanding of local tissue properties and its relation to the underlying microstructure. Here, we combine large-strain biomechanical tests with enzyme-linked immunosorbent assays (ELISA) and develop an extended type of constitutive artificial neural networks (CANNs) that can account for viscoelastic effects. We show that our viscoelastic constitutive artificial neural network is able to describe the tissue response in different brain regions and quantify the relevance of different cellular and extracellular components for time-independent (nonlinearity, compression-tension-asymmetry) and time-dependent (hysteresis, conditioning, stress relaxation) tissue mechanics, respectively. Our results suggest that the content of the extracellular matrix protein fibronectin is highly relevant for both the quasi-elastic behavior and viscoelastic effects of brain tissue. While the quasi-elastic response seems to be largely controlled by extracellular matrix proteins from the basement membrane, cellular components have a higher relevance for the viscoelastic response. Our findings advance our understanding of microstructure - mechanics relations in human brain tissue and are valuable to further advance predictive material models for finite element simulations or to design biomaterials for tissue engineering and 3D printing applications.
Collapse
Affiliation(s)
- Kevin Linka
- Institute of Continuum and Material Mechanics, Hamburg University of Technology, Hamburg, Germany
| | - Nina Reiter
- Institute of Applied Mechanics, Department Mechanical Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Jasmin Würges
- Institute of Applied Mechanics, Department Mechanical Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Schicht
- Institute of Functional and Clinical Anatomy, Faculty of Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Lars Bräuer
- Institute of Functional and Clinical Anatomy, Faculty of Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Christian J Cyron
- Institute of Continuum and Material Mechanics, Hamburg University of Technology, Hamburg, Germany.,Institute of Material Systems Modeling, Helmholtz-Zentrum Hereon, Geesthacht, Germany
| | - Friedrich Paulsen
- Institute of Functional and Clinical Anatomy, Faculty of Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany.,Department of Operative Surgery and Topographic Anatomy, Sechenov University, Moscow, Russia
| | - Silvia Budday
- Institute of Applied Mechanics, Department Mechanical Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
42
|
Ghorbani S, Yong VW. The extracellular matrix as modifier of neuroinflammation and remyelination in multiple sclerosis. Brain 2021; 144:1958-1973. [PMID: 33889940 PMCID: PMC8370400 DOI: 10.1093/brain/awab059] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Remyelination failure contributes to axonal loss and progression of disability in multiple sclerosis. The failed repair process could be due to ongoing toxic neuroinflammation and to an inhibitory lesion microenvironment that prevents recruitment and/or differentiation of oligodendrocyte progenitor cells into myelin-forming oligodendrocytes. The extracellular matrix molecules deposited into lesions provide both an altered microenvironment that inhibits oligodendrocyte progenitor cells, and a fuel that exacerbates inflammatory responses within lesions. In this review, we discuss the extracellular matrix and where its molecules are normally distributed in an uninjured adult brain, specifically at the basement membranes of cerebral vessels, in perineuronal nets that surround the soma of certain populations of neurons, and in interstitial matrix between neural cells. We then highlight the deposition of different extracellular matrix members in multiple sclerosis lesions, including chondroitin sulphate proteoglycans, collagens, laminins, fibronectin, fibrinogen, thrombospondin and others. We consider reasons behind changes in extracellular matrix components in multiple sclerosis lesions, mainly due to deposition by cells such as reactive astrocytes and microglia/macrophages. We next discuss the consequences of an altered extracellular matrix in multiple sclerosis lesions. Besides impairing oligodendrocyte recruitment, many of the extracellular matrix components elevated in multiple sclerosis lesions are pro-inflammatory and they enhance inflammatory processes through several mechanisms. However, molecules such as thrombospondin-1 may counter inflammatory processes, and laminins appear to favour repair. Overall, we emphasize the crosstalk between the extracellular matrix, immune responses and remyelination in modulating lesions for recovery or worsening. Finally, we review potential therapeutic approaches to target extracellular matrix components to reduce detrimental neuroinflammation and to promote recruitment and maturation of oligodendrocyte lineage cells to enhance remyelination.
Collapse
Affiliation(s)
- Samira Ghorbani
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
43
|
Dansu DK, Sauma S, Casaccia P. Oligodendrocyte progenitors as environmental biosensors. Semin Cell Dev Biol 2021; 116:38-44. [PMID: 33092959 PMCID: PMC8053729 DOI: 10.1016/j.semcdb.2020.09.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 01/10/2023]
Abstract
The past decade has seen an important revision of the traditional concept of the role and function of glial cells. From "passive support" for neurons, oligodendrocyte lineage cells are now recognized as metabolic exchangers with neurons, a cellular interface with blood vessels and responders to gut-derived metabolites or changes in the social environment. In the developing brain, the differentiation of neonatal oligodendrocyte progenitors (nOPCs) is required for normal brain function. In adulthood, the differentiation of adult OPCs (aOPCs) serves an important role in learning, behavioral adaptation and response to myelin injury. Here, we propose the concept of OPCs as environmental biosensors, which "sense" chemical and physical stimuli over time and adjust to the new challenges by modifying their epigenome and consequent transcriptome. Because epigenetics defines the ability of the cell to "adapt" gene expression to changes in the environment, we propose a model of OPC differentiation resulting from time-dependent changes of the epigenomic landscape in response to declining mitogens, raising hormone levels, neuronal activity, changes in space constraints or stiffness of the extracellular matrix. We propose that the intrinsically different functional properties of aOPCs compared to nOPCs result from the accrual of "epigenetic memories" of distinct events, which are "recorded" in the nuclei of OPCs as histone and DNA marks, defining a "unique epigenomic landscape" over time.
Collapse
Affiliation(s)
- David K Dansu
- Graduate Program in Biochemistry, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA
| | - Sami Sauma
- Graduate Program in Biology, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA
| | - Patrizia Casaccia
- Graduate Program in Biochemistry, Graduate Center of the City University of New York, New York, NY, USA; Graduate Program in Biology, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA.
| |
Collapse
|
44
|
Marinval N, Chew SY. Mechanotransduction assays for neural regeneration strategies: A focus on glial cells. APL Bioeng 2021; 5:021505. [PMID: 33948526 PMCID: PMC8088332 DOI: 10.1063/5.0037814] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/19/2021] [Indexed: 01/22/2023] Open
Abstract
Glial cells are mechanosensitive, and thus, engineered systems have taken a step forward to design mechanotransduction platforms in order to impart diverse mechanical stresses to cells. Mechanical strain encountered in the central nervous system can arise from diverse mechanisms, such as tissue reorganization, fluid flow, and axon growth, as well as pathological events including axon swelling or mechanical trauma. Biomechanical relevance of the in vitro mechanical testing requires to be placed in line with the physiological and mechanical changes in central nervous tissues that occur during the progression of neurodegenerative diseases. Mechanotransduction signaling utilized by glial cells and the recent approaches intended to model altered microenvironment adapted to pathological context are discussed in this review. New insights in systems merging substrate's stiffness and topography should be considered for further glial mechanotransduction studies, while testing platforms for drug discoveries promise great advancements in pharmacotherapy. Potential leads and strategies for clinical outcomes are expected to be developed following the exploration of these glial mechanosensitive signaling pathways.
Collapse
Affiliation(s)
- Nicolas Marinval
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459
| | - Sing Yian Chew
- Author to whom correspondence should be addressed: . Tel.: +65 6316 8812. Fax: +65 6794 7553
| |
Collapse
|
45
|
Powell R, Eleftheriadou D, Kellaway S, Phillips JB. Natural Biomaterials as Instructive Engineered Microenvironments That Direct Cellular Function in Peripheral Nerve Tissue Engineering. Front Bioeng Biotechnol 2021; 9:674473. [PMID: 34113607 PMCID: PMC8185204 DOI: 10.3389/fbioe.2021.674473] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/16/2021] [Indexed: 12/25/2022] Open
Abstract
Nerve tissue function and regeneration depend on precise and well-synchronised spatial and temporal control of biological, physical, and chemotactic cues, which are provided by cellular components and the surrounding extracellular matrix. Therefore, natural biomaterials currently used in peripheral nerve tissue engineering are selected on the basis that they can act as instructive extracellular microenvironments. Despite emerging knowledge regarding cell-matrix interactions, the exact mechanisms through which these biomaterials alter the behaviour of the host and implanted cells, including neurons, Schwann cells and immune cells, remain largely unclear. Here, we review some of the physical processes by which natural biomaterials mimic the function of the extracellular matrix and regulate cellular behaviour. We also highlight some representative cases of controllable cell microenvironments developed by combining cell biology and tissue engineering principles.
Collapse
Affiliation(s)
- Rebecca Powell
- UCL Centre for Nerve Engineering, University College London, London, United Kingdom.,Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| | - Despoina Eleftheriadou
- UCL Centre for Nerve Engineering, University College London, London, United Kingdom.,Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom.,Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Simon Kellaway
- UCL Centre for Nerve Engineering, University College London, London, United Kingdom.,Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| | - James B Phillips
- UCL Centre for Nerve Engineering, University College London, London, United Kingdom.,Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| |
Collapse
|
46
|
Xu Z, Orkwis JA, Harris GM. Cell Shape and Matrix Stiffness Impact Schwann Cell Plasticity via YAP/TAZ and Rho GTPases. Int J Mol Sci 2021; 22:ijms22094821. [PMID: 34062912 PMCID: PMC8124465 DOI: 10.3390/ijms22094821] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/01/2023] Open
Abstract
Schwann cells (SCs) are a highly plastic cell type capable of undergoing phenotypic changes following injury or disease. SCs are able to upregulate genes associated with nerve regeneration and ultimately achieve functional recovery. During the regeneration process, the extracellular matrix (ECM) and cell morphology play a cooperative, critical role in regulating SCs, and therefore highly impact nerve regeneration outcomes. However, the roles of the ECM and mechanotransduction relating to SC phenotype are largely unknown. Here, we describe the role that matrix stiffness and cell morphology play in SC phenotype specification via known mechanotransducers YAP/TAZ and RhoA. Using engineered microenvironments to precisely control ECM stiffness, cell shape, and cell spreading, we show that ECM stiffness and SC spreading downregulated SC regenerative associated proteins by the activation of RhoA and YAP/TAZ. Additionally, cell elongation promoted a distinct SC regenerative capacity by the upregulation of Rac1/MKK7/JNK, both necessary for the ECM and morphology changes found during nerve regeneration. These results confirm the role of ECM signaling in peripheral nerve regeneration as well as provide insight to the design of future biomaterials and cellular therapies for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Zhenyuan Xu
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (Z.X.); (J.A.O.)
| | - Jacob A. Orkwis
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (Z.X.); (J.A.O.)
| | - Greg M. Harris
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (Z.X.); (J.A.O.)
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- Correspondence: ; Tel.: +1-(513)-556-4167
| |
Collapse
|
47
|
Cayre M, Falque M, Mercier O, Magalon K, Durbec P. Myelin Repair: From Animal Models to Humans. Front Cell Neurosci 2021; 15:604865. [PMID: 33935649 PMCID: PMC8079744 DOI: 10.3389/fncel.2021.604865] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/15/2021] [Indexed: 12/20/2022] Open
Abstract
It is widely thought that brain repair does not occur, but myelin regeneration provides clear evidence to the contrary. Spontaneous remyelination may occur after injury or in multiple sclerosis (MS). However, the efficiency of remyelination varies considerably between MS patients and between the lesions of each patient. Myelin repair is essential for optimal functional recovery, so a profound understanding of the cells and mechanisms involved in this process is required for the development of new therapeutic strategies. In this review, we describe how animal models and modern cell tracing and imaging methods have helped to identify the cell types involved in myelin regeneration. In addition to the oligodendrocyte progenitor cells identified in the 1990s as the principal source of remyelinating cells in the central nervous system (CNS), other cell populations, including subventricular zone-derived neural progenitors, Schwann cells, and even spared mature oligodendrocytes, have more recently emerged as potential contributors to CNS remyelination. We will also highlight the conditions known to limit endogenous repair, such as aging, chronic inflammation, and the production of extracellular matrix proteins, and the role of astrocytes and microglia in these processes. Finally, we will present the discrepancies between observations in humans and in rodents, discussing the relationship of findings in experimental models to myelin repair in humans. These considerations are particularly important from a therapeutic standpoint.
Collapse
Affiliation(s)
- Myriam Cayre
- Aix Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut de Biologie du Développement de Marseille (IBDM-UMR 7288), Marseille, France
| | | | | | | | | |
Collapse
|
48
|
Fornaro M, Marcus D, Rattin J, Goral J. Dynamic Environmental Physical Cues Activate Mechanosensitive Responses in the Repair Schwann Cell Phenotype. Cells 2021; 10:cells10020425. [PMID: 33671410 PMCID: PMC7922665 DOI: 10.3390/cells10020425] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 01/10/2023] Open
Abstract
Schwann cells plastically change in response to nerve injury to become a newly reconfigured repair phenotype. This cell is equipped to sense and interact with the evolving and unusual physical conditions characterizing the injured nerve environment and activate intracellular adaptive reprogramming as a consequence of external stimuli. Summarizing the literature contributions on this matter, this review is aimed at highlighting the importance of the environmental cues of the regenerating nerve as key factors to induce morphological and functional changes in the Schwann cell population. We identified four different microenvironments characterized by physical cues the Schwann cells sense via interposition of the extracellular matrix. We discussed how the physical cues of the microenvironment initiate changes in Schwann cell behavior, from wrapping the axon to becoming a multifunctional denervated repair cell and back to reestablishing contact with regenerated axons.
Collapse
Affiliation(s)
- Michele Fornaro
- Department of Anatomy, College of Graduate Studies (CGS), Midwestern University, Downers Grove, IL 60515, USA;
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
- Correspondence: ; Tel.: +001-630-515-6055
| | - Dominic Marcus
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
| | - Jacob Rattin
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
| | - Joanna Goral
- Department of Anatomy, College of Graduate Studies (CGS), Midwestern University, Downers Grove, IL 60515, USA;
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
| |
Collapse
|
49
|
Feltri ML, Weaver MR, Belin S, Poitelon Y. The Hippo pathway: Horizons for innovative treatments of peripheral nerve diseases. J Peripher Nerv Syst 2021; 26:4-16. [PMID: 33449435 DOI: 10.1111/jns.12431] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 12/19/2022]
Abstract
Initially identified in Drosophila, the Hippo signaling pathway regulates how cells respond to their environment by controlling proliferation, migration and differentiation. Many recent studies have focused on characterizing Hippo pathway function and regulation in mammalian cells. Here, we present a brief overview of the major components of the Hippo pathway, as well as their regulation and function. We comprehensively review the studies that have contributed to our understanding of the Hippo pathway in the function of the peripheral nervous system and in peripheral nerve diseases. Finally, we discuss innovative approaches that aim to modulate Hippo pathway components in diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- M Laura Feltri
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Michael R Weaver
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| |
Collapse
|
50
|
Jeanette H, Marziali LN, Bhatia U, Hellman A, Herron J, Kopec AM, Feltri ML, Poitelon Y, Belin S. YAP and TAZ regulate Schwann cell proliferation and differentiation during peripheral nerve regeneration. Glia 2020; 69:1061-1074. [PMID: 33336855 PMCID: PMC7898398 DOI: 10.1002/glia.23949] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/27/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022]
Abstract
YAP and TAZ are effectors of the Hippo pathway that controls multicellular development by integrating chemical and mechanical signals. Peripheral nervous system development depends on the Hippo pathway. We previously showed that loss of YAP and TAZ impairs the development of peripheral nerve as well as Schwann cell myelination. The role of the Hippo pathway in peripheral nerve regeneration has just started to be explored. After injury, Schwann cells adopt new identities to promote regeneration by converting to a repair‐promoting phenotype. While the reprogramming of Schwann cells to repair cells has been well characterized, the maintenance of such repair phenotype cannot be sustained for a very long period, which limits nerve repair in human. First, we show that short or long‐term myelin maintenance is not affected by defect in YAP and TAZ expression. Using crush nerve injury and conditional mutagenesis in mice, we also show that YAP and TAZ are regulators of repair Schwann cell proliferation and differentiation. We found that YAP and TAZ are required in repair Schwann cells for their redifferentiation into myelinating Schwann cell following crush injury. In this present study, we describe how the Hippo pathway and YAP and TAZ regulate remyelination over time during peripheral nerve regeneration.
Collapse
Affiliation(s)
- Haley Jeanette
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Leandro N Marziali
- Department of Biochemistry, Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Urja Bhatia
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Abigail Hellman
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Jacob Herron
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Ashley M Kopec
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Maria Laura Feltri
- Department of Biochemistry, Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA.,Department of Neurology, Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| |
Collapse
|