1
|
Vroemen PAMM, Seijas-Gamardo A, Palmen R, Wieringa PA, Webers CAB, Moroni L, Gorgels TGMF. The Importance of Coating Surface and Composition for Attachment and Survival of Neuronal Cells Under Mechanical Stimulation. J Biomed Mater Res A 2025; 113:e37919. [PMID: 40285752 DOI: 10.1002/jbm.a.37919] [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: 07/22/2024] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
Cell culture of neuronal cells places high demands on the surface for these cells to adhere to and grow on. Native extracellular matrix (ECM) proteins are often applied to the cell culture surface. The substrate is even more important when mechanical strain is applied to the cells in culture. These cells will easily detach and die, precluding the study of how mechanical factors affect these cells. Mechanical factors are, for example, important in the eye disorder glaucoma, which is characterized by the loss of the retinal ganglion cells (RGCs), the retinal neurons that transfer the visual information from the retina via the optic nerve to the brain. High intraocular pressure is the main risk factor of glaucoma. Here, we aimed to find an optimal coating formulation for mechanical testing of the two cell types that are often used for in vitro studies on glaucoma: primary rat retinal ganglion cells (RGCs) and the neuronal PC-12 cell line. Glass and polymer coverslips as well as well plate wells were coated with various substrates: fibronectin, collagen 1, RGD peptide, polyethyleneimine (PEI), poly-D-lysine (PDL), and laminin. We used a thermomixer for 1 min at 500RPM and 37°C to apply mechanical strain and test cell attachment in medium throughput during mechanical stimulation. Cell density, morphology, and cell death were measured to evaluate the coatings. First, a screen of various surfaces and coatings was performed using PC-12 cells, after which a selection of coating strategies was tested with RGCs. For PC-12 cells, the best results were obtained using a coating with a mixture of 10 μg/mL PDL with 2 or 50 μg/mL laminin in PBS (M2). This resulted in the highest cell density, with and without mechanical stimulation. Many other coating strategies failed to provide an effective substrate for adherence and growth of PC-12 cells. Coating composition as well as coating strategy influenced cell attachment and survival. Contrary to PC-12 cells, RGCs performed better in a sequential coating of first 10 μg/mL PDL and then 2 μg/mL laminin (S2). With this protocol, RGCs showed best neurite growth and highest cell density. Based on this difference between PC-12 cells and RGCs, we conclude that the optimal coating depends on the cell type. When reporting cell culture studies, it is important to fully specify culture surface, surface treatment, and coating protocol since all these factors influence cell attachment, growth, and survival.
Collapse
Affiliation(s)
- Pascal A M M Vroemen
- Maastricht UMC+, Maastricht University Medical Centre+, University Eye Clinic, Maastricht, the Netherlands
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Adrián Seijas-Gamardo
- Maastricht UMC+, Maastricht University Medical Centre+, University Eye Clinic, Maastricht, the Netherlands
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Roy Palmen
- Maastricht UMC+, Maastricht University Medical Centre+, University Eye Clinic, Maastricht, the Netherlands
| | - Paul A Wieringa
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Carroll A B Webers
- Maastricht UMC+, Maastricht University Medical Centre+, University Eye Clinic, Maastricht, the Netherlands
| | - Lorenzo Moroni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Theo G M F Gorgels
- Maastricht UMC+, Maastricht University Medical Centre+, University Eye Clinic, Maastricht, the Netherlands
| |
Collapse
|
2
|
Zhurenkov KE, Svirskis D, Connor B, Malmström J. Actuated Hydrogel Platforms To Study Brain Cell Behavior. Adv Healthc Mater 2025; 14:e2404484. [PMID: 40091290 PMCID: PMC12004428 DOI: 10.1002/adhm.202404484] [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: 11/12/2024] [Revised: 02/16/2025] [Indexed: 03/19/2025]
Abstract
The human brain is a highly complex organ characterized by intricate neural networks, biochemical signaling, and unique mechanical properties. The soft and dynamic viscoelastic extracellular matrix (ECM) plays a crucial role in supporting different types of brain cells and influencing their behavior. Understanding how brain cells respond to mechanical stimuli within this complex environment is essential for unraveling fundamental mechanisms of healthy, unhealthy, and regenerative functions within the central nervous system. This requires the development of advanced materials and techniques to study the interplay between mechanical cues and cell responses. Hydrogels have become essential in this research, mimicking the brain's ECM in both chemical composition and mechanical behavior. Conventional hydrogels, while helpful, are static and lack dynamic stimulation. On the other hand, dynamic hydrogels provide reversible, dynamic stimulation, closely replicating the brain's ECM properties. This review discusses current hydrogel platforms used to investigate brain function in health and disease, focusing on traumatic brain injury (TBI)-like conditions and brain tumors. These dynamic materials offer sophisticated tools for understanding brain cell mechanobiology and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Kirill E. Zhurenkov
- Department of Chemical and Materials EngineeringThe University of AucklandAuckland1010New Zealand
- MacDiarmid Institute for Advanced Materials and NanotechnologyWellington6140New Zealand
| | - Darren Svirskis
- School of PharmacyFaculty of Medical and Health SciencesThe University of AucklandAuckland1023New Zealand
| | - Bronwen Connor
- Department of Pharmacology and Clinical PharmacologySchool of Medical Sciences, Faculty of Medical and Health SciencesThe University of AucklandAuckland1023New Zealand
| | - Jenny Malmström
- Department of Chemical and Materials EngineeringThe University of AucklandAuckland1010New Zealand
- MacDiarmid Institute for Advanced Materials and NanotechnologyWellington6140New Zealand
| |
Collapse
|
3
|
Babaliari E, Kavatzikidou P, Xydias D, Psilodimitrakopoulos S, Ranella A, Stratakis E. Flow-Induced Shear Stress Combined with Microtopography Inhibits the Differentiation of Neuro-2a Cells. MICROMACHINES 2025; 16:341. [PMID: 40141952 PMCID: PMC11945430 DOI: 10.3390/mi16030341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/01/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025]
Abstract
Considering that neurological injuries cannot typically self-recover, there is a need to develop new methods to study neuronal outgrowth in a controllable manner in vitro. In this study, a precise flow-controlled microfluidic system featuring custom-designed chambers that integrate laser-microstructured polyethylene terephthalate (PET) substrates comprising microgrooves (MGs) was developed to investigate the combined effect of shear stress and topography on Neuro-2a (N2a) cells' behavior. The MGs were positioned parallel to the flow direction and the response of N2a cells was evaluated in terms of growth and differentiation. Our results demonstrate that flow-induced shear stress could inhibit the differentiation of N2a cells. This microfluidic system could potentially be used as a new model system to study the impact of shear stress on cell differentiation.
Collapse
Affiliation(s)
- Eleftheria Babaliari
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece; (E.B.); (P.K.); (D.X.); (S.P.)
| | - Paraskevi Kavatzikidou
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece; (E.B.); (P.K.); (D.X.); (S.P.)
| | - Dionysios Xydias
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece; (E.B.); (P.K.); (D.X.); (S.P.)
- Department of Materials Science and Technology, University of Crete, 70013 Crete, Greece
| | - Sotiris Psilodimitrakopoulos
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece; (E.B.); (P.K.); (D.X.); (S.P.)
| | - Anthi Ranella
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece; (E.B.); (P.K.); (D.X.); (S.P.)
| | - Emmanuel Stratakis
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece; (E.B.); (P.K.); (D.X.); (S.P.)
- Department of Physics, University of Crete, 70013 Crete, Greece
| |
Collapse
|
4
|
Kwokdinata C, Chew SY. Additive manufacturing in spatial patterning for spinal cord injury treatment. Adv Drug Deliv Rev 2025; 218:115523. [PMID: 39880332 DOI: 10.1016/j.addr.2025.115523] [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: 11/11/2024] [Revised: 01/03/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Combinatorial treatments integrating cells and biomolecules within scaffolds have been investigated to address the multifactorial nature of spinal cord injury (SCI). Current regenerative treatments have been ineffective as they do not consider the spatial positions of various cell types to effectively form functional neural pathways. Emulating the complex heterogeneity of cells in the native spinal cord requires translating the existing biological understanding of spatial patterning in neural development, as well as the influence of biomolecule and mechanical patterning on regional specification and axonal regeneration, to engineer a scaffold for spinal cord regeneration. This review explores the potential of 3D bioprinting to precisely control material, cell and drug patterns in scaffolds, achieving spatial phenotype specification and providing axonal guidance to form appropriate connections. We also discuss the application of extrusion-based and digital light processing bioprinting in integrating mechanical, chemical and biological cues within a scaffold to advance spatially patterned 3D bioprinted scaffold, as well as current challenges and future perspectives in these bioengineering strategies.
Collapse
Affiliation(s)
- Christy Kwokdinata
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University 637459 Singapore
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University 637459 Singapore; Critical Analytics for Manufacturing Personalized-Medicine Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology, Campus for Research Excellence and Technological Enterprise 138602 Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University 308232 Singapore; School of Materials Science and Engineering 639798 Singapore; National Neuroscience Institute, 11 Jalan Tan Tock Seng 308433 Singapore.
| |
Collapse
|
5
|
Neuman K, Zhang X, Lejeune BT, Pizzarella D, Vázquez M, Lewis LH, Koppes AN, Koppes RA. Static Magnetic Stimulation and Magnetic Microwires Synergistically Enhance and Guide Neurite Outgrowth. Adv Healthc Mater 2025; 14:e2403956. [PMID: 39568232 PMCID: PMC11773108 DOI: 10.1002/adhm.202403956] [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: 10/11/2024] [Indexed: 11/22/2024]
Abstract
Axonal growth is heavily influenced by topography and biophysical stimuli including magnetic and electrical fields. Despite extensive investigation, the degree of influence and the underlying genetic mechanisms remain poorly understood. Here, a novel approach to guide neurite growth is undertaken using an innovative ferromagnetic composite material - glass-coated magnetic microwire - to furnish a synergistic combination of magnetic and topographical cues. Whole rat dorsal root ganglia (DRG) are cultured under five different conditions: control, static magnetic field, magnetic microwire, static magnetic field + glass fiber, and static magnetic field + magnetic microwire. DRG outgrowth responses under each condition, including total neurite outgrowth and directionality, are compared. The combination of both magnetic stimulation and topography significantly increases total neurite outgrowth compared to the controls. The combination of magnetic stimulation and magnetic microwire lead to a strong directional bias of growth along the microwire, double what is observed with the glass fiber. Next generation RNA sequencing of DRG exposed to static magnetic field + magnetic microwire reveals the downregulation of genes relating to the immune response, interleukin signaling, and signal transduction. These results set the stage for contemplating future biophysical stimulation for axonal guidance and improved understanding of material-tissue interactions.
Collapse
Affiliation(s)
- Katelyn Neuman
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| | - Xiaoyu Zhang
- Dept. of Mechanical and Industrial EngineeringNortheastern UniversityBostonMA02115USA
| | - Brian. T. Lejeune
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| | | | - Manuel Vázquez
- Instituto de Ciencia de Materiales de MadridCSICMadrid28049Spain
| | - Laura H. Lewis
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
- Dept. of Mechanical and Industrial EngineeringNortheastern UniversityBostonMA02115USA
| | - Abigail N. Koppes
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
- Dept. of BioengineeringNortheastern UniversityBostonMA02115USA
- Dept. of BiologyNortheastern UniversityBostonMA02115USA
| | - Ryan A. Koppes
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| |
Collapse
|
6
|
Habibey R, Striebel J, Meinert M, Latiftikhereshki R, Schmieder F, Nasiri R, Latifi S. Engineered modular neuronal networks-on-chip represent structure-function relationship. Biosens Bioelectron 2024; 261:116518. [PMID: 38924816 DOI: 10.1016/j.bios.2024.116518] [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: 02/08/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Brain function is substantially linked to the highly organized modular structure of neuronal networks. However, the structure of in vitro assembled neuronal circuits often exhibits variability, complicating the consistent recording of network functional output and its correlation to network structure. Therefore, engineering neuronal structures with predefined geometry and reproducible functional features is essential to precisely model in vivo neuronal circuits. Here, we engineered microchannel devices to assemble 2D and 3D modular networks. The microchannel devices were coupled with a multi-electrode array (MEA) electrophysiology system to enable recordings from circuits. Each network consisted of 64 modules connected to their adjacent modules by micron-sized channels. Modular circuits within microchannel devices showed enhanced activity and functional connectivity traits. This includes metrics such as connection weights, clustering coefficient, global efficiency, and the number of hub neurons with higher betweenness centrality. In addition, modular networks demonstrated an increased functional modularity score compared to the randomly formed circuits. Neurons within individual modules displayed uniform network characteristics and predominantly participated in their respective functional communities within the same or neighboring physical modules. These observations highlight that the modular network structure promotes the development of segregated functional connectivity traits while simultaneously enhancing the efficiency of overall network connectivity. Our findings emphasize the significant impact of physical constraints on the activity patterns and functional organization within engineered modular networks. These circuits, characterized by stable modular architecture and intricate functional dynamics-key features of the brain networks-offer a robust in vitro model for advancing neuroscience research.
Collapse
Affiliation(s)
- Rouhollah Habibey
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany; CRTD - Center for Regenerative Therapies TU Dresden, 01307, Dresden, Germany; Dept. Neuroscience, Italian Institute of Technology. Genova, Italy.
| | - Johannes Striebel
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Melissa Meinert
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Roshanak Latiftikhereshki
- Department of Computer Engineering, Faculty of Engineering, Kermanshah Branch, Azad University, Kermanshah, Iran
| | - Felix Schmieder
- Laboratory of Measurement and Sensor System Technique, Faculty of Electrical and Computer Engineering, TU Dresden, Helmholtzstraße 18, 01069, Dresden, Germany
| | - Rohollah Nasiri
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden; AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solna, Sweden
| | - Shahrzad Latifi
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Neuroscience, Rockefeller Neuroscience Institute West Virginia University, Morgantown, WV, 26506, USA
| |
Collapse
|
7
|
Dillon AP, Moslehi S, Brouse B, Keremane S, Philliber S, Griffiths W, Rowland C, Smith JH, Taylor RP. Evolution of Retinal Neuron Fractality When Interfacing with Carbon Nanotube Electrodes. Bioengineering (Basel) 2024; 11:823. [PMID: 39199781 PMCID: PMC11351692 DOI: 10.3390/bioengineering11080823] [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: 07/30/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
Exploring how neurons in the mammalian body interact with the artificial interface of implants can be used to learn about fundamental cell behavior and to refine medical applications. For fundamental and applied research, it is crucial to determine the conditions that encourage neurons to maintain their natural behavior during interactions with non-natural interfaces. Our previous investigations quantified the deterioration of neuronal connectivity when their dendrites deviate from their natural fractal geometry. Fractal resonance proposes that neurons will exhibit enhanced connectivity if an implant's electrode geometry is matched to the fractal geometry of the neurons. Here, we use in vitro imaging to quantify the fractal geometry of mouse retinal neurons and show that they change during interaction with the electrode. Our results demonstrate that it is crucial to understand these changes in the fractal properties of neurons for fractal resonance to be effective in the in vivo mammalian system.
Collapse
Affiliation(s)
- Aiden P. Dillon
- Department of Physics, University of Oregon, Eugene, OR 97403, USA
- Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| | - Saba Moslehi
- Department of Physics, University of Oregon, Eugene, OR 97403, USA
- Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| | - Bret Brouse
- Department of Physics, University of Oregon, Eugene, OR 97403, USA
- Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| | - Saumya Keremane
- Department of Biology, University of Oregon, Eugene, OR 97403, USA
- Department of Biology, Institute of Neurobiology, University of Oregon, Eugene, OR 97403, USA
| | - Sam Philliber
- Department of Physics, University of Oregon, Eugene, OR 97403, USA
- Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| | - Willem Griffiths
- Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Conor Rowland
- Department of Physics, University of Oregon, Eugene, OR 97403, USA
- Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| | - Julian H. Smith
- Department of Physics, University of Oregon, Eugene, OR 97403, USA
- Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| | - Richard P. Taylor
- Department of Physics, University of Oregon, Eugene, OR 97403, USA
- Materials Science Institute, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
8
|
Zhao H, Xiong T, Chu Y, Hao W, Zhao T, Sun X, Zhuang Y, Chen B, Zhao Y, Wang J, Chen Y, Dai J. Biomimetic Dual-Network Collagen Fibers with Porous and Mechanical Cues Reconstruct Neural Stem Cell Niche via AKT/YAP Mechanotransduction after Spinal Cord Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311456. [PMID: 38497893 DOI: 10.1002/smll.202311456] [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: 12/08/2023] [Revised: 02/21/2024] [Indexed: 03/19/2024]
Abstract
Tissue engineering scaffolds can mediate the maneuverability of neural stem cell (NSC) niche to influence NSC behavior, such as cell self-renewal, proliferation, and differentiation direction, showing the promising application in spinal cord injury (SCI) repair. Here, dual-network porous collagen fibers (PCFS) are developed as neurogenesis scaffolds by employing biomimetic plasma ammonia oxidase catalysis and conventional amidation cross-linking. Following optimizing the mechanical parameters of PCFS, the well-matched Young's modulus and physiological dynamic adaptability of PCFS (4.0 wt%) have been identified as a neurogenetic exciter after SCI. Remarkably, porous topographies and curving wall-like protrusions are generated on the surface of PCFS by simple and non-toxic CO2 bubble-water replacement. As expected, PCFS with porous and matched mechanical properties can considerably activate the cadherin receptor of NSCs and induce a series of serine-threonine kinase/yes-associated protein mechanotransduction signal pathways, encouraging cellular orientation, neuron differentiation, and adhesion. In SCI rats, implanted PCFS with matched mechanical properties further integrated into the injured spinal cords, inhibited the inflammatory progression and decreased glial and fibrous scar formation. Wall-like protrusions of PCFS drive multiple neuron subtypes formation and even functional neural circuits, suggesting a viable therapeutic strategy for nerve regeneration and functional recovery after SCI.
Collapse
Affiliation(s)
- Haitao Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
| | - Tiandi Xiong
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yun Chu
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Wangping Hao
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
| | - Tongtong Zhao
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Xinyue Sun
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yan Zhuang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Bing Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology Chinese Academy of Sciences, Beijing, 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology Chinese Academy of Sciences, Beijing, 100101, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Yanyan Chen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Jianwu Dai
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
9
|
Sasaki S, Tsukamoto S, Ishida Y, Kobayashi Y, Inagaki Y, Mano T, Kitamura T, Seriu N, Nakagawa I, Kido A. The Karnofsky Performance Status at Discharge Is a Prognostic Indicator of Life Expectancy in Patients With Glioblastoma. Cureus 2024; 16:e66226. [PMID: 39238708 PMCID: PMC11376000 DOI: 10.7759/cureus.66226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2024] [Indexed: 09/07/2024] Open
Abstract
Background Glioblastoma (GBM) is the most frequent invasive brain tumor and a rapidly progressive disease with a poor prognosis that predominantly affects middle-aged and older adults. The relationship between daily functioning and prognosis in patients with GBM will become more important as advances in multimodality treatment are expected to increase the number of long-term survivors. Methods Sixty-seven patients were initially diagnosed with GBM at our hospital between December 2013 and December 2022. All patients were divided into two groups: those who survived for one year or longer from the date of discharge (Group A) and those who died within one year from the date of discharge (Group B). Muscle strength, nutritional status, and Karnofsky Performance Status (KPS) were examined upon admission (p1), post-surgery (p2), and discharge (p3), and their relationships with prognosis were investigated. Results Group A was significantly younger than Group B, with a significant difference in the total radiation dose. There were no significant differences in the anatomical tumor location, whether the tumor occurred on the left or right side, or tumor size. KPS at discharge (p3) and the degree of improvement in the KPS between p1 and p3 were associated with a good prognosis. Conclusions The KPS varies throughout the treatment. When considering the KPS as a prognostic indicator, the KPS at discharge is the most important, given the structure of the disability and the course of treatment for GBM.
Collapse
Affiliation(s)
- Shogo Sasaki
- Rehabilitation Medicine, Nara Medical University, Kashihara, JPN
| | | | - Yukako Ishida
- Rehabilitation Medicine, Nara Medical University, Kashihara, JPN
| | - Yasuyo Kobayashi
- Rehabilitation Medicine, Nara Medical University, Kashihara, JPN
| | - Yusuke Inagaki
- Rehabilitation Medicine, Nara Medical University, Kashihara, JPN
| | - Tomoo Mano
- Rehabilitation Medicine, Nara Medical University, Kashihara, JPN
| | - Tetsuro Kitamura
- Rehabilitation Medicine, Nara Medical University, Kashihara, JPN
| | - Naoto Seriu
- Rehabilitation Medicine, Nara Medical University, Kashihara, JPN
| | | | - Akira Kido
- Rehabilitation Medicine, Nara Medical University, Kashihara, JPN
| |
Collapse
|
10
|
Marciuš T, Deftu AF, Vuka I, Braeken D, Sapunar D. Electrophysiological properties of dorsal root ganglion neurons cultured on 3D silicon micro-pillar substrates. J Neurosci Methods 2024; 407:110143. [PMID: 38670536 DOI: 10.1016/j.jneumeth.2024.110143] [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: 11/20/2023] [Revised: 02/27/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Silicon-based micro-pillar substrates (MPS), as three-dimensional cell culture platforms with vertically aligned micro-patterned scaffolding structures, are known to facilitate high-quality growth and morphology of dorsal root ganglion (DRG) sensory neurons, promote neurite outgrowth and enhance neurite alignment. However, the electrophysiological aspects of DRG neurons cultured on silicon MPSs have not been thoroughly investigated, which is of greatest importance to ensure that such substrates do not disrupt neuronal homeostasis and function before their widespread adoption in diverse biomedical applications. NEW METHOD We conducted whole-cell patch-clamp recordings to explore the electrophysiological properties of DRG neurons cultured on MPS arrays, utilizing a custom-made upright patch-clamp setup. RESULTS Our findings revealed that DRG neurons exhibited similar electrophysiological responses on patterned MPS samples when compared to the control planar glass surfaces. Notably, there were no significant differences observed in the action potential parameters or firing patterns of action potentials between neurons grown on either substrate. COMPARISON WITH EXISTING METHODS In the current study we for the first time confirmed that successful electrophysiological recordings can be obtained from the cells grown on MPS. CONCLUSION Our results imply that, despite the potential alterations caused by the cumulative trauma of tissue harvest and cell dissociation, essential functional cell properties of DRG neurons appear to be relatively maintained on MPS surfaces. Therefore, vertically aligned silicon MPSs could be considered as a potentially effective three-dimensional system for supporting a controlled cellular environment in culture.
Collapse
Affiliation(s)
- Tihana Marciuš
- Laboratory for Pain Research, University of Split School of Medicine, Split 21000, Croatia
| | - Alexandru-Florian Deftu
- Pain Center, Department of Anesthesiology, Lausanne University Hospital and Department of Fundamental Neurosciences, University of Lausanne, Lausanne 1011, Switzerland
| | - Ivana Vuka
- Technology Transfer Office, Department of Science and Innovation, University of Split, Split 21000, Croatia
| | - Dries Braeken
- Life Sciences Technologies, Imec, Leuven 3001, Belgium
| | - Damir Sapunar
- Laboratory for Pain Research, University of Split School of Medicine, Split 21000, Croatia.
| |
Collapse
|
11
|
Sun Y, Zhang Y, Guo Y, He D, Xu W, Fang W, Zhang C, Zuo Y, Zhang Z. Electrical aligned polyurethane nerve guidance conduit modulates macrophage polarization and facilitates immunoregulatory peripheral nerve regeneration. J Nanobiotechnology 2024; 22:244. [PMID: 38735969 PMCID: PMC11089704 DOI: 10.1186/s12951-024-02507-3] [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: 12/18/2023] [Accepted: 04/29/2024] [Indexed: 05/14/2024] Open
Abstract
Biomaterials can modulate the local immune microenvironments to promote peripheral nerve regeneration. Inspired by the spatial orderly distribution and endogenous electric field of nerve fibers, we aimed to investigate the synergistic effects of electrical and topological cues on immune microenvironments of peripheral nerve regeneration. Nerve guidance conduits (NGCs) with aligned electrospun nanofibers were fabricated using a polyurethane copolymer containing a conductive aniline trimer and degradable L-lysine (PUAT). In vitro experiments showed that the aligned PUAT (A-PUAT) membranes promoted the recruitment of macrophages and induced their polarization towards the pro-healing M2 phenotype, which subsequently facilitated the migration and myelination of Schwann cells. Furthermore, NGCs fabricated from A-PUAT increased the proportion of pro-healing macrophages and improved peripheral nerve regeneration in a rat model of sciatic nerve injury. In conclusion, this study demonstrated the potential application of NGCs in peripheral nerve regeneration from an immunomodulatory perspective and revealed A-PUAT as a clinically-actionable strategy for peripheral nerve injury.
Collapse
Affiliation(s)
- Yiting Sun
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yinglong Zhang
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, China
| | - Yibo Guo
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Dongming He
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Wanlin Xu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Wei Fang
- MOE Key Laboratory of Low-Grade Energy, Utilization Technologies and Systems, CQU-NUS Renewable, Energy Materials & Devices Joint Laboratory, School of Energy & Power Engineering, Chongqing University, Chongqing, 400044, China
| | - Chenping Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yi Zuo
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu, 610064, China.
| | - Zhen Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China.
- Department of Oral and Maxillofacial Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
12
|
Sharifi M, Kamalabadi-Farahani M, Salehi M, Ebrahimi-Barough S, Alizadeh M. Recent advances in enhances peripheral nerve orientation: the synergy of micro or nano patterns with therapeutic tactics. J Nanobiotechnology 2024; 22:194. [PMID: 38643117 PMCID: PMC11031871 DOI: 10.1186/s12951-024-02475-8] [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: 02/23/2024] [Accepted: 04/11/2024] [Indexed: 04/22/2024] Open
Abstract
Several studies suggest that topographical patterns influence nerve cell fate. Efforts have been made to improve nerve cell functionality through this approach, focusing on therapeutic strategies that enhance nerve cell function and support structures. However, inadequate nerve cell orientation can impede long-term efficiency, affecting nerve tissue repair. Therefore, enhancing neurites/axons directional growth and cell orientation is crucial for better therapeutic outcomes, reducing nerve coiling, and ensuring accurate nerve fiber connections. Conflicting results exist regarding the effects of micro- or nano-patterns on nerve cell migration, directional growth, immunogenic response, and angiogenesis, complicating their clinical use. Nevertheless, advances in lithography, electrospinning, casting, and molding techniques to intentionally control the fate and neuronal cells orientation are being explored to rapidly and sustainably improve nerve tissue efficiency. It appears that this can be accomplished by combining micro- and nano-patterns with nanomaterials, biological gradients, and electrical stimulation. Despite promising outcomes, the unclear mechanism of action, the presence of growth cones in various directions, and the restriction of outcomes to morphological and functional nerve cell markers have presented challenges in utilizing this method. This review seeks to clarify how micro- or nano-patterns affect nerve cell morphology and function, highlighting the potential benefits of cell orientation, especially in combined approaches.
Collapse
Affiliation(s)
- Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
| | | | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Alizadeh
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
13
|
Mateus JC, Sousa MM, Burrone J, Aguiar P. Beyond a Transmission Cable-New Technologies to Reveal the Richness in Axonal Electrophysiology. J Neurosci 2024; 44:e1446232023. [PMID: 38479812 PMCID: PMC10941245 DOI: 10.1523/jneurosci.1446-23.2023] [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: 08/07/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 03/17/2024] Open
Abstract
The axon is a neuronal structure capable of processing, encoding, and transmitting information. This assessment contrasts with a limiting, but deeply rooted, perspective where the axon functions solely as a transmission cable of somatodendritic activity, sending signals in the form of stereotypical action potentials. This perspective arose, at least partially, because of the technical difficulties in probing axons: their extreme length-to-diameter ratio and intricate growth paths preclude the study of their dynamics through traditional techniques. Recent findings are challenging this view and revealing a much larger repertoire of axonal computations. Axons display complex signaling processes and structure-function relationships, which can be modulated via diverse activity-dependent mechanisms. Additionally, axons can exhibit patterns of activity that are dramatically different from those of their corresponding soma. Not surprisingly, many of these recent discoveries have been driven by novel technology developments, which allow for in vitro axon electrophysiology with unprecedented spatiotemporal resolution and signal-to-noise ratio. In this review, we outline the state-of-the-art in vitro toolset for axonal electrophysiology and summarize the recent discoveries in axon function it has enabled. We also review the increasing repertoire of microtechnologies for controlling axon guidance which, in combination with the available cutting-edge electrophysiology and imaging approaches, have the potential for more controlled and high-throughput in vitro studies. We anticipate that a larger adoption of these new technologies by the neuroscience community will drive a new era of experimental opportunities in the study of axon physiology and consequently, neuronal function.
Collapse
Affiliation(s)
- J C Mateus
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - M M Sousa
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - J Burrone
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
| | - P Aguiar
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
14
|
Duncan JL, Wang JJ, Glusauskas G, Weagraff GR, Gao Y, Hoeferlin GF, Hunter AH, Hess-Dunning A, Ereifej ES, Capadona JR. In Vivo Characterization of Intracortical Probes with Focused Ion Beam-Etched Nanopatterned Topographies. MICROMACHINES 2024; 15:286. [PMID: 38399014 PMCID: PMC10893395 DOI: 10.3390/mi15020286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
(1) Background: Intracortical microelectrodes (IMEs) are an important part of interfacing with the central nervous system (CNS) and recording neural signals. However, recording electrodes have shown a characteristic steady decline in recording performance owing to chronic neuroinflammation. The topography of implanted devices has been explored to mimic the nanoscale three-dimensional architecture of the extracellular matrix. Our previous work used histology to study the implant sites of non-recording probes and showed that a nanoscale topography at the probe surface mitigated the neuroinflammatory response compared to probes with smooth surfaces. Here, we hypothesized that the improvement in the neuroinflammatory response for probes with nanoscale surface topography would extend to improved recording performance. (2) Methods: A novel design modification was implemented on planar silicon-based neural probes by etching nanopatterned grooves (with a 500 nm pitch) into the probe shank. To assess the hypothesis, two groups of rats were implanted with either nanopatterned (n = 6) or smooth control (n = 6) probes, and their recording performance was evaluated over 4 weeks. Postmortem gene expression analysis was performed to compare the neuroinflammatory response from the two groups. (3) Results: Nanopatterned probes demonstrated an increased impedance and noise floor compared to controls. However, the recording performances of the nanopatterned and smooth probes were similar, with active electrode yields for control probes and nanopatterned probes being approximately 50% and 45%, respectively, by 4 weeks post-implantation. Gene expression analysis showed one gene, Sirt1, differentially expressed out of 152 in the panel. (4) Conclusions: this study provides a foundation for investigating novel nanoscale topographies on neural probes.
Collapse
Affiliation(s)
- Jonathan L. Duncan
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Jaime J. Wang
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Gabriele Glusauskas
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Gwendolyn R. Weagraff
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Yue Gao
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - George F. Hoeferlin
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Allen H. Hunter
- Michigan Center for Materials Characterization, University of Michigan, 500 S. State St, Ann Arbor, MI 48109, USA
| | - Allison Hess-Dunning
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Evon S. Ereifej
- Department of Biomedical Engineering, University of Michigan, 500 S. State St, Ann Arbor, MI 48109, USA
- Veterans Affairs Hospital, 2215 Fuller Rd, Ann Arbor, MI 48105, USA
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| |
Collapse
|
15
|
Wan Z, Li M, Wang Z, Tan H, Li W, Yu L, Samuel DJ. CellT-Net: A Composite Transformer Method for 2-D Cell Instance Segmentation. IEEE J Biomed Health Inform 2024; 28:730-741. [PMID: 37023158 DOI: 10.1109/jbhi.2023.3265006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Cell instance segmentation (CIS) via light microscopy and artificial intelligence (AI) is essential to cell and gene therapy-based health care management, which offers the hope of revolutionary health care. An effective CIS method can help clinicians to diagnose neurological disorders and quantify how well these deadly disorders respond to treatment. To address the CIS task challenged by dataset characteristics such as irregular morphology, variation in sizes, cell adhesion, and obscure contours, we propose a novel deep learning model named CellT-Net to actualize effective cell instance segmentation. In particular, the Swin transformer (Swin-T) is used as the basic model to construct the CellT-Net backbone, as the self-attention mechanism can adaptively focus on useful image regions while suppressing irrelevant background information. Moreover, CellT-Net incorporating Swin-T constructs a hierarchical representation and generates multi-scale feature maps that are suitable for detecting and segmenting cells at different scales. A novel composite style named cross-level composition (CLC) is proposed to build composite connections between identical Swin-T models in the CellT-Net backbone and generate more representational features. The earth mover's distance (EMD) loss and binary cross entropy loss are used to train CellT-Net and actualize the precise segmentation of overlapped cells. The LiveCELL and Sartorius datasets are utilized to validate the model effectiveness, and the results demonstrate that CellT-Net can achieve better model performance for dealing with the challenges arising from the characteristics of cell datasets than state-of-the-art models.
Collapse
|
16
|
Chu Q, Han W, He Z, Hao L, Fu X. Suppression of LPS-activated inflammatory responses and chromosomal histone modifications in macrophages by micropattern-induced nuclear deformation. J Biomed Mater Res A 2024; 112:250-259. [PMID: 37740539 DOI: 10.1002/jbm.a.37617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/24/2023] [Accepted: 09/07/2023] [Indexed: 09/24/2023]
Abstract
Macrophages are important immune effector cells which participate various physiological and pathological conditions. Numerous studies have demonstrated the regulation of macrophage phenotype by micropatterns. It is well accepted that micropatterns affect cellular behaviors through changing cell shape and modulating the associated mechanical sensors on the plasma membrane and cytoskeleton. However, the role of nucleus, which serves as a critical physical sensing device, is often ignored. Herein, we found the nuclear deformation and the subsequently increased chromosomal histone methylation (H3K36me2) may contribute to the micropattern-induced suppression of macrophage inflammatory responses. Specifically, macrophages on micropatterned surfaces expressed lower levels of key inflammatory genes, compared with those on flat surfaces. Further investigation on macrophage nuclei showed that micropatterned surfaces cause shrinkage of nucleus volume and compaction of chromatin. Moreover, micropatterned surfaces elevated the methylation level of H3K36me2 in macrophages, while decreased the methylation level of H3K4me3. Our study provides new mechanistic insight into how micropatterns affect macrophage phenotype and highlights the importance of nuclear shape and chromatin histone modification in mediating micropattern-induced change in cell behaviors.
Collapse
Affiliation(s)
- Qi Chu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, People's Republic of China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China
| | - Weiju Han
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, People's Republic of China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, People's Republic of China
| | - Zhichun He
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, People's Republic of China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China
| | - Lijing Hao
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, People's Republic of China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, People's Republic of China
| | - Xiaoling Fu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou, People's Republic of China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, People's Republic of China
| |
Collapse
|
17
|
Rowland C, Moslehi S, Smith JH, Harland B, Dalrymple-Alford J, Taylor RP. Fractal Resonance: Can Fractal Geometry Be Used to Optimize the Connectivity of Neurons to Artificial Implants? ADVANCES IN NEUROBIOLOGY 2024; 36:877-906. [PMID: 38468068 DOI: 10.1007/978-3-031-47606-8_44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
In parallel to medical applications, exploring how neurons interact with the artificial interface of implants in the human body can be used to learn about their fundamental behavior. For both fundamental and applied research, it is important to determine the conditions that encourage neurons to maintain their natural behavior during these interactions. Whereas previous biocompatibility studies have focused on the material properties of the neuron-implant interface, here we discuss the concept of fractal resonance - the possibility that favorable connectivity properties might emerge by matching the fractal geometry of the implant surface to that of the neurons.To investigate fractal resonance, we first determine the degree to which neurons are fractal and the impact of this fractality on their functionality. By analyzing three-dimensional images of rat hippocampal neurons, we find that the way their dendrites fork and weave through space is important for generating their fractal-like behavior. By modeling variations in neuron connectivity along with the associated energetic and material costs, we highlight how the neurons' fractal dimension optimizes these constraints. To simulate neuron interactions with implant interfaces, we distort the neuron models away from their natural form by modifying the dendrites' fork and weaving patterns. We find that small deviations can induce large changes in fractal dimension, causing the balance between connectivity and cost to deteriorate rapidly. We propose that implant surfaces should be patterned to match the fractal dimension of the neurons, allowing them to maintain their natural functionality as they interact with the implant.
Collapse
Affiliation(s)
- C Rowland
- Physics Department, University of Oregon, Eugene, OR, USA
| | - S Moslehi
- Physics Department, University of Oregon, Eugene, OR, USA
| | - J H Smith
- Physics Department, University of Oregon, Eugene, OR, USA
| | - B Harland
- School of Pharmacy, University of Auckland, Auckland, New Zealand
| | - J Dalrymple-Alford
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - R P Taylor
- Physics Department, University of Oregon, Eugene, OR, USA.
| |
Collapse
|
18
|
Bianchi M, Guzzo S, Lunghi A, Greco P, Pisciotta A, Murgia M, Carnevale G, Fadiga L, Biscarini F. Synergy of Nanotopography and Electrical Conductivity of PEDOT/PSS for Enhanced Neuronal Development. ACS APPLIED MATERIALS & INTERFACES 2023; 15:59224-59235. [PMID: 38091494 PMCID: PMC10755694 DOI: 10.1021/acsami.3c15278] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/28/2023]
Abstract
Biomaterials able to promote neuronal development and neurite outgrowth are highly desired in neural tissue engineering for the repair of damaged or disrupted neural tissue and restoring the axonal connection. For this purpose, the use of either electroactive or micro- and nanostructured materials has been separately investigated. Here, the use of a nanomodulated conductive poly(3,4-ethylendioxithiophene) poly(styrenesulfonate) (PEDOT/PSS) substrate that exhibits instructive topographical and electrical cues at the same time was investigated for the first time. In particular, thin films featuring grooves with sizes comparable with those of neuronal neurites (NanoPEDOT) were fabricated by electrochemical polymerization of PEDOT/PSS on a nanomodulated polycarbonate template. The ability of NanoPEDOT to support neuronal development and direct neurite outgrowth was demonstrated by assessing cell viability and proliferation, expression of neuronal markers, average neurite length, and direction of neuroblastoma N2A cells induced to differentiate on this novel support. In addition to the beneficial effect of the nanogrooved topography, a 30% increase was shown in the average length of neurites when differentiating cells were subjected to an electrical stimulation of a few microamperes for 6 h. The results reported here suggest a favorable effect on the neuronal development of the synergistic combination of nanotopography and electrical stimulation, supporting the use of NanoPEDOT in neural tissue engineering to promote physical and functional reconnection of impaired neural networks.
Collapse
Affiliation(s)
- Michele Bianchi
- Department
of Life Sciences, Università degli
Studi di Modena e Reggio Emilia, 44125 Modena, Italy
- Center
for Translational Neurophysiology of Speech and Communication, Istituto Italiano di Tecnologia, 44121 Ferrara, Italy
| | - Sonia Guzzo
- Center
for Translational Neurophysiology of Speech and Communication, Istituto Italiano di Tecnologia, 44121 Ferrara, Italy
- Section
of Physiology, Università di Ferrara, 44121 Ferrara, Italy
| | - Alice Lunghi
- Center
for Translational Neurophysiology of Speech and Communication, Istituto Italiano di Tecnologia, 44121 Ferrara, Italy
- Section
of Physiology, Università di Ferrara, 44121 Ferrara, Italy
| | - Pierpaolo Greco
- Section
of Physiology, Università di Ferrara, 44121 Ferrara, Italy
| | - Alessandra Pisciotta
- Department
of Surgery, Medicine, Dentistry and Morphological Sciences with Interest
in Transplant, Oncology and Regenerative Medicine, Università di Modena e Reggio Emilia, 44125 Modena, Italy
| | - Mauro Murgia
- Center
for Translational Neurophysiology of Speech and Communication, Istituto Italiano di Tecnologia, 44121 Ferrara, Italy
- Istituto
per lo Studio dei Materiali Nanostrutturati (ISMN-CNR), 40129 Bologna, Italy
| | - Gianluca Carnevale
- Department
of Surgery, Medicine, Dentistry and Morphological Sciences with Interest
in Transplant, Oncology and Regenerative Medicine, Università di Modena e Reggio Emilia, 44125 Modena, Italy
| | - Luciano Fadiga
- Center
for Translational Neurophysiology of Speech and Communication, Istituto Italiano di Tecnologia, 44121 Ferrara, Italy
- Section
of Physiology, Università di Ferrara, 44121 Ferrara, Italy
| | - Fabio Biscarini
- Department
of Life Sciences, Università degli
Studi di Modena e Reggio Emilia, 44125 Modena, Italy
- Center
for Translational Neurophysiology of Speech and Communication, Istituto Italiano di Tecnologia, 44121 Ferrara, Italy
| |
Collapse
|
19
|
Dong X, Zhang H, Duan P, Liu K, Yu Y, Wei W, Wang W, Liu Y, Cheng Q, Liang X, Huo Y, Yan L, Yu A, Dai H. An injectable and adaptable hydrogen sulfide delivery system for modulating neuroregenerative microenvironment. SCIENCE ADVANCES 2023; 9:eadi1078. [PMID: 38117891 PMCID: PMC10732521 DOI: 10.1126/sciadv.adi1078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 11/17/2023] [Indexed: 12/22/2023]
Abstract
Peripheral nerve regeneration is a complex physiological process. Single-function nerve scaffolds often struggle to quickly adapt to the imbalanced regenerative microenvironment, leading to slow nerve regeneration and limited functional recovery. In this study, we demonstrate a "pleiotropic gas transmitter" strategy based on endogenous reactive oxygen species (ROS), which trigger the on-demand H2S release at the defect area for transected peripheral nerve injury (PNI) repair through concurrent neuroregeneration and neuroprotection processing. This H2S delivery system consists of an H2S donor (peroxyTCM) encapsulated in a ROS-responsive polymer (mPEG-PMet) and loaded into a temperature-sensitive poly (amino acid) hydrogel (mPEG-PA-PP). This multi-effect combination strategy greatly promotes the regeneration of PNI, attributed to the physiological effects of H2S. These effects include the inhibition of inflammation and oxidative stress, protection of nerve cells, promotion of angiogenesis, and the restoration of normal mitochondrial function. The adaptive release of pleiotropic messengers to modulate the tissue regeneration microenvironment offers promising peripheral nerve repair and tissue engineering opportunities.
Collapse
Affiliation(s)
- Xianzhen Dong
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, China
| | - Hao Zhang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ping Duan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Kun Liu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, China
| | - Yifeng Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wenying Wei
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, China
| | - Weixing Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yuhang Liu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qiang Cheng
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, China
| | - Xinyue Liang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, China
| | - Yuanfang Huo
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, China
| | - Lesan Yan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, China
| | - Aixi Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan 430070, China
- Shenzhen Research Institute of Wuhan University of Technology, Shenzhen 518000, China
| |
Collapse
|
20
|
Han S, Kim J, Kim SH, Youn W, Kim J, Ji GY, Yang S, Park J, Lee GM, Kim Y, Choi IS. In vitro induction of in vivo-relevant stellate astrocytes in 3D brain-derived, decellularized extracellular matrices. Acta Biomater 2023; 172:218-233. [PMID: 37788738 DOI: 10.1016/j.actbio.2023.09.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/05/2023]
Abstract
In vitro fabrication of 3D cell culture systems that could provide in vivo tissue-like, structural, and biochemical environments to neural cells is essential not only for fundamental studies on brain function and behavior, but also for tissue engineering and regenerative medicine applicable to neural injury and neurodegenerative diseases. In particular, for astrocytes-which actively respond to the surroundings and exhibit varied morphologies based on stimuli (e.g., stiffness and chemicals) in vitro, as well as physiological or pathological conditions in vivo-it is crucial to establish an appropriate milieu in in vitro culture platforms. Herein, we report the induction of in vivo-relevant, stellate-shaped astrocytes derived from cortices of Rattus norvegicus by constructing the 3D cell culture systems of brain-derived, decellularized extracellular matrices (bdECMs). The bdECM hydrogels were mechanically stable and soft, and the bdECM-based 3D scaffolds supplied biochemically active environments that astrocytes could interact with, leading to the development of in vivo-like stellate structures. In addition to the distinct morphology with actively elongated endfeet, the astrocytes, cultured in 3D bdECM scaffolds, would have neurosupportive characteristics, indicated by the accelerated neurite outgrowth in the astrocyte-conditioned media. Furthermore, next-generation sequencing showed that the gene expression profiles of astrocytes cultured in bdECMs were significantly different from those cultured on 2D surfaces. The stellate-shaped astrocytes in the bdECMs were analyzed to have reached a more mature state, for instance, with decreased expression of genes for scaffold ECMs, actin filaments, and cell division. The results suggest that the bdECM-based 3D culture system offers an advanced platform for culturing primary cortical astrocytes and their mixtures with other neural cells, providing a brain-like, structural and biochemical milieu that promotes the maturity and in vivo-like characteristics of astrocytes in both form and gene expression. STATEMENT OF SIGNIFICANCE: Decellularized extracellular matrices (dECMs) have emerged as strong candidates for the construction of three-dimensional (3D) cell cultures in vitro, owing to the potential to provide native biochemical and physical environments. In this study, we fabricated hydrogels of brain-derived dECMs (bdECMs) and cultured primary astrocytes within the bdECM hydrogels in a 3D context. The cultured astrocytes exhibited a stellate morphology distinct from conventional 2D cultures, featuring tridimensionally elongated endfeet. qRT-PCR and NGS-based transcriptomic analyses revealed gene expression patterns indicative of a more mature state, compared with the 2D culture. Moreover, astrocytes cultured in bdECMs showed neurosupportive characteristics, as demonstrated by the accelerated neurite outgrowth in astrocyte-conditioned media. We believe that the bdECM hydrogel-based culture system can serve as an in vitro model system for astrocytes and their coculture with other neural cells, holding significant potential for neural engineering and therapeutic applications.
Collapse
Affiliation(s)
- Sol Han
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Jungnam Kim
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Su Hyun Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, South Korea
| | - Wongu Youn
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Jihoo Kim
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Gil Yong Ji
- Cannabis Medical, Inc., Asan 31418, South Korea
| | - Seoin Yang
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Joohyouck Park
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea
| | - Gyun Min Lee
- Department of Biological Sciences, KAIST, Daejeon 34141, South Korea
| | | | - Insung S Choi
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, South Korea; Department of Bio and Brain Engineering, KAIST, Daejeon 34141, South Korea.
| |
Collapse
|
21
|
Babaliari E, Ranella A, Stratakis E. Microfluidic Systems for Neural Cell Studies. Bioengineering (Basel) 2023; 10:902. [PMID: 37627787 PMCID: PMC10451731 DOI: 10.3390/bioengineering10080902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/05/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Whereas the axons of the peripheral nervous system (PNS) spontaneously regenerate after an injury, the occurring regeneration is rarely successful because axons are usually directed by inappropriate cues. Therefore, finding successful ways to guide neurite outgrowth, in vitro, is essential for neurogenesis. Microfluidic systems reflect more appropriately the in vivo environment of cells in tissues such as the normal fluid flow within the body, consistent nutrient delivery, effective waste removal, and mechanical stimulation due to fluid shear forces. At the same time, it has been well reported that topography affects neuronal outgrowth, orientation, and differentiation. In this review, we demonstrate how topography and microfluidic flow affect neuronal behavior, either separately or in synergy, and highlight the efficacy of microfluidic systems in promoting neuronal outgrowth.
Collapse
Affiliation(s)
- Eleftheria Babaliari
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
| | - Anthi Ranella
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
| | - Emmanuel Stratakis
- Foundation for Research and Technology—Hellas (F.O.R.T.H.), Institute of Electronic Structure and Laser (I.E.S.L.), Vasilika Vouton, 70013 Heraklion, Greece;
- Department of Physics, University of Crete, 70013 Heraklion, Greece
| |
Collapse
|
22
|
Drewry MD, Rothermund K, Syed-Picard FN. Topographical and Chemical Inductive Cues Synergistically Enhance the Schwann Cell Differentiation of Aligned Dental Pulp Stem Cell Sheets. J Tissue Eng Regen Med 2023; 2023:7958770. [PMID: 40226400 PMCID: PMC11918939 DOI: 10.1155/2023/7958770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 04/15/2025]
Abstract
Peripheral nerves have an inherent capacity for regeneration, but these Schwann cell-mediated mechanisms are insufficient for severe injuries. With current clinical treatments, slow regeneration and aberrant reinnervation result in poor functional outcomes. Dental pulp stem cells (DPSCs) offer a promising source of therapeutic neurotrophic factors (NTFs), growth factors that stimulate axon regeneration. Previously, we established that DPSCs can generate scaffold-free sheets with a linearly aligned extracellular matrix (ECM). These sheets provide trophic cues via the DPSCs and directional cues through the aligned ECM to both accelerate and orient axon outgrowth, thus providing a biomaterial capable of addressing the current clinical challenges. DPSCs have a propensity for differentiating into Schwann cells (SC-DPSCs), further enhancing their endogenous NTF expression. Here, we evaluated the effect of inducing SC differentiation on the neuroregenerative bioactivity of our DPSC sheets. These sheets were formed on substrates with linear microgrooves to direct the cells to deposit an aligned ECM. Inducing differentiation using an SC differentiation medium (SCDM) increased NTF expression 2-fold compared to unaligned uDPSC sheets, and this effect was amplified in linearly oriented SC-DPSC sheets by up to 8-fold. Furthermore, these aligned SC-DPSC sheets remodeled the sheet ECM to more closely emulate a regenerative neural microenvironment, expressing 8-fold and 2 × 107-fold more collagen IV and laminin, respectively, than unaligned uDPSC sheets. These data demonstrate that the chemical cues of the SCDM and the mechanotransductive cues of the aligned cell sheet synergistically enhanced the differentiation of DPSCs into repair SC-like cells. To evaluate their functional effects on neuritogenesis, the DPSC sheets were directly cocultured with neuronally differentiated neuroblastoma SH-SY5Y cells. In this in vitro culture system, the aligned SC-DPSC sheets promoted oriented neurite-like outgrowth similar to aligned uninduced DPSC sheets and increased collateral branching, which may emulate stages associated with natural SC-mediated repair processes. Therefore, linearly aligned SC-DPSC sheets have the potential to both promote nerve regeneration and reduce aberrant reinnervation, thus providing a promising biomaterial for applications to improve the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Michelle D. Drewry
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kristi Rothermund
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fatima N. Syed-Picard
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| |
Collapse
|
23
|
Naghilou A, Peter K, Millesi F, Stadlmayr S, Wolf S, Rad A, Semmler L, Supper P, Ploszczanski L, Liu J, Burghammer M, Riekel C, Bismarck A, Backus EHG, Lichtenegger H, Radtke C. Insights into the material properties of dragline spider silk affecting Schwann cell migration. Int J Biol Macromol 2023:125398. [PMID: 37330085 DOI: 10.1016/j.ijbiomac.2023.125398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023]
Abstract
Dragline silk of Trichonephila spiders has attracted attention in various applications. One of the most fascinating uses of dragline silk is in nerve regeneration as a luminal filling for nerve guidance conduits. In fact, conduits filled with spider silk can measure up to autologous nerve transplantation, but the reasons behind the success of silk fibers are not yet understood. In this study dragline fibers of Trichonephila edulis were sterilized with ethanol, UV radiation, and autoclaving and the resulting material properties were characterized with regard to the silk's suitability for nerve regeneration. Rat Schwann cells (rSCs) were seeded on these silks in vitro and their migration and proliferation were investigated as an indication for the fiber's ability to support the growth of nerves. It was found that rSCs migrate faster on ethanol treated fibers. To elucidate the reasons behind this behavior, the fiber's morphology, surface chemistry, secondary protein structure, crystallinity, and mechanical properties were studied. The results demonstrate that the synergy of dragline silk's stiffness and its composition has a crucial effect on the migration of rSCs. These findings pave the way towards understanding the response of SCs to silk fibers as well as the targeted production of synthetic alternatives for regenerative medicine applications.
Collapse
Affiliation(s)
- Aida Naghilou
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Karolina Peter
- University of Natural Resources and Life Sciences, Department of Material Sciences and Process Engineering, Institute of Physics and Materials Science, Peter-Jordan-Strasse 82, 1190 Vienna, Austria
| | - Flavia Millesi
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sarah Stadlmayr
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sonja Wolf
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Anda Rad
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Lorenz Semmler
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Paul Supper
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Leon Ploszczanski
- University of Natural Resources and Life Sciences, Department of Material Sciences and Process Engineering, Institute of Physics and Materials Science, Peter-Jordan-Strasse 82, 1190 Vienna, Austria
| | - Jiliang Liu
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, 38000 Grenoble, France
| | - Manfred Burghammer
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, 38000 Grenoble, France
| | - Christian Riekel
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, 38000 Grenoble, France
| | - Alexander Bismarck
- University of Vienna, Faculty of Chemistry, Institute of Materials Chemistry & Research, Währingerstraße 42, 1090 Vienna, Austria
| | - Ellen H G Backus
- University of Vienna, Faculty of Chemistry, Institute of Physical Chemistry, Währingerstraße 42, 1090 Vienna, Austria
| | - Helga Lichtenegger
- University of Natural Resources and Life Sciences, Department of Material Sciences and Process Engineering, Institute of Physics and Materials Science, Peter-Jordan-Strasse 82, 1190 Vienna, Austria
| | - Christine Radtke
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
24
|
Wu X, Yang K, He S, Zhu F, Kang S, Liu B, Sun C, Pang W, Wang Y. Dual-functional gold nanorods micro pattern guiding cell alignment and cellular microenvironment monitoring. J Colloid Interface Sci 2023; 647:429-437. [PMID: 37269739 DOI: 10.1016/j.jcis.2023.05.167] [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: 01/06/2023] [Revised: 05/10/2023] [Accepted: 05/27/2023] [Indexed: 06/05/2023]
Abstract
Surface topography has become a powerful tool to control cell behaviors, however, it's still difficult to monitor cellular microenvironment changes during topography-induced cell responses. Here, a dual-functional platform integrating cell alignment with extracellular pH (pHe) measurement is proposed. The platform is fabricated by assembling gold nanorods (AuNRs) into micro pattern via wettability difference interface method, which provides topographical cues and surface-enhanced Raman scattering (SERS) effect for cell alignment and biochemical detection respectively. Results demonstrate that contact guidance and cell morphology changes are achieved by the AuNRs micro pattern, and pHe are also obtained by the changes of SERS spectra during cell alignment, where the pHe near cytoplasm is lower than nucleus, revealing the heterogeneity of extracellular microenvironment. Moreover, a correlation between lower extracellular pH and higher cell migration ability is revealed, and AuNRs micro pattern can differentiate cells with different migration ability, which may be an inheritable character during cell division. Furthermore, mesenchymal stem cells response dramatically to AuNRs micro pattern, showing different morphology and increased pHe level, offering the potential of impacting stem cell differentiation. This approach provides a new idea for the research of cell regulation and response mechanism.
Collapse
Affiliation(s)
- Xiaoyu Wu
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Kai Yang
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Shan He
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Feng Zhu
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Shenghui Kang
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Bohua Liu
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Chongling Sun
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Wei Pang
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China
| | - Yanyan Wang
- State Key Laboratory of Precision Measuring Technology and Instruments, College of Precision Instruments and Opto-electronics Engineering, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
25
|
Dai Y, Wang W, Zhou X, li L, Tang Y, Shao M, Lyu F. Biomimetic Electrospun PLLA/PPSB Nanofibrous Scaffold Combined with Human Neural Stem Cells for Spinal Cord Injury Repair. ACS APPLIED NANO MATERIALS 2023; 6:5980-5993. [DOI: 10.1021/acsanm.3c00374] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Yuan Dai
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Weizhong Wang
- Shanghai Fifth People’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200240, China
| | - Xiaojun Zhou
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Linli li
- Department of Orthopedics, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
| | - Yuyi Tang
- Shanghai Fifth People’s Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200240, China
| | - Minghao Shao
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Feizhou Lyu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Orthopedics, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
| |
Collapse
|
26
|
Losero E, Jagannath S, Pezzoli M, Goblot V, Babashah H, Lashuel HA, Galland C, Quack N. Neuronal growth on high-aspect-ratio diamond nanopillar arrays for biosensing applications. Sci Rep 2023; 13:5909. [PMID: 37041255 PMCID: PMC10090193 DOI: 10.1038/s41598-023-32235-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/24/2023] [Indexed: 04/13/2023] Open
Abstract
Monitoring neuronal activity with simultaneously high spatial and temporal resolution in living cell cultures is crucial to advance understanding of the development and functioning of our brain, and to gain further insights in the origin of brain disorders. While it has been demonstrated that the quantum sensing capabilities of nitrogen-vacancy (NV) centers in diamond allow real time detection of action potentials from large neurons in marine invertebrates, quantum monitoring of mammalian neurons (presenting much smaller dimensions and thus producing much lower signal and requiring higher spatial resolution) has hitherto remained elusive. In this context, diamond nanostructuring can offer the opportunity to boost the diamond platform sensitivity to the required level. However, a comprehensive analysis of the impact of a nanostructured diamond surface on the neuronal viability and growth was lacking. Here, we pattern a single crystal diamond surface with large-scale nanopillar arrays and we successfully demonstrate growth of a network of living and functional primary mouse hippocampal neurons on it. Our study on geometrical parameters reveals preferential growth along the nanopillar grid axes with excellent physical contact between cell membrane and nanopillar apex. Our results suggest that neuron growth can be tailored on diamond nanopillars to realize a nanophotonic quantum sensing platform for wide-field and label-free neuronal activity recording with sub-cellular resolution.
Collapse
Affiliation(s)
- Elena Losero
- School of Basic Sciences, Institute of Physics, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland.
- Division of Quantum Metrology and Nanotechnologies, Istituto Nazionale di Ricerca Metrologica (INRiM), Strada delle Cacce 91, 10135, Torino, Italy.
- School of Engineering, Institute of Electrical and Micro Engineering, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland.
| | - Somanath Jagannath
- School of Life Sciences, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Maurizio Pezzoli
- School of Life Sciences, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Valentin Goblot
- School of Basic Sciences, Institute of Physics, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Hossein Babashah
- School of Basic Sciences, Institute of Physics, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Hilal A Lashuel
- School of Life Sciences, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Christophe Galland
- School of Basic Sciences, Institute of Physics, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| | - Niels Quack
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, NSW, Australia
- School of Engineering, Institute of Electrical and Micro Engineering, EPFL, Rte Cantonale, 1015, Lausanne, Switzerland
| |
Collapse
|
27
|
Xuan Z, Peng Q, Larsen T, Gurevich L, de Claville Christiansen J, Zachar V, Pennisi CP. Tailoring Hydrogel Composition and Stiffness to Control Smooth Muscle Cell Differentiation in Bioprinted Constructs. Tissue Eng Regen Med 2023; 20:199-212. [PMID: 36401768 PMCID: PMC10070577 DOI: 10.1007/s13770-022-00500-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/23/2022] [Accepted: 10/04/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Reliable in vitro cellular models are needed to study the phenotypic modulation of smooth muscle cells (SMCs) in health and disease. The aim of this study was to optimize gelatin methacrylate (GelMA)/alginate hydrogels for bioprinting three-dimensional (3D) SMC constructs. METHODS Four different hydrogel groups were prepared by mixing different concentrations (% w/v) of GelMA and alginate: G1 (5/1.5), G2 (5/3), G3 (7.5/1.5), and G4 (7.5/3). GelMA 10% was used as control (G5). A circular structure containing human bladder SMCs was fabricated by using an extrusion-based bioprinter. The effects of the mixing ratios on printability, viability, proliferation, and differentiation of the cells were investigated. RESULTS Rheological analysis showed that the addition of alginate significantly stabilized the change in mechanical properties with temperature variations. The group with the highest GelMA and alginate concentrations (G4) exhibited the highest viscosity, resulting in better stability of the 3D construct after crosslinking. Compared to other hydrogel compositions, cells in G4 maintained high viability (> 80%), exhibited spindle-shaped morphology, and showed a significantly higher proliferation rate within an 8-day period. More importantly, G4 provided an optimal environment for the induction of a SMC contractile phenotype, as evidenced by significant changes in the expression of marker proteins and morphological parameters. CONCLUSION Adjusting the composition of GelMA/alginate hydrogels is an effective means of controlling the SMC phenotype. These hydrogels support bioprinting of 3D models to study phenotypic smooth muscle adaptation, with the prospect of using the constructs in the study of therapies for the treatment of urethral strictures.
Collapse
Affiliation(s)
- Zongzhe Xuan
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, Frederik Bajers Vej 3B, 9220, Aalborg Ø, Denmark
| | - Qiuyue Peng
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, Frederik Bajers Vej 3B, 9220, Aalborg Ø, Denmark
| | - Thomas Larsen
- Materials Science and Engineering Group, Department of Materials and Production, Aalborg University, Pontoppidanstræde 103, 9220, Aalborg, Denmark
| | - Leonid Gurevich
- Materials Science and Engineering Group, Department of Materials and Production, Aalborg University, Pontoppidanstræde 103, 9220, Aalborg, Denmark
| | - Jesper de Claville Christiansen
- Materials Science and Engineering Group, Department of Materials and Production, Aalborg University, Pontoppidanstræde 103, 9220, Aalborg, Denmark
| | - Vladimir Zachar
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, Frederik Bajers Vej 3B, 9220, Aalborg Ø, Denmark
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, Frederik Bajers Vej 3B, 9220, Aalborg Ø, Denmark.
| |
Collapse
|
28
|
LCST-mediated phase separation strategy for preparation of poly(amino alcohol ethers) microparticle-gelatin hydrogel composites. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2023.111983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
29
|
Zheng K, Bai J, Yang H, Xu Y, Pan G, Wang H, Geng D. Nanomaterial-assisted theranosis of bone diseases. Bioact Mater 2022; 24:263-312. [PMID: 36632509 PMCID: PMC9813540 DOI: 10.1016/j.bioactmat.2022.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/27/2022] Open
Abstract
Bone-related diseases refer to a group of skeletal disorders that are characterized by bone and cartilage destruction. Conventional approaches can regulate bone homeostasis to a certain extent. However, these therapies are still associated with some undesirable problems. Fortunately, recent advances in nanomaterials have provided unprecedented opportunities for diagnosis and therapy of bone-related diseases. This review provides a comprehensive and up-to-date overview of current advanced theranostic nanomaterials in bone-related diseases. First, the potential utility of nanomaterials for biological imaging and biomarker detection is illustrated. Second, nanomaterials serve as therapeutic delivery platforms with special functions for bone homeostasis regulation and cellular modulation are highlighted. Finally, perspectives in this field are offered, including current key bottlenecks and future directions, which may be helpful for exploiting nanomaterials with novel properties and unique functions. This review will provide scientific guidance to enhance the development of advanced nanomaterials for the diagnosis and therapy of bone-related diseases.
Collapse
Affiliation(s)
- Kai Zheng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China,Corresponding author.Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Huaiyu Wang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China,Corresponding author.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China,Corresponding author. Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
30
|
Angelaki D, Kavatzikidou P, Fotakis C, Stratakis E, Ranella A. Laser-Structured Si and PLGA Inhibit the Neuro2a Differentiation in Mono- and Co-Culture with Glia. Tissue Eng Regen Med 2022; 20:111-125. [PMID: 36538193 PMCID: PMC9852401 DOI: 10.1007/s13770-022-00497-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/31/2022] [Accepted: 09/25/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The first step towards a successful neural tissue engineering therapy is the development of an appropriate scaffold and the in vitro study of the cellular response onto it. METHODS Here, we fabricated nano- and micro- patterned Si surfaces via direct ultrafast laser irradiation, as well as their replicas in the biodegradable poly(lactide-co-glycolide), in order to use them as culture substrates for neuronal cells. The differentiation of neuro2a cells on the Si platforms and their replicas was studied both in a mono-culture and in a co-culture with glial cells (Schwann-SW10). RESULTS It was found that the substrate's roughness inhibits the differentiation of the neuronal cells even in the presence of the differentiation medium, and the higher the roughness is, the more the differentiation gets limited. CONCLUSION Our results highlight the importance of the substrate's topography for the controlled growth and differentiation of the neuronal cells and their further study via protein screening methods could shed light on the factors that lead to limited differentiation; thus, contributing to the long standing request for culture substrates that induce cells to differentiate.
Collapse
Affiliation(s)
- Despoina Angelaki
- Institute of Electronic Structure and Laser, Foundation for Research and Technology- Hellas (IESL- FORTH), 711 10 Heraklion, Greece ,Department of Physics, University of Crete, 710 03 Heraklion, Greece
| | - Paraskevi Kavatzikidou
- Institute of Electronic Structure and Laser, Foundation for Research and Technology- Hellas (IESL- FORTH), 711 10 Heraklion, Greece
| | - Costas Fotakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology- Hellas (IESL- FORTH), 711 10 Heraklion, Greece ,Department of Physics, University of Crete, 710 03 Heraklion, Greece
| | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology- Hellas (IESL- FORTH), 711 10 Heraklion, Greece ,Department of Physics, University of Crete, 710 03 Heraklion, Greece
| | - Anthi Ranella
- Institute of Electronic Structure and Laser, Foundation for Research and Technology- Hellas (IESL- FORTH), 711 10 Heraklion, Greece
| |
Collapse
|
31
|
Zhang Y, Jiang N, Gan Z. Poly(ε-Caprolactone) Substrates with Micro/Nanohierarchical Patterned Structures for Cell Culture. Macromol Biosci 2022; 22:e2200300. [PMID: 36086924 DOI: 10.1002/mabi.202200300] [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: 07/20/2022] [Revised: 08/23/2022] [Indexed: 01/15/2023]
Abstract
A simple, efficient and controllable one-step template method is proposed to fabricate poly(ε-caprolactone) substrates with micro/nanohierarchical patterned structures. Two kinds of geometric patterns with and without nanowires, i.e., hexagonal and strip with controllable island size and spacing are designed and fabricated. Furthermore, the influence of geometric patterns, island size, island spacing, and patterned nanowires (pNW) on the growth behavior of MG-63 cells is studied in terms of cell density, distribution, proliferation, morphogenesis, and cellular alignment. It is found that MG-63 cells prefer to adhere and grow on the substrate with smaller island size or spacing. Moreover, unlike the hexagonal structure, the strip structure can guide cellular alignment on its surface. In addition, the microisland structures and the pNW play different roles in promoting cell proliferation, distribution, and morphogenesis. It is concluded that the growth behavior of MG-63 cells can be well controlled by precisely adjusting the micro/nanostructure of the substrate surface. A simple and effective method is provided here for the regulation of cell growth behavior.
Collapse
Affiliation(s)
- Yuan Zhang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ni Jiang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhihua Gan
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
32
|
Veiko V, Karlagina Y, Zernitckaia E, Egorova E, Radaev M, Yaremenko A, Chernenko G, Romanov V, Shchedrina N, Ivanova E, Chichkov B, Odintsova G. Laser-Induced µ-Rooms for Osteocytes on Implant Surface: An In Vivo Study. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:4229. [PMID: 36500852 PMCID: PMC9737095 DOI: 10.3390/nano12234229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
Laser processing of dental implant surfaces is becoming a more widespread replacement for classical techniques due to its undeniable advantages, including control of oxide formation and structure and surface relief at the microscale. Thus, using a laser, we created several biomimetic topographies of various shapes on the surface of titanium screw-shaped implants to research their success and survival rates. A distinctive feature of the topographies is the presence of "µ-rooms", which are special spaces created by the depressions and elevations and are analogous to the µ-sized room in which the osteocyte will potentially live. We conducted the comparable in vivo study using dental implants with continuous (G-topography with µ-canals), discrete (S-topography with μ-cavities), and irregular (I-topography) laser-induced topographies. A histological analysis performed with the statistical method (with p-value less than 0.05) was conducted, which showed that G-topography had the highest BIC parameter and contained the highest number of mature osteocytes, indicating the best secondary stability and osseointegration.
Collapse
Affiliation(s)
- Vadim Veiko
- Institute of Laser Technologies, ITMO University, Saint-Petersburg 197101, Russia
| | - Yuliya Karlagina
- Institute of Laser Technologies, ITMO University, Saint-Petersburg 197101, Russia
| | - Ekaterina Zernitckaia
- Department of Dental Surgery and Maxillofacial Surgery, Pavlov First Saint-Petersburg State Medical University, Saint-Petersburg 197022, Russia
| | - Elena Egorova
- Institute of Laser Technologies, ITMO University, Saint-Petersburg 197101, Russia
| | - Maxim Radaev
- Institute of Laser Technologies, ITMO University, Saint-Petersburg 197101, Russia
| | - Andrey Yaremenko
- Department of Dental Surgery and Maxillofacial Surgery, Pavlov First Saint-Petersburg State Medical University, Saint-Petersburg 197022, Russia
| | - Gennadiy Chernenko
- Lenmiriot Dental Implant Prosthetics Manufacture, Saint-Petersburg 193079, Russia
| | - Valery Romanov
- Institute of Laser Technologies, ITMO University, Saint-Petersburg 197101, Russia
| | - Nadezhda Shchedrina
- Institute of Laser Technologies, ITMO University, Saint-Petersburg 197101, Russia
| | - Elena Ivanova
- STEM, School of Science, RMIT University, Melbourne 3000, Australia
| | - Boris Chichkov
- Institute of Quantum Optics, Leibniz University of Hanover, 30167 Hannover, Germany
| | - Galina Odintsova
- Institute of Laser Technologies, ITMO University, Saint-Petersburg 197101, Russia
| |
Collapse
|
33
|
Matino L, Mariano A, Ausilio C, Garg R, Cohen-Karni T, Santoro F. Modulation of Early Stage Neuronal Outgrowth through Out-of-Plane Graphene. NANO LETTERS 2022; 22:8633-8640. [PMID: 36301701 DOI: 10.1021/acs.nanolett.2c03171] [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] [Indexed: 06/16/2023]
Abstract
The correct wiring of a neural network requires neuron to integrate an incredible repertoire of cues found in their extracellular environment. The astonishing efficiency of this process plays a pivotal role in the correct wiring of the brain during development and axon regeneration. Biologically inspired micro- and nanostructured substrates have been shown to regulate axonal outgrowth. In parallel, several studies investigated graphene's potential as a conductive neural interface, able to enhance cell adhesion, neurite sprouting and outgrowth. Here, we engineered a 3D single- to few-layer fuzzy graphene morphology (3DFG), 3DFG on a collapsed Si nanowire (SiNW) mesh template (NT-3DFGc), and 3DFG on a noncollapsed SiNW mesh template (NT-3DFGnc) as neural-instructive materials. The micrometric protruding features of the NWs templates dictated neuronal growth cone establishment, as well as influencing axon elongation and branching. Furthermore, neurons-to-graphene coupling was investigated with comprehensive view of integrin-mediated contact adhesion points and plasma membrane curvature processes.
Collapse
Affiliation(s)
- Laura Matino
- Tissue Electronics, Istituto Italiano di Tecnologia, Naples 80125, Italy
- Dipartimento di Ingegneria Chimica, dei Materiali e delle Produzioni Industriali, DICMAPI, Università "Federico II", Naples 80125, Italy
| | - Anna Mariano
- Tissue Electronics, Istituto Italiano di Tecnologia, Naples 80125, Italy
| | - Chiara Ausilio
- Tissue Electronics, Istituto Italiano di Tecnologia, Naples 80125, Italy
| | - Raghav Garg
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Tzahi Cohen-Karni
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Francesca Santoro
- Tissue Electronics, Istituto Italiano di Tecnologia, Naples 80125, Italy
- Faculty of Electrical Engineering and IT, RWTH Aachen 52074, Germany
- Institute for Biological Information Processing-Bioelectronics, IBI-3, Forschungszentrum Juelich 52428, Germany
| |
Collapse
|
34
|
Joodaki M, Müller B, Schift H, Nallathambi A, Osmani B. Micro-patterned cellulose films for flexible electrodes in medical implants. MICRO AND NANO ENGINEERING 2022. [DOI: 10.1016/j.mne.2022.100162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
35
|
Tudureanu R, Handrea-Dragan IM, Boca S, Botiz I. Insight and Recent Advances into the Role of Topography on the Cell Differentiation and Proliferation on Biopolymeric Surfaces. Int J Mol Sci 2022; 23:7731. [PMID: 35887079 PMCID: PMC9315624 DOI: 10.3390/ijms23147731] [Citation(s) in RCA: 15] [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: 06/09/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 01/27/2023] Open
Abstract
It is well known that surface topography plays an important role in cell behavior, including adhesion, migration, orientation, elongation, proliferation and differentiation. Studying these cell functions is essential in order to better understand and control specific characteristics of the cells and thus to enhance their potential in various biomedical applications. This review proposes to investigate the extent to which various surface relief patterns, imprinted in biopolymer films or in polymeric films coated with biopolymers, by utilizing specific lithographic techniques, influence cell behavior and development. We aim to understand how characteristics such as shape, dimension or chemical functionality of surface relief patterns alter the orientation and elongation of cells, and thus, finally make their mark on the cell proliferation and differentiation. We infer that such an insight is a prerequisite for pushing forward the comprehension of the methodologies and technologies used in tissue engineering applications and products, including skin or bone implants and wound or fracture healing.
Collapse
Affiliation(s)
- Raluca Tudureanu
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babeș-Bolyai University, 400271 Cluj-Napoca, Romania; (R.T.); (I.M.H.-D.); (S.B.)
- Faculty of Physics, Babeș-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Iuliana M. Handrea-Dragan
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babeș-Bolyai University, 400271 Cluj-Napoca, Romania; (R.T.); (I.M.H.-D.); (S.B.)
- Faculty of Physics, Babeș-Bolyai University, 400084 Cluj-Napoca, Romania
| | - Sanda Boca
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babeș-Bolyai University, 400271 Cluj-Napoca, Romania; (R.T.); (I.M.H.-D.); (S.B.)
| | - Ioan Botiz
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babeș-Bolyai University, 400271 Cluj-Napoca, Romania; (R.T.); (I.M.H.-D.); (S.B.)
| |
Collapse
|
36
|
Hsu CC, Serio A, Gopal S, Gelmi A, Chiappini C, Desai RA, Stevens MM. Biophysical Regulations of Epigenetic State and Notch Signaling in Neural Development Using Microgroove Substrates. ACS APPLIED MATERIALS & INTERFACES 2022; 14:32773-32787. [PMID: 35830496 PMCID: PMC9335410 DOI: 10.1021/acsami.2c01996] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A number of studies have recently shown how surface topography can alter the behavior and differentiation patterns of different types of stem cells. Although the exact mechanisms and molecular pathways involved remain unclear, a consistent portion of the literature points to epigenetic changes induced by nuclear remodeling. In this study, we investigate the behavior of clinically relevant neural populations derived from human pluripotent stem cells when cultured on polydimethylsiloxane microgrooves (3 and 10 μm depth grooves) to investigate what mechanisms are responsible for their differentiation capacity and functional behavior. Our results show that microgrooves enhance cell alignment, modify nuclear geometry, and significantly increase cellular stiffness, which we were able to measure at high resolution with a combination of light and electron microscopy, scanning ion conductance microscopy (SICM), and atomic force microscopy (AFM) coupled with quantitative image analysis. The microgrooves promoted significant changes in the epigenetic landscape, as revealed by the expression of key histone modification markers. The main behavioral change of neural stem cells on microgrooves was an increase of neuronal differentiation under basal conditions on the microgrooves. Through measurements of cleaved Notch1 levels, we found that microgrooves downregulate Notch signaling. We in fact propose that microgroove topography affects the differentiation potential of neural stem cells by indirectly altering Notch signaling through geometric segregation and that this mechanism in parallel with topography-dependent epigenetic modulations acts in concert to enhance stem cell neuronal differentiation.
Collapse
Affiliation(s)
- Chia-Chen Hsu
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Andrea Serio
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Sahana Gopal
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Amy Gelmi
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Ciro Chiappini
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Ravi A. Desai
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| | - Molly M. Stevens
- Department
of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Department
of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, U.K.
- Institute
of Biomedical Engineering, Imperial College
London, Exhibition Road, London SW7 2AZ, U.K.
| |
Collapse
|
37
|
Patel M, Ahn S, Koh WG. Topographical pattern for neuronal tissue engineering. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
38
|
McNaughton R, Huo Y, Li G, Ioschpe ADV, Yan L, Man HY, Zhang X. Regulatory Effects of Gradient Microtopographies on Synapse Formation and Neurite Growth in Hippocampal Neurons. JOURNAL OF MICROMECHANICS AND MICROENGINEERING : STRUCTURES, DEVICES, AND SYSTEMS 2022; 32:075005. [PMID: 35814808 PMCID: PMC9262107 DOI: 10.1088/1361-6439/ac73d7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Among approaches aiming toward functional nervous system restoration, those implementing microfabrication techniques allow the manufacture of platforms with distinct geometry where neurons can develop and be guided to form patterned connections in vitro. The interplay between neuronal development and the microenvironment, shaped by the physical limitations, remains largely unknown. Therefore, it is crucial to have an efficient way to quantify neuronal morphological changes induced by physical or contact guidance of the microenvironment. In this study, we first devise and assess a method to prepare anisotropic, gradient poly(dimethylsiloxane) micro-ridge/groove arrays featuring variable local pattern width. We then demonstrate the ability of this single substrate to simultaneously profile the morphologcial and synaptic connectivity changes of primary cultured hippocampal neurons reacting to variable physical conditons, throughout neurodevelopment, in vitro. The gradient microtopography enhanced adhesion within microgrooves, increasing soma density with decreasing pattern width. Decreasing pattern width also reduced dendritic arborization and increased preferential axon growth. Finally, decreasing pattern geometry inhibited presynaptic puncta architecture. Collectively, a method to examine structural development and connectivity in response to physical stimuli is established, and potentially provides insight into microfabricated geometries which promote neural regeneration and repair.
Collapse
Affiliation(s)
- Ryan McNaughton
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Yuda Huo
- Department of Biology, Boston University, Boston, MA, USA
| | - Guicai Li
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | | | - Lei Yan
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Xin Zhang
- Department of Mechanical Engineering, Boston University, Boston, MA, USA
- Photonics Center, Boston University, Boston, MA, USA
| |
Collapse
|
39
|
Zhang SY, Zhao J, Ni JJ, Li H, Quan ZZ, Qing H. Application and prospects of high-throughput screening for in vitro neurogenesis. World J Stem Cells 2022; 14:393-419. [PMID: 35949394 PMCID: PMC9244953 DOI: 10.4252/wjsc.v14.i6.393] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/07/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Over the past few decades, high-throughput screening (HTS) has made great contributions to new drug discovery. HTS technology is equipped with higher throughput, minimized platforms, more automated and computerized operating systems, more efficient and sensitive detection devices, and rapid data processing systems. At the same time, in vitro neurogenesis is gradually becoming important in establishing models to investigate the mechanisms of neural disease or developmental processes. However, challenges remain in generating more mature and functional neurons with specific subtypes and in establishing robust and standardized three-dimensional (3D) in vitro models with neural cells cultured in 3D matrices or organoids representing specific brain regions. Here, we review the applications of HTS technologies on in vitro neurogenesis, especially aiming at identifying the essential genes, chemical small molecules and adaptive microenvironments that hold great prospects for generating functional neurons or more reproductive and homogeneous 3D organoids. We also discuss the developmental tendency of HTS technology, e.g., so-called next-generation screening, which utilizes 3D organoid-based screening combined with microfluidic devices to narrow the gap between in vitro models and in vivo situations both physiologically and pathologically.
Collapse
Affiliation(s)
- Shu-Yuan Zhang
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Juan Zhao
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China
| | - Jun-Jun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hui Li
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Zhen-Zhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
40
|
Mai P, Hampl J, Baca M, Brauer D, Singh S, Weise F, Borowiec J, Schmidt A, Küstner JM, Klett M, Gebinoga M, Schroeder IS, Markert UR, Glahn F, Schumann B, Eckstein D, Schober A. MatriGrid® Based Biological Morphologies: Tools for 3D Cell Culturing. Bioengineering (Basel) 2022; 9:bioengineering9050220. [PMID: 35621498 PMCID: PMC9138054 DOI: 10.3390/bioengineering9050220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 02/06/2023] Open
Abstract
Recent trends in 3D cell culturing has placed organotypic tissue models at another level. Now, not only is the microenvironment at the cynosure of this research, but rather, microscopic geometrical parameters are also decisive for mimicking a tissue model. Over the years, technologies such as micromachining, 3D printing, and hydrogels are making the foundation of this field. However, mimicking the topography of a particular tissue-relevant substrate can be achieved relatively simply with so-called template or morphology transfer techniques. Over the last 15 years, in one such research venture, we have been investigating a micro thermoforming technique as a facile tool for generating bioinspired topographies. We call them MatriGrid®s. In this research account, we summarize our learning outcome from this technique in terms of the influence of 3D micro morphologies on different cell cultures that we have tested in our laboratory. An integral part of this research is the evolution of unavoidable aspects such as possible label-free sensing and fluidic automatization. The development in the research field is also documented in this account.
Collapse
Affiliation(s)
- Patrick Mai
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Jörg Hampl
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| | - Martin Baca
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Dana Brauer
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Sukhdeep Singh
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Frank Weise
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Justyna Borowiec
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - André Schmidt
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Johanna Merle Küstner
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Maren Klett
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Michael Gebinoga
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
| | - Insa S. Schroeder
- Biophysics Division, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany;
| | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, 07747 Jena, Germany; (A.S.); (U.R.M.)
| | - Felix Glahn
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Berit Schumann
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Diana Eckstein
- Institute of Environmental Toxicology, Martin-Luther-University Halle-Wittenberg, 06097 Halle, Germany; (F.G.); (B.S.); (D.E.)
| | - Andreas Schober
- Department of Nano-Biosystems Engineering, Institute of Chemistry and Biotechnology, Ilmenau University of Technology, 98693 Ilmenau, Germany; (P.M.); (M.B.); (D.B.); (S.S.); (F.W.); (J.B.); (J.M.K.); (M.K.); (M.G.)
- Correspondence: (J.H.); (A.S.); Tel.: +49-3677-6933387 (A.S.)
| |
Collapse
|
41
|
Beiriger J, Habib A, Jovanovich N, Kodavali CV, Edwards L, Amankulor N, Zinn PO. The Subventricular Zone in Glioblastoma: Genesis, Maintenance, and Modeling. Front Oncol 2022; 12:790976. [PMID: 35359410 PMCID: PMC8960165 DOI: 10.3389/fonc.2022.790976] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/07/2022] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is a malignant tumor with a median survival rate of 15-16 months with standard care; however, cases of successful treatment offer hope that an enhanced understanding of the pathology will improve the prognosis. The cell of origin in GBM remains controversial. Recent evidence has implicated stem cells as cells of origin in many cancers. Neural stem/precursor cells (NSCs) are being evaluated as potential initiators of GBM tumorigenesis. The NSCs in the subventricular zone (SVZ) have demonstrated similar molecular profiles and share several distinctive characteristics to proliferative glioblastoma stem cells (GSCs) in GBM. Genomic and proteomic studies comparing the SVZ and GBM support the hypothesis that the tumor cells and SVZ cells are related. Animal models corroborate this connection, demonstrating migratory patterns from the SVZ to the tumor. Along with laboratory and animal research, clinical studies have demonstrated improved progression-free survival in patients with GBM after radiation to the ipsilateral SVZ. Additionally, key genetic mutations in GBM for the most part carry regulatory roles in the SVZ as well. An exciting avenue towards SVZ modeling and determining its role in gliomagenesis in the human context is human brain organoids. Here we comprehensively discuss and review the role of the SVZ in GBM genesis, maintenance, and modeling.
Collapse
Affiliation(s)
- Jamison Beiriger
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Ahmed Habib
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Nicolina Jovanovich
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Chowdari V. Kodavali
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Lincoln Edwards
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Nduka Amankulor
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Pascal O. Zinn
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| |
Collapse
|
42
|
Pribyl M, Taus P, Prado-López S, Dozio SM, Schrenk W, Haslinger MJ, Kopp S, Mühlberger M, Wanzenboeck HD. Dense high aspect ratio nanostructures for cell chip applications - Fabrication, replication, and cell interactions. MICRO AND NANO ENGINEERING 2022. [DOI: 10.1016/j.mne.2022.100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
43
|
Drewry M, Dailey MT, Rothermund K, Backman C, Dahl KN, Syed-Picard FN. Promoting and Orienting Axon Extension Using Scaffold-Free Dental Pulp Stem Cell Sheets. ACS Biomater Sci Eng 2022; 8:814-825. [PMID: 34982537 PMCID: PMC9821555 DOI: 10.1021/acsbiomaterials.1c01517] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Current treatments of facial nerve injury result in poor functional outcomes due to slow and inefficient axon regeneration and aberrant reinnervation. To address these clinical challenges, bioactive scaffold-free cell sheets were engineered using neurotrophic dental pulp stem/progenitor cells (DPCs) and their aligned extracellular matrix (ECM). DPCs endogenously supply high levels of neurotrophic factors (NTFs), growth factors capable of stimulating axonal regeneration, and an aligned ECM provides guidance cues to direct axon extension. Human DPCs were grown on a substrate comprising parallel microgrooves, inducing the cells to align and deposit a linearly aligned, collagenous ECM. The resulting cell sheets were robust and could be easily removed from the underlying substrate. DPC sheets produced NTFs at levels previously shown capable of promoting axon regeneration, and, moreover, inducing DPC alignment increased the expression of select NTFs relative to unaligned controls. Furthermore, the aligned DPC sheets were able to stimulate functional neuritogenic effects in neuron-like cells in vitro. Neuronally differentiated neuroblastoma SH-SY5Y cells produced neurites that were significantly more oriented and less branched when cultured on aligned cell sheets relative to unaligned sheets. These data demonstrate that the linearly aligned DPC sheets can biomechanically support axon regeneration and improve axonal guidance which, when applied to a facial nerve injury, will result in more accurate reinnervation. The aligned DPC sheets generated here could be used in combination with commercially available nerve conduits to enhance their bioactivity or be formed into stand-alone scaffold-free nerve conduits capable of facilitating improved facial nerve recovery.
Collapse
Affiliation(s)
- Michelle
D. Drewry
- Department
of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Matthew T. Dailey
- Department
of Oral and Maxillofacial Surgery, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Kristi Rothermund
- Department
of Oral Biology and Center for Craniofacial Regeneration, School of
Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Charles Backman
- Department
of Chemical Engineering, College of Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Kris N. Dahl
- Department
of Chemical Engineering, College of Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States,Department
of Biomedical Engineering, College of Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States,McGowan
Institute for Regenerative Medicine, Pittsburgh, Pennsylvania 15219, United States,Forensics, Thornton Tomasetti, New York, New York 10271, United States
| | - Fatima N. Syed-Picard
- Department
of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States,Department
of Oral Biology and Center for Craniofacial Regeneration, School of
Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States,McGowan
Institute for Regenerative Medicine, Pittsburgh, Pennsylvania 15219, United States,. Phone: 412-648-8824
| |
Collapse
|
44
|
Zhang X, Meng Y, Gong B, Wang T, Lu Y, Zhang L, Xue J. Electrospun Nanofibers for Manipulating the Soft Tissue Regeneration. J Mater Chem B 2022; 10:7281-7308. [DOI: 10.1039/d2tb00609j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Soft tissue damage is a common clinical problem that affects the lives of a large number of patients all over the world. It is of great importance to develop functional...
Collapse
|
45
|
Luo B, Tiwari AP, Chen N, Ramakrishna S, Yang IH. Development of an Axon-Guiding Aligned Nanofiber-Integrated Compartmentalized Microfluidic Neuron Culture System. ACS APPLIED BIO MATERIALS 2021; 4:8424-8432. [PMID: 35005947 DOI: 10.1021/acsabm.1c00960] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Microfluidic-based neuron cell culture systems have recently gained a lot of attention due to their efficiency in supporting the spatial and temporal control of cellular microenvironments. However, the lack of axon guidance is the key limitation in current culture systems. To combat this, we have developed electrospun aligned nanofiber-integrated compartmentalized microfluidic neuron culture systems (NIMSs), where the nanofibers have enabled axonal guidance and stability. The resulting platform significantly improved axon alignment, length, and stability for both rat primary embryonic motor neurons (MNs) and dorsal root ganglia (DRG) neurons compared to the conventional glass-based microfluidic systems (GMSs). The results showed that axonal growth covered more than two times the area on the axonal chamber of NIMSs compared to the area covered for GMSs. Overall, this platform can be used as a valuable tool for fundamental neuroscience research, drug screening, and biomaterial testing.
Collapse
Affiliation(s)
- Baiwen Luo
- N.1 Institute for Health, National University of Singapore, 28 Medical Drive, #05-COR, 117456 Singapore
| | - Arjun Prasad Tiwari
- Department of Mechanical Engineering and Engineering Science, Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, United States
| | - Nuan Chen
- N.1 Institute for Health, National University of Singapore, 28 Medical Drive, #05-COR, 117456 Singapore.,Center of Nanofibers & Nanotechnology, Department of Mechanical Engineering, National University of Singapore, 117575 Singapore
| | - Seeram Ramakrishna
- Center of Nanofibers & Nanotechnology, Department of Mechanical Engineering, National University of Singapore, 117575 Singapore
| | - In Hong Yang
- Department of Mechanical Engineering and Engineering Science, Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, United States
| |
Collapse
|
46
|
Harberts J, Bours K, Siegmund M, Hedrich C, Glatza M, Schöler HR, Haferkamp U, Pless O, Zierold R, Blick RH. Culturing human iPSC-derived neural progenitor cells on nanowire arrays: mapping the impact of nanowire length and array pitch on proliferation, viability, and membrane deformation. NANOSCALE 2021; 13:20052-20066. [PMID: 34842880 DOI: 10.1039/d1nr04352h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Nanowire arrays used as cell culture substrates build a potent tool for advanced biological applications such as cargo delivery and biosensing. The unique topography of nanowire arrays, however, renders them a challenging growth environment for cells and explains why only basic cell lines have been employed in existing studies. Here, we present the culturing of human induced pluripotent stem cell-derived neural progenitor cells on rectangularly arranged nanowire arrays: In detail, we mapped the impact on proliferation, viability, and topography-induced membrane deformation across a multitude of array pitches (1, 3, 5, 10 μm) and nanowire lengths (1.5, 3, 5 μm). Against the intuitive expectation, a reduced proliferation was found on the arrays with the smallest array pitch of 1 μm and long NWs. Typically, cells settle in a fakir-like state on such densely-spaced nanowires and thus experience no substantial stress caused by nanowires indenting the cell membrane. However, imaging of F-actin showed a distinct reorganization of the cytoskeleton along the nanowire tips in the case of small array pitches interfering with regular proliferation. For larger pitches, the cell numbers depend on the NW lengths but proliferation generally continued although heavy deformations of the cell membrane were observed caused by the encapsulation of the nanowires. Moreover, we noticed a strong interaction of the nanowires with the nucleus in terms of squeezing and indenting. Remarkably, the cell viability is maintained at about 85% despite the massive deformation of the cells. Considering the enormous potential of human induced stem cells to study neurodegenerative diseases and the high cellular viability combined with a strong interaction with nanowire arrays, we believe that our results pave the way to apply nanowire arrays to human stem cells for future applications in stem cell research and regenerative medicine.
Collapse
Affiliation(s)
- Jann Harberts
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Katja Bours
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Malte Siegmund
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Carina Hedrich
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Michael Glatza
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Hans R Schöler
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Robert Zierold
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| | - Robert H Blick
- Center for Hybrid Nanostructures (CHyN), Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
- Material Science and Engineering, College of Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
47
|
Dong X, Wu P, Yan L, Liu K, Wei W, Cheng Q, Liang X, Chen Y, Dai H. Oriented nanofibrous P(MMD-co-LA)/Deferoxamine nerve scaffold facilitates peripheral nerve regeneration by regulating macrophage phenotype and revascularization. Biomaterials 2021; 280:121288. [PMID: 34894585 DOI: 10.1016/j.biomaterials.2021.121288] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/12/2021] [Accepted: 11/25/2021] [Indexed: 12/14/2022]
Abstract
Delayed injured nerve regeneration remains a clinical problem, partly ascribing to the lack of regulation of regenerative microenvironment, topographical cues, and blood nourishment. Functional electrospun conduits have been established as an efficacious strategy to facilitate nerve regeneration by providing structural guidance, regulating the regenerative immune microenvironment, and improving vascular regeneration. However, the synthetic polymers conventionally used to fabricate electrospinning scaffolds, such as poly(L-lactic acid), poly(glycolic acid), and poly(lactic-co-glycolic acid), can cause aseptic inflammation due to acidic degradation products. Therefore, a poly[3(S)-methyl-morpholine-2,5-dione-co-lactic] [P(MMD-co-LA)] containing alanine units with good mechanical properties and reduced acid degradation products, was obtained by melt ring-opening polymerization (ROP). Here, we aimed to explore the effect of oriented nanofiber/Deferoxamine (DFO, a hydrophilic angiogenic drug) scaffold in the rapid construction of a favorable regenerative microenvironment, including cell bridge, polarized vascular system, and immune microenvironment. In vitro studies have shown that the scaffold can sustainably release DFO, which accelerates the migration and tube formation of human umbilical vein endothelial cells (HUVECs), as well as the expression of genes related to angiogenesis. The physical clues provided by the arranged nanofibers can regulate the polarization of macrophages and reduce the expression of inflammatory factors. Furthermore, the in vivo results demonstrated a higher M2 polarization level of the oriented nanofibrous scaffold treatment group with reducedinflammation reaction in the injured nerve. Moreover, the in-situ release of DFO up-regulated the expression of HIF1-α and SDF-1α genes, as well as the expression of HIF1-α's target gene VEGF, further promoting revascularization and enhancing nerve regeneration at the defect site. The obtained results provide essential insights on accelerating the creation of the nerve regeneration microenvironment by combining the physiological processes of nerve regeneration with topographical cues and chemical signal induction.
Collapse
Affiliation(s)
- Xianzhen Dong
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Ping Wu
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lesan Yan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Kun Liu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Wenying Wei
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Qiang Cheng
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Xinyue Liang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Yun Chen
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune-Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China; Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu Hydrogen Valley, Foshan, 528200, China.
| |
Collapse
|
48
|
Czerwińska-Główka D, Krukiewicz K. Guidelines for a Morphometric Analysis of Prokaryotic and Eukaryotic Cells by Scanning Electron Microscopy. Cells 2021; 10:3304. [PMID: 34943812 PMCID: PMC8699492 DOI: 10.3390/cells10123304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/11/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
The invention of a scanning electron microscopy (SEM) pushed the imaging methods and allowed for the observation of cell details with a high resolution. Currently, SEM appears as an extremely useful tool to analyse the morphology of biological samples. The aim of this paper is to provide a set of guidelines for using SEM to analyse morphology of prokaryotic and eukaryotic cells, taking as model cases Escherichia coli bacteria and B-35 rat neuroblastoma cells. Herein, we discuss the necessity of a careful sample preparation and provide an optimised protocol that allows to observe the details of cell ultrastructure (≥ 50 nm) with a minimum processing effort. Highlighting the versatility of morphometric descriptors, we present the most informative parameters and couple them with molecular processes. In this way, we indicate the wide range of information that can be collected through SEM imaging of biological materials that makes SEM a convenient screening method to detect cell pathology.
Collapse
Affiliation(s)
| | - Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, 44-100 Gliwice, Poland;
| |
Collapse
|
49
|
Wang S, Zhu C, Zhang B, Hu J, Xu J, Xue C, Bao S, Gu X, Ding F, Yang Y, Gu X, Gu Y. BMSC-derived extracellular matrix better optimizes the microenvironment to support nerve regeneration. Biomaterials 2021; 280:121251. [PMID: 34810037 DOI: 10.1016/j.biomaterials.2021.121251] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 12/26/2022]
Abstract
A favorable microenvironment plays an important role in nerve regeneration. Extracellular matrix (ECM) derived from cultured cells or natural tissues can facilitate nerve regeneration in the presence of various microenvironmental cues, including biochemical, spatial, and biomechanical factors. This study, through proteomics and three-dimensional image analysis, determines that the components and spatial organization of the ECM secreted by bone marrow mesenchymal cells (BMSCs) are more similar to acellular nerves than those of the ECMs derived from Schwann cells (SCs), skin-derived precursor Schwann cells (SKP-SCs), or fibroblasts (FBs). ECM-modified nerve grafts (ECM-NGs) are engineered by co-cultivating BMSCs, SCs, FBs, SKP-SCs with well-designed nerve grafts used to bridge nerve defects. BMSC-ECM-NGs exhibit the most promising nerve repair properties based on the histology, neurophysiology, and behavioral analyses. The regeneration microenvironment formed by the ECM-NGs is also characterized by proteomics, and the advantages of BMSC-ECM-NGs are evidenced by the enhanced expression of factors related to neural regeneration and reduced immune response. Together, these findings indicate that BMSC-derived ECMs create a more superior microenvironment for nerve regeneration than that by the other ECMs and may, therefore, represent a potential alternative for the clinical repair of peripheral nerve defects.
Collapse
Affiliation(s)
- Shengran Wang
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Changlai Zhu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Bin Zhang
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Junxia Hu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Jinghui Xu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Chengbin Xue
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Shuangxi Bao
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Xiaokun Gu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Fei Ding
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Yumin Yang
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Xiaosong Gu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China.
| | - Yun Gu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China.
| |
Collapse
|
50
|
Hydrogel, Electrospun and Composite Materials for Bone/Cartilage and Neural Tissue Engineering. MATERIALS 2021; 14:ma14226899. [PMID: 34832300 PMCID: PMC8624846 DOI: 10.3390/ma14226899] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022]
Abstract
Injuries of the bone/cartilage and central nervous system are still a serious socio-economic problem. They are an effect of diversified, difficult-to-access tissue structures as well as complex regeneration mechanisms. Currently, commercially available materials partially solve this problem, but they do not fulfill all of the bone/cartilage and neural tissue engineering requirements such as mechanical properties, biochemical cues or adequate biodegradation. There are still many things to do to provide complete restoration of injured tissues. Recent reports in bone/cartilage and neural tissue engineering give high hopes in designing scaffolds for complete tissue regeneration. This review thoroughly discusses the advantages and disadvantages of currently available commercial scaffolds and sheds new light on the designing of novel polymeric scaffolds composed of hydrogels, electrospun nanofibers, or hydrogels loaded with nano-additives.
Collapse
|