1
|
Schwartzman JD, McCall M, Ghattas Y, Pugazhendhi AS, Wei F, Ngo C, Ruiz J, Seal S, Coathup MJ. Multifunctional scaffolds for bone repair following age-related biological decline: Promising prospects for smart biomaterial-driven technologies. Biomaterials 2024; 311:122683. [PMID: 38954959 DOI: 10.1016/j.biomaterials.2024.122683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/09/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
The repair of large bone defects due to trauma, disease, and infection can be exceptionally challenging in the elderly. Despite best clinical practice, bone regeneration within contemporary, surgically implanted synthetic scaffolds is often problematic, inconsistent, and insufficient where additional osteobiological support is required to restore bone. Emergent smart multifunctional biomaterials may drive important and dynamic cellular crosstalk that directly targets, signals, stimulates, and promotes an innate bone repair response following age-related biological decline and when in the presence of disease or infection. However, their role remains largely undetermined. By highlighting their mechanism/s and mode/s of action, this review spotlights smart technologies that favorably align in their conceivable ability to directly target and enhance bone repair and thus are highly promising for future discovery for use in the elderly. The four degrees of interactive scaffold smartness are presented, with a focus on bioactive, bioresponsive, and the yet-to-be-developed autonomous scaffold activity. Further, cell- and biomolecular-assisted approaches were excluded, allowing for contemporary examination of the capabilities, demands, vision, and future requisites of next-generation biomaterial-induced technologies only. Data strongly supports that smart scaffolds hold significant promise in the promotion of bone repair in patients with a reduced osteobiological response. Importantly, many techniques have yet to be tested in preclinical models of aging. Thus, greater clarity on their proficiency to counteract the many unresolved challenges within the scope of aging bone is highly warranted and is arguably the next frontier in the field. This review demonstrates that the use of multifunctional smart synthetic scaffolds with an engineered strategy to circumvent the biological insufficiencies associated with aging bone is a viable route for achieving next-generation therapeutic success in the elderly population.
Collapse
Affiliation(s)
| | - Max McCall
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Yasmine Ghattas
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Abinaya Sindu Pugazhendhi
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Fei Wei
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Christopher Ngo
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Jonathan Ruiz
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Sudipta Seal
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA; Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, USA, Orlando, FL
| | - Melanie J Coathup
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
2
|
Grigore A, Coman OA, Păunescu H, Costescu M, Fulga I. Latest Insights into the In Vivo Studies in Murine Regarding the Role of TRP Channels in Wound Healing-A Review. Int J Mol Sci 2024; 25:6753. [PMID: 38928459 PMCID: PMC11204351 DOI: 10.3390/ijms25126753] [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: 05/15/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Wound healing involves physical, chemical and immunological processes. Transient receptor potential (TRP) and other ion channels are implicated in epidermal re-epithelization. Ion movement across ion channels can induce transmembrane potential that leads to transepithelial potential (TEP) changes. TEP is present in epidermis surrounding the lesion decreases and induces an endogenous direct current generating an epithelial electric field (EF) that could be implicated in wound re-epithelialization. TRP channels are involved in the activation of immune cells during mainly the inflammatory phase of wound healing. The aim of the study was to review the mechanisms of ion channel involvement in wound healing in in vivo experiments in murine (mice, rats) and how can this process be influenced. This review used the latest results published in scientific journals over the last year and this year to date (1 January 2023-31 December 3000) in order to include the in-press articles. Some types of TRP channels, such as TRPV1, TRPV3 and TRPA1, are expressed in immune cells and can be activated by inflammatory mediators. The most beneficial effects in wound healing are produced using agonists of TRPV1, TRPV4 and TRPA1 channels or by inhibiting with antagonists, antisense oligonucleotides or knocking down TRPV3 and TRPM8 channels.
Collapse
Affiliation(s)
| | | | - Horia Păunescu
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucureșt, Romania; (A.G.); (O.A.C.); (M.C.); (I.F.)
| | | | | |
Collapse
|
3
|
Lim PLK, Balakrishnan Y, Goh G, Tham KC, Ng YZ, Lunny DP, Leavesley DI, Bonnard C. Automated Electrical Stimulation Therapy Accelerates Re-Epithelialization in a Three-Dimensional In Vitro Human Skin Wound Model. Adv Wound Care (New Rochelle) 2024; 13:217-234. [PMID: 38062745 DOI: 10.1089/wound.2023.0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024] Open
Affiliation(s)
- Priscilla L K Lim
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Yamini Balakrishnan
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Gracia Goh
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Khek-Chian Tham
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Yi Zhen Ng
- Tissue Technologies, Skin Research Institute of Singapore (SRIS), A*STAR, Singapore, Republic of Singapore
| | - Declan P Lunny
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Asian Skin Biobank, Skin Research Institute of Singapore (SRIS), A*STAR, Singapore, Republic of Singapore
| | - David I Leavesley
- Tissue Technologies, Skin Research Institute of Singapore (SRIS), A*STAR, Singapore, Republic of Singapore
| | - Carine Bonnard
- Model Development, A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Asian Skin Biobank, Skin Research Institute of Singapore (SRIS), A*STAR, Singapore, Republic of Singapore
| |
Collapse
|
4
|
Wu J, Xue W, Yun Z, Liu Q, Sun X. Biomedical applications of stimuli-responsive "smart" interpenetrating polymer network hydrogels. Mater Today Bio 2024; 25:100998. [PMID: 38390342 PMCID: PMC10882133 DOI: 10.1016/j.mtbio.2024.100998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/04/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
In recent years, owing to the ongoing advancements in polymer materials, hydrogels have found increasing applications in the biomedical domain, notably in the realm of stimuli-responsive "smart" hydrogels. Nonetheless, conventional single-network stimuli-responsive "smart" hydrogels frequently exhibit deficiencies, including low mechanical strength, limited biocompatibility, and extended response times. In response, researchers have addressed these challenges by introducing a second network to create stimuli-responsive "smart" Interpenetrating Polymer Network (IPN) hydrogels. The mechanical strength of the material can be significantly improved due to the topological entanglement and physical interactions within the interpenetrating structure. Simultaneously, combining different network structures enhances the biocompatibility and stimulus responsiveness of the gel, endowing it with unique properties such as cell adhesion, conductivity, hemostasis/antioxidation, and color-changing capabilities. This article primarily aims to elucidate the stimulus-inducing factors in stimuli-responsive "smart" IPN hydrogels, the impact of the gels on cell behaviors and their biomedical application range. Additionally, we also offer an in-depth exposition of their categorization, mechanisms, performance characteristics, and related aspects. This review furnishes a comprehensive assessment and outlook for the advancement of stimuli-responsive "smart" IPN hydrogels within the biomedical arena. We believe that, as the biomedical field increasingly demands novel materials featuring improved mechanical properties, robust biocompatibility, and heightened stimulus responsiveness, stimuli-responsive "smart" IPN hydrogels will hold substantial promise for wide-ranging applications in this domain.
Collapse
Affiliation(s)
- Jiuping Wu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Wu Xue
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Zhihe Yun
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Qinyi Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Xinzhi Sun
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
5
|
Urabe H, Akimoto R, Kamiya S, Hosoki K, Ichikawa H, Nishiyama T. Pulsed electrical stimulation and amino acid derivatives promote collagen gene expression in human dermal fibroblasts. Cytotechnology 2024; 76:139-151. [PMID: 38304625 PMCID: PMC10828296 DOI: 10.1007/s10616-023-00604-z] [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: 04/18/2023] [Accepted: 10/19/2023] [Indexed: 02/03/2024] Open
Abstract
Several collagen types are important for maintaining skin structure and function. Previous reports show that l-hydroxyproline (Hyp), N-acetyl-l-hydroxyproline (AHyp), and l-alanyl-l-glutamine (Aln-Gln) are biological active substances with collagen synthesis-promoting effects. In this study, we combined the promotive effects of pulsed electrical stimulation (PES) with three amino acid derivatives in human dermal fibroblasts. Fibroblasts were exposed to PES with a 4,800 Hz pulse frequency and a voltage at 1 or 5 V for 15 min. The gene expression of type I and III collagen (fibrillar collagen), type IV and VII collagen (basement membrane collagen and anchoring fibril collagen) were measured by RT-PCR 48 h after PES. PES alone promoted the expression of COL1A1 and COL3A1 at 5 V but did not alter that of COL4A1 and COL7A1. Each AAD and the AAD mixture promoted the expression of COL4A1 and COL7A1 but either repressed, or did not alter, that of COL1A1 and COL3A1. Compared to treatment with each AAD, PES at 5 V with Hyp promoted the expression of COL1A1 and COL3A1, enhanced COL3A1 expression with AHyp, and stimulated COL3A1 expression with Aln-Gln, while COL4A1 and COL7A1 expressions were not affected. PES and the AAD mixture significantly promoted COL4A1 expression in a voltage-dependent manner, and COL1A1 and COL3A1 demonstrated a similar but nonsignificant trend, whereas COL7A1 expression was not affected. The combination of PES with each AAD or the AAD mixture may improve skin structure and function by increasing the expression of basement membrane collagen and dermal fibrillar collagen.
Collapse
Affiliation(s)
- Hiroya Urabe
- Homer Ion Laboratory Co., Ltd, 17-2 Shinsen-cho, Shibuya-ku, Tokyo, 150-0045 Japan
| | - Ryuji Akimoto
- Homer Ion Laboratory Co., Ltd, 17-2 Shinsen-cho, Shibuya-ku, Tokyo, 150-0045 Japan
| | - Shohei Kamiya
- Homer Ion Laboratory Co., Ltd, 17-2 Shinsen-cho, Shibuya-ku, Tokyo, 150-0045 Japan
| | - Katsu Hosoki
- Homer Ion Laboratory Co., Ltd, 17-2 Shinsen-cho, Shibuya-ku, Tokyo, 150-0045 Japan
| | - Hideyuki Ichikawa
- Homer Ion Laboratory Co., Ltd, 17-2 Shinsen-cho, Shibuya-ku, Tokyo, 150-0045 Japan
| | - Toshio Nishiyama
- Homer Ion Laboratory Co., Ltd, 17-2 Shinsen-cho, Shibuya-ku, Tokyo, 150-0045 Japan
- Tokyo University of Agriculture and Technology, Fuchu, Tokyo 183-8509 Japan
| |
Collapse
|
6
|
Liu L, Liu X, Chen Y, Kong M, Zhang J, Jiang M, Zhou H, Yang J, Chen X, Zhang Z, Wu C, Jiang X, Zhang J. Paxillin/HDAC6 regulates microtubule acetylation to promote directional migration of keratinocytes driven by electric fields. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119628. [PMID: 37949303 DOI: 10.1016/j.bbamcr.2023.119628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/18/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
Endogenous electric fields (EFs) have been demonstrated to facilitate wound healing by directing the migration of epidermal cells. Despite the identification of numerous molecules and signaling pathways that are crucial for the directional migration of keratinocytes under EFs, the underlying molecular mechanisms remain undefined. Previous studies have indicated that microtubule (MT) acetylation is linked to cell migration, while Paxillin exerts a significant influence on cell motility. Therefore, we postulated that Paxillin could enhance EF-induced directional migration of keratinocytes by modulating MT acetylation. In the present study, we observed that EFs (200 mV/mm) induced migration of human immortalized epidermal cells (HaCaT) towards the anode, while upregulating Paxillin, downregulating HDAC6, and increasing the level of microtubule acetylation. Our findings suggested that Paxillin plays a pivotal role in inhibiting HDAC6-mediated microtubule acetylation during directional migration under EF regulation. Conversely, downregulation of Paxillin decreased microtubule acetylation and electrotaxis of epidermal cells by promoting HDAC6 expression, and this effect could be reversed by the addition of tubacin, an HDAC6-specific inhibitor. Furthermore, we observed that EFs also mediated the polarization of Paxillin and acetylated α-tubulin, which is critical for directional migration. In conclusion, our study revealed that MT acetylation in EF-guided keratinocyte migration is regulated by the Paxillin/HDAC6 signaling pathway, providing a novel theoretical foundation for the molecular mechanism of EF-guided directional migration of keratinocytes.
Collapse
Affiliation(s)
- Luojia Liu
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Xiaoqiang Liu
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Ying Chen
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Meng Kong
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Jinghong Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Min Jiang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Hongling Zhou
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Jinrui Yang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Xu Chen
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Ze Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Chao Wu
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Xupin Jiang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China.
| | - Jiaping Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China.
| |
Collapse
|
7
|
Li E, Yan R, Qiao H, Sun J, Zou P, Chang J, Li S, Ma Q, Zhang R, Liao B. Combined transcriptomics and proteomics studies on the effect of electrical stimulation on spinal cord injury in rats. Heliyon 2024; 10:e23960. [PMID: 38226269 PMCID: PMC10788535 DOI: 10.1016/j.heliyon.2023.e23960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 11/20/2023] [Accepted: 12/19/2023] [Indexed: 01/17/2024] Open
Abstract
Electrical stimulation (ES) of the spinal cord is a promising therapy for functional rehabilitation after spinal cord injury (SCI). However, the specific mechanism of action is poorly understood. We designed and applied an implanted ES device in the SCI area in rats and determined the effect of ES on the treatment of motor dysfunction after SCI using behavioral scores. Additionally, we examined the molecular characteristics of the samples using proteomic and transcriptomic sequencing. The differential molecules between groups were identified using statistical analyses. Molecular, network, and pathway-based analyses were used to identify group-specific biological features. ES (0.5 mA, 0.1 ms, 50 Hz) had a positive effect on motor dysfunction and neuronal regeneration in rats after SCI. Six samples (three independent replicates in each group) were used for transcriptome sequencing; we obtained 1026 differential genes, comprising 274 upregulated genes and 752 downregulated genes. A total of 10 samples were obtained: four samples in the ES group and six samples in the SCI group; for the proteome sequencing, 48 differential proteins were identified, including 45 up-regulated and three down-regulated proteins. Combined transcriptomic and proteomic studies have shown that the main enrichment pathway is the hedgehog signaling pathway. Western blot results showed that the expression levels of Sonic hedgehog (SHH) (P < 0.001), Smoothened (SMO) (P = 0.0338), and GLI-1 (P < 0.01) proteins in the ES treatment group were significantly higher than those in the SCI group. The immunofluorescence results showed significantly increased expression of SHH (P = 0.0181), SMO (P = 0.021), and GLI-1 (P = 0.0126) in the ES group compared with that in the SCI group. In conclusion, ES after SCI had a positive effect on motor dysfunction and anti-inflammatory effects in rats. Moreover, transcriptomic and proteomic sequencing also provided unique perspectives on the complex relationships between ES on SCI, where the SHH signaling pathway plays a critical role. Our study provides a significant theoretical foundation for the clinical implementation of ES therapy in patients with SCI.
Collapse
Affiliation(s)
- Erliang Li
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Rongbao Yan
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huanhuan Qiao
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Jin Sun
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Peng Zou
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiaqi Chang
- School of Automation Science and Electrical Engineering, Beihang University, 37th Xueyuan Road, Beijing, China
| | - Shuang Li
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Qiong Ma
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Rui Zhang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Bo Liao
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
8
|
Murugan NJ, Cariba S, Abeygunawardena S, Rouleau N, Payne SL. Biophysical control of plasticity and patterning in regeneration and cancer. Cell Mol Life Sci 2023; 81:9. [PMID: 38099951 PMCID: PMC10724343 DOI: 10.1007/s00018-023-05054-6] [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: 08/18/2023] [Revised: 10/12/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023]
Abstract
Cells and tissues display a remarkable range of plasticity and tissue-patterning activities that are emergent of complex signaling dynamics within their microenvironments. These properties, which when operating normally guide embryogenesis and regeneration, become highly disordered in diseases such as cancer. While morphogens and other molecular factors help determine the shapes of tissues and their patterned cellular organization, the parallel contributions of biophysical control mechanisms must be considered to accurately predict and model important processes such as growth, maturation, injury, repair, and senescence. We now know that mechanical, optical, electric, and electromagnetic signals are integral to cellular plasticity and tissue patterning. Because biophysical modalities underly interactions between cells and their extracellular matrices, including cell cycle, metabolism, migration, and differentiation, their applications as tuning dials for regenerative and anti-cancer therapies are being rapidly exploited. Despite this, the importance of cellular communication through biophysical signaling remains disproportionately underrepresented in the literature. Here, we provide a review of biophysical signaling modalities and known mechanisms that initiate, modulate, or inhibit plasticity and tissue patterning in models of regeneration and cancer. We also discuss current approaches in biomedical engineering that harness biophysical control mechanisms to model, characterize, diagnose, and treat disease states.
Collapse
Affiliation(s)
- Nirosha J Murugan
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada.
- Allen Discovery Center, Tufts University, Medford, MA, USA.
| | - Solsa Cariba
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | | | - Nicolas Rouleau
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada
- Allen Discovery Center, Tufts University, Medford, MA, USA
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Samantha L Payne
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
9
|
Casal D, Casimiro MH, Ferreira LM, Leal JP, Rodrigues G, Lopes R, Moura DL, Gonçalves L, Lago JB, Pais D, Santos PMP. Review of Piezoelectrical Materials Potentially Useful for Peripheral Nerve Repair. Biomedicines 2023; 11:3195. [PMID: 38137416 PMCID: PMC10740581 DOI: 10.3390/biomedicines11123195] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
It has increasingly been recognized that electrical currents play a pivotal role in cell migration and tissue repair, in a process named "galvanotaxis". In this review, we summarize the current evidence supporting the potential benefits of electric stimulation (ES) in the physiology of peripheral nerve repair (PNR). Moreover, we discuss the potential of piezoelectric materials in this context. The use of these materials has deserved great attention, as the movement of the body or of the external environment can be used to power internally the electrical properties of devices used for providing ES or acting as sensory receptors in artificial skin (e-skin). The fact that organic materials sustain spontaneous degradation inside the body means their piezoelectric effect is limited in duration. In the case of PNR, this is not necessarily problematic, as ES is only required during the regeneration period. Arguably, piezoelectric materials have the potential to revolutionize PNR with new biomedical devices that range from scaffolds and nerve-guiding conduits to sensory or efferent components of e-skin. However, much remains to be learned regarding piezoelectric materials, their use in manufacturing of biomedical devices, and their sterilization process, to fine-tune their safe, effective, and predictable in vivo application.
Collapse
Affiliation(s)
- Diogo Casal
- Departamento de Anatomia, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (L.G.); (D.P.)
- Plastic and Reconstructive Surgery Department and Burn Unit, Centro Hospitalar Universitário de Lisboa Central, Rua José António Serrano, 1169-045 Lisbon, Portugal
| | - Maria Helena Casimiro
- Centro de Ciências e Tecnologias Nucleares (C2TN), Instituto Superior Técnico (IST), Universidade de Lisboa, 2695-066 Bobadela, Portugal; (M.H.C.); (P.M.P.S.)
| | - Luís M. Ferreira
- Departamento de Engenharia e Ciências Nucleares (DECN), Instituto Superior Técnico (IST), Universidade de Lisboa, 2695-066 Bobadela, Portugal;
| | - João Paulo Leal
- Centro de Química Estrutural (CQE), Institute of Molecular Sciences (IMS), Instituto Superior Técnico (IST), Universidade de Lisboa, 2695-066 Bobadela, Portugal;
| | - Gabriela Rodrigues
- Centro de Ecologia, Evolução e Alterações Ambientais (cE3c) & CHANGE—Global Change and Sustainability Institute, Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa (FCUL), 1749-016 Lisboa, Portugal;
| | - Raquel Lopes
- Gynaecology and Obstetrics Department, Maternidade Alfredo da Costa, Centro Hospitalar Universitário de Lisboa Central, R. Viriato 1, 2890-495 Lisboa, Portugal;
| | - Diogo Lino Moura
- Anatomy Institute and Orthopedics Department, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal;
- Spine Unit, Orthopedics Department, Coimbra University Hospital, 3000-602 Coimbra, Portugal
| | - Luís Gonçalves
- Departamento de Anatomia, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (L.G.); (D.P.)
| | - João B. Lago
- Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa (FCUL), 1749-016 Lisboa, Portugal;
| | - Diogo Pais
- Departamento de Anatomia, NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (L.G.); (D.P.)
| | - Pedro M. P. Santos
- Centro de Ciências e Tecnologias Nucleares (C2TN), Instituto Superior Técnico (IST), Universidade de Lisboa, 2695-066 Bobadela, Portugal; (M.H.C.); (P.M.P.S.)
| |
Collapse
|
10
|
Jang HJ, Tiruneh DM, Ryu H, Yoon JK. Piezoelectric and Triboelectric Nanogenerators for Enhanced Wound Healing. Biomimetics (Basel) 2023; 8:517. [PMID: 37999158 PMCID: PMC10669670 DOI: 10.3390/biomimetics8070517] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/22/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023] Open
Abstract
Wound healing is a highly orchestrated biological process characterized by sequential phases involving inflammation, proliferation, and tissue remodeling, and the role of endogenous electrical signals in regulating these phases has been highlighted. Recently, external electrostimulation has been shown to enhance these processes by promoting cell migration, extracellular matrix formation, and growth factor release while suppressing pro-inflammatory signals and reducing the risk of infection. Among the innovative approaches, piezoelectric and triboelectric nanogenerators have emerged as the next generation of flexible and wireless electronics designed for energy harvesting and efficiently converting mechanical energy into electrical power. In this review, we discuss recent advances in the emerging field of nanogenerators for harnessing electrical stimulation to accelerate wound healing. We elucidate the fundamental mechanisms of wound healing and relevant bioelectric physiology, as well as the principles underlying each nanogenerator technology, and review their preclinical applications. In addition, we address the prominent challenges and outline the future prospects for this emerging era of electrical wound-healing devices.
Collapse
Affiliation(s)
- Hye-Jeong Jang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea;
| | - Daniel Manaye Tiruneh
- Department of Intelligence Energy and Industry, Chung-Ang University, Seoul 06974, Republic of Korea;
| | - Hanjun Ryu
- Department of Intelligence Energy and Industry, Chung-Ang University, Seoul 06974, Republic of Korea;
- Department of Advanced Materials Engineering, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea;
| |
Collapse
|
11
|
Alvarez-Lorenzo C, Zarur M, Seijo-Rabina A, Blanco-Fernandez B, Rodríguez-Moldes I, Concheiro A. Physical stimuli-emitting scaffolds: The role of piezoelectricity in tissue regeneration. Mater Today Bio 2023; 22:100740. [PMID: 37521523 PMCID: PMC10374602 DOI: 10.1016/j.mtbio.2023.100740] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/01/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023] Open
Abstract
The imbalance between life expectancy and quality of life is increasing due to the raising prevalence of chronic diseases. Musculoskeletal disorders and chronic wounds affect a growing percentage of people and demand more efficient tools for regenerative medicine. Scaffolds that can better mimic the natural physical stimuli that tissues receive under healthy conditions and during healing may significantly aid the regeneration process. Shape, mechanical properties, pore size and interconnectivity have already been demonstrated to be relevant scaffold features that can determine cell adhesion and differentiation. Much less attention has been paid to scaffolds that can deliver more dynamic physical stimuli, such as electrical signals. Recent developments in the precise measurement of electrical fields in vivo have revealed their key role in cell movement (galvanotaxis), growth, activation of secondary cascades, and differentiation to different lineages in a variety of tissues, not just neural. Piezoelectric scaffolds can mimic the natural bioelectric potentials and gradients in an autonomous way by generating the electric stimuli themselves when subjected to mechanical loads or, if the patient or the tissue lacks mobility, ultrasound irradiation. This review provides an analysis on endogenous bioelectrical signals, recent developments on piezoelectric scaffolds for bone, cartilage, tendon and nerve regeneration, and their main outcomes in vivo. Wound healing with piezoelectric dressings is addressed in the last section with relevant examples of performance in animal models. Results evidence that a fine adjustment of material composition and processing (electrospinning, corona poling, 3D printing, annealing) provides scaffolds that act as true emitters of electrical stimuli that activate endogenous signaling pathways for more efficient and long-term tissue repair.
Collapse
Affiliation(s)
- Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Mariana Zarur
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Alejandro Seijo-Rabina
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Barbara Blanco-Fernandez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Isabel Rodríguez-Moldes
- Grupo NEURODEVO, Departamento de Bioloxía Funcional, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| |
Collapse
|
12
|
Leal J, Shaner S, Jedrusik N, Savelyeva A, Asplund M. Electrotaxis evokes directional separation of co-cultured keratinocytes and fibroblasts. Sci Rep 2023; 13:11444. [PMID: 37454232 PMCID: PMC10349865 DOI: 10.1038/s41598-023-38664-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Bioelectric communication plays a significant role in several cellular processes and biological mechanisms, such as division, differentiation, migration, cancer metastasis, and wound healing. Ion flow across cellular walls leads to potential gradients and subsequent formation of constant or time-varying electric fields(EFs), which regulate cellular processes. An EF is natively generated towards the wound center during epithelial wound healing, aiming to align and guide cell migration, particularly of macrophages, fibroblasts, and keratinocytes. While this phenomenon, known as electrotaxis or galvanotaxis, has been extensively investigated across many cell types, it is typically explored one cell type at a time, which does not accurately represent cellular interactions during complex biological processes. Here we show the co-cultured electrotaxis of epidermal keratinocytes and dermal fibroblasts with a salt-bridgeless microfluidic approach for the first time. The electrotactic response of these cells was first assessed in mono-culture to establish a baseline, resulting in the characteristic cathodic migration for keratinocytes and anodic for fibroblasts. Both cell types retained their electrotactic properties in co-culture leading to clear cellular partition even in the presence of cellular collisions. The methods leveraged here pave the way for future co-culture electrotaxis experiments where the concurrent influence of cell types can be thoroughly investigated.
Collapse
Affiliation(s)
- José Leal
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany.
- BrainLinks-BrainTools Center, University of Freiburg, Freiburg, Germany.
| | - Sebastian Shaner
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- BrainLinks-BrainTools Center, University of Freiburg, Freiburg, Germany
| | - Nicole Jedrusik
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- BrainLinks-BrainTools Center, University of Freiburg, Freiburg, Germany
| | - Anna Savelyeva
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- BrainLinks-BrainTools Center, University of Freiburg, Freiburg, Germany
| | - Maria Asplund
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany.
- BrainLinks-BrainTools Center, University of Freiburg, Freiburg, Germany.
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany.
- Department of Microtechnology and Nanoscience, Chalmers University of Technology, Gothenburg, Sweden.
- Division of Nursing and Medical Technology, Luleå University of Technology, 97187, Luleå, Sweden.
| |
Collapse
|
13
|
Shahemi NH, Mahat MM, Asri NAN, Amir MA, Ab Rahim S, Kasri MA. Application of Conductive Hydrogels on Spinal Cord Injury Repair: A Review. ACS Biomater Sci Eng 2023. [PMID: 37364251 DOI: 10.1021/acsbiomaterials.3c00194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Spinal cord injury (SCI) causes severe motor or sensory damage that leads to long-term disabilities due to disruption of electrical conduction in neuronal pathways. Despite current clinical therapies being used to limit the propagation of cell or tissue damage, the need for neuroregenerative therapies remains. Conductive hydrogels have been considered a promising neuroregenerative therapy due to their ability to provide a pro-regenerative microenvironment and flexible structure, which conforms to a complex SCI lesion. Furthermore, their conductivity can be utilized for noninvasive electrical signaling in dictating neuronal cell behavior. However, the ability of hydrogels to guide directional axon growth to reach the distal end for complete nerve reconnection remains a critical challenge. In this Review, we highlight recent advances in conductive hydrogels, including the incorporation of conductive materials, fabrication techniques, and cross-linking interactions. We also discuss important characteristics for designing conductive hydrogels for directional growth and regenerative therapy. We propose insights into electrical conductivity properties in a hydrogel that could be implemented as guidance for directional cell growth for SCI applications. Specifically, we highlight the practical implications of recent findings in the field, including the potential for conductive hydrogels to be used in clinical applications. We conclude that conductive hydrogels are a promising neuroregenerative therapy for SCI and that further research is needed to optimize their design and application.
Collapse
Affiliation(s)
- Nur Hidayah Shahemi
- Faculty of Applied Sciences, Universiti Teknologi MARA, 40450 Shah Alam, Selangor, Malaysia
| | - Mohd Muzamir Mahat
- Faculty of Applied Sciences, Universiti Teknologi MARA, 40450 Shah Alam, Selangor, Malaysia
| | - Nurul Ain Najihah Asri
- Faculty of Applied Sciences, Universiti Teknologi MARA, 40450 Shah Alam, Selangor, Malaysia
| | - Muhammad Abid Amir
- Faculty of Medicine, Sungai Buloh Campus, Universiti Teknologi MARA, 47000 Sungai Buloh, Selangor, Malaysia
| | - Sharaniza Ab Rahim
- Faculty of Medicine, Sungai Buloh Campus, Universiti Teknologi MARA, 47000 Sungai Buloh, Selangor, Malaysia
| | - Mohamad Arif Kasri
- Kulliyyah of Science, International Islamic University Malaysia, 25200 Kuantan, Pahang, Malaysia
| |
Collapse
|
14
|
Luo B, Zhou Q, Chen W, Sun L, Yang L, Guo Y, Liu H, Wu Z, Neisiany RE, Qin X, Pan J, You Z. Nonadjacent Wireless Electrotherapy for Tissue Repair by a 3D-Printed Bioresorbable Fully Soft Triboelectric Nanogenerator. NANO LETTERS 2023; 23:2927-2937. [PMID: 36926930 DOI: 10.1021/acs.nanolett.3c00300] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Electrotherapy is a promising tissue repair technique. However, electrotherapy devices are frequently complex and must be placed adjacent to injured tissue, thereby limiting their clinical application. Here, we propose a general strategy to facilitate tissue repair by modulating endogenous electric fields with nonadjacent (approximately 44 mm) wireless electrotherapy through a 3D-printed entirely soft and bioresorbable triboelectric nanogenerator based stimulator, without any electrical accessories, which has biomimetic mechanical properties similar to those of soft tissue. In addition, the feasibility of using the stimulator to construct an electrical double layer with tissue for nonadjacent wireless electrotherapy was demonstrated by skin and muscle injury models. The treated groups showed significantly improved tissue repair compared with the control group. In conclusion, we developed a promising electrotherapy strategy and may inspire next-generation electrotherapy for tissue repair.
Collapse
Affiliation(s)
- Bin Luo
- College of Textiles, State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Donghua University, Shanghai 201620, People's Republic of China
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, People's Republic of China
| | - Qiangqiang Zhou
- Department of Endodontics, Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai 200001, People's Republic of China
| | - Wenyi Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, People's Republic of China
| | - Lijie Sun
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, People's Republic of China
| | - Lei Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, People's Republic of China
| | - Yifan Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, People's Republic of China
| | - Huijie Liu
- College of Textiles, State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Donghua University, Shanghai 201620, People's Republic of China
| | - Zekai Wu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, People's Republic of China
| | - Rasoul Esmaeely Neisiany
- Department of Materials and Polymer Engineering, Faculty of Engineering, Hakim Sabzevari University, Sabzevar 9617976487, Iran
| | - Xiaohong Qin
- College of Textiles, State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Donghua University, Shanghai 201620, People's Republic of China
| | - Jie Pan
- Department of Orthodontics, Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai 200001, People's Republic of China
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Institute of Functional Materials, Research Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society), Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, Donghua University, Shanghai 201620, People's Republic of China
| |
Collapse
|
15
|
Shaner S, Savelyeva A, Kvartuh A, Jedrusik N, Matter L, Leal J, Asplund M. Bioelectronic microfluidic wound healing: a platform for investigating direct current stimulation of injured cell collectives. LAB ON A CHIP 2023; 23:1531-1546. [PMID: 36723025 PMCID: PMC10013350 DOI: 10.1039/d2lc01045c] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/14/2023] [Indexed: 06/18/2023]
Abstract
Upon cutaneous injury, the human body naturally forms an electric field (EF) that acts as a guidance cue for relevant cellular and tissue repair and reorganization. However, the direct current (DC) flow imparted by this EF can be impacted by a variety of diseases. This work delves into the impact of DC stimulation on both healthy and diabetic in vitro wound healing models of human keratinocytes, the most prevalent cell type of the skin. The culmination of non-metal electrode materials and prudent microfluidic design allowed us to create a compact bioelectronic platform to study the effects of different sustained (12 hours galvanostatic DC) EF configurations on wound closure dynamics. Specifically, we compared if electrotactically closing a wound's gap from one wound edge (i.e., uni-directional EF) is as effective as compared to alternatingly polarizing both the wound's edges (i.e., pseudo-converging EF) as both of these spatial stimulation strategies are fundamental to the eventual translational electrode design and strategy. We found that uni-directional electric guidance cues were superior in group keratinocyte healing dynamics by enhancing the wound closure rate nearly three-fold for both healthy and diabetic-like keratinocyte collectives, compared to their non-stimulated respective controls. The motility-inhibited and diabetic-like keratinocytes regained wound closure rates with uni-directional electrical stimulation (increase from 1.0 to 2.8% h-1) comparable to their healthy non-stimulated keratinocyte counterparts (3.5% h-1). Our results bring hope that electrical stimulation delivered in a controlled manner can be a viable pathway to accelerate wound repair, and also by providing a baseline for other researchers trying to find an optimal electrode blueprint for in vivo DC stimulation.
Collapse
Affiliation(s)
- Sebastian Shaner
- Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 201, 79110, Freiburg, Germany
- Brainlinks-Braintools Center, Georges-Köhler-Allee 201, 79110, Freiburg, Germany.
| | - Anna Savelyeva
- Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 201, 79110, Freiburg, Germany
- Brainlinks-Braintools Center, Georges-Köhler-Allee 201, 79110, Freiburg, Germany.
| | - Anja Kvartuh
- Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 201, 79110, Freiburg, Germany
| | - Nicole Jedrusik
- Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 201, 79110, Freiburg, Germany
- Brainlinks-Braintools Center, Georges-Köhler-Allee 201, 79110, Freiburg, Germany.
| | - Lukas Matter
- Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 201, 79110, Freiburg, Germany
| | - José Leal
- Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 201, 79110, Freiburg, Germany
- Brainlinks-Braintools Center, Georges-Köhler-Allee 201, 79110, Freiburg, Germany.
| | - Maria Asplund
- Department of Microsystems Engineering, University of Freiburg, Georges-Köhler-Allee 201, 79110, Freiburg, Germany
- Brainlinks-Braintools Center, Georges-Köhler-Allee 201, 79110, Freiburg, Germany.
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Albertstr. 19, 79104, Freiburg, Germany
- Division of Nursing and Medical Technology, Luleå University of Technology, 971 87, Luleå, Sweden
- Department of Microtechnology and Nanoscience, Chalmers University of Technology, Kemivägen 9, 412 58, Gothenburg, Sweden.
| |
Collapse
|
16
|
Huynh QS, Holsinger RMD. Fiber and Electrical Field Alignment Increases BDNF Expression in SH-SY5Y Cells following Electrical Stimulation. Pharmaceuticals (Basel) 2023; 16:138. [PMID: 37259290 PMCID: PMC9960882 DOI: 10.3390/ph16020138] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 09/13/2024] Open
Abstract
The limited expression of neurotrophic factors that can be included in neural tissue engineering scaffolds is insufficient for sustained neural regeneration. A localized and sustained method of introducing neurotrophic factors is required. We describe our attempt at inducing neuroblastoma cells to express trophic factors following electrical stimulation. Human SH-SY5Y neuroblastoma cells, cultured on polycaprolactone electrospun nanofibers, were electrically stimulated using a 100 mV/mm electric field. Nuclear morphology and brain-derived neurotrophic factor (BDNF) expression were analyzed. Cells were classified based on the type of fiber orientation and the alignment of these fibers in relation to the electric field. Nuclear deformation was mainly influenced by fiber orientation rather than the electrical field. Similarly, fiber orientation also induced BDNF expression. Although electrical field alone had no significant effect on BDNF expression, combining fiber orientation with electrical field resulted in BDNF expression in cells that grew on electrospun fibers that were aligned perpendicular to the electrical field.
Collapse
Affiliation(s)
- Quy-Susan Huynh
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
17
|
Liu Z, Wei W, Tremblay PL, Zhang T. Electrostimulation of fibroblast proliferation by an electrospun poly (lactide-co-glycolide)/polydopamine/chitosan membrane in a humid environment. Colloids Surf B Biointerfaces 2022; 220:112902. [DOI: 10.1016/j.colsurfb.2022.112902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/29/2022] [Accepted: 10/02/2022] [Indexed: 11/18/2022]
|
18
|
Lee S, M Silva S, Caballero Aguilar LM, Eom T, Moulton SE, Shim BS. Biodegradable bioelectronics for biomedical applications. J Mater Chem B 2022; 10:8575-8595. [PMID: 36214325 DOI: 10.1039/d2tb01475k] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Biodegradable polymers have been widely used in tissue engineering with the potential to be replaced by regenerative tissue. While conventional bionic interfaces are designed to be implanted in living tissue and organs permanently, biocompatible and biodegradable electronic materials are now progressing a paradigm shift towards transient and regenerative bionic engineering. For example, biodegradable bioelectronics can monitor physiologies in a body, transiently rehabilitate disease symptoms, and seamlessly form regenerative interfaces from synthetic electronic devices to tissues by reducing inflammatory foreign-body responses. Conventional electronic materials have not readily been considered biodegradable. However, several strategies have been adopted for designing electroactive and biodegradable materials systems: (1) conductive materials blended with biodegradable components, (2) molecularly engineered conjugated polymers with biodegradable moieties, (3) naturally derived conjugated biopolymers, and (4) aqueously dissolvable metals with encapsulating layers. In this review, we endeavor to present the technical bridges from electrically active and biodegradable material systems to edible and biodegradable electronics as well as transient bioelectronics with pre-clinical bio-instrumental applications, including biodegradable sensors, neural and tissue engineering, and intelligent drug delivery systems.
Collapse
Affiliation(s)
- Seunghyeon Lee
- Program in Biomedical Science & Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon, Republic of Korea. .,Department of Chemical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon, Republic of Korea
| | - Saimon M Silva
- ARC Centre of Excellence for Electromaterials Science, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Victoria 3122, Australia.,Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, Victoria 3122, Australia. .,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia
| | - Lilith M Caballero Aguilar
- ARC Centre of Excellence for Electromaterials Science, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Victoria 3122, Australia.,Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, Victoria 3122, Australia. .,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia
| | - Taesik Eom
- Program in Biomedical Science & Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon, Republic of Korea. .,Department of Chemical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon, Republic of Korea
| | - Simon E Moulton
- ARC Centre of Excellence for Electromaterials Science, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Victoria 3122, Australia.,Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, Victoria 3122, Australia. .,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia
| | - Bong Sup Shim
- Program in Biomedical Science & Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon, Republic of Korea. .,Department of Chemical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon, Republic of Korea
| |
Collapse
|
19
|
Walker JC, Jorgensen AM, Sarkar A, Gent SP, Messerli MA. Anionic polymers amplify electrokinetic perfusion through extracellular matrices. Front Bioeng Biotechnol 2022; 10:983317. [PMID: 36225599 PMCID: PMC9548625 DOI: 10.3389/fbioe.2022.983317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
Electrical stimulation (ES) promotes healing of chronic epidermal wounds and delays degeneration of articular cartilage. Despite electrotherapeutic treatment of these non-excitable tissues, the mechanisms by which ES promotes repair are unknown. We hypothesize that a beneficial role of ES is dependent on electrokinetic perfusion in the extracellular space and that it mimics the effects of interstitial flow. In vivo, the extracellular space contains mixtures of extracellular proteins and negatively charged glycosaminoglycans and proteoglycans surrounding cells. While these anionic macromolecules promote water retention and increase mechanical support under compression, in the presence of ES they should also enhance electro-osmotic flow (EOF) to a greater extent than proteins alone. To test this hypothesis, we compare EOF rates between artificial matrices of gelatin (denatured collagen) with matrices of gelatin mixed with anionic polymers to mimic endogenous charged macromolecules. We report that addition of anionic polymers amplifies EOF and that a matrix comprised of 0.5% polyacrylate and 1.5% gelatin generates EOF with similar rates to those reported in cartilage. The enhanced EOF reduces mortality of cells at lower applied voltage compared to gelatin matrices alone. We also use modeling to describe the range of thermal changes that occur during these electrokinetic experiments and during electrokinetic perfusion of soft tissues. We conclude that the negative charge density of native extracellular matrices promotes electrokinetic perfusion during electrical therapies in soft tissues and may promote survival of artificial tissues and organs prior to vascularization and during transplantation.
Collapse
Affiliation(s)
- Joseph C. Walker
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, United States
| | - Ashley M. Jorgensen
- Department of Mechanical Engineering, South Dakota State University, Brookings, SD, United States
| | - Anyesha Sarkar
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, United States
| | - Stephen P. Gent
- Department of Mechanical Engineering, South Dakota State University, Brookings, SD, United States
| | - Mark A. Messerli
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, United States
- *Correspondence: Mark A. Messerli,
| |
Collapse
|
20
|
Pai VP, Cooper BG, Levin M. Screening Biophysical Sensors and Neurite Outgrowth Actuators in Human Induced-Pluripotent-Stem-Cell-Derived Neurons. Cells 2022; 11:cells11162470. [PMID: 36010547 PMCID: PMC9406775 DOI: 10.3390/cells11162470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/26/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
All living cells maintain a charge distribution across their cell membrane (membrane potential) by carefully controlled ion fluxes. These bioelectric signals regulate cell behavior (such as migration, proliferation, differentiation) as well as higher-level tissue and organ patterning. Thus, voltage gradients represent an important parameter for diagnostics as well as a promising target for therapeutic interventions in birth defects, injury, and cancer. However, despite much progress in cell and molecular biology, little is known about bioelectric states in human stem cells. Here, we present simple methods to simultaneously track ion dynamics, membrane voltage, cell morphology, and cell activity (pH and ROS), using fluorescent reporter dyes in living human neurons derived from induced neural stem cells (hiNSC). We developed and tested functional protocols for manipulating ion fluxes, membrane potential, and cell activity, and tracking neural responses to injury and reinnervation in vitro. Finally, using morphology sensor, we tested and quantified the ability of physiological actuators (neurotransmitters and pH) to manipulate nerve repair and reinnervation. These methods are not specific to a particular cell type and should be broadly applicable to the study of bioelectrical controls across a wide range of combinations of models and endpoints.
Collapse
Affiliation(s)
- Vaibhav P. Pai
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA
| | - Ben G. Cooper
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, MA 02155, USA
- Correspondence:
| |
Collapse
|
21
|
Reinboldt-Jockenhöfer F, Traber J, Liesch G, Bittner C, Benecke U, Dissemond J. Concurrent optical and magnetic stimulation therapy in patients with lower extremity hard-to-heal wounds. J Wound Care 2022; 31:S12-S21. [PMID: 35678774 DOI: 10.12968/jowc.2022.31.sup6.s12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE The treatment of patients with hard-to-heal wounds represents a major multidisciplinary challenge. Therefore, the development and clinical validation of new technologies remains extremely important. The novel application of concurrent optical and magnetic stimulation (COMS) offers a promising noninvasive approach to support physiological wound healing processes, especially in hard-to-heal wounds. METHOD In a multicentre, prospective, comparative, clinical trial, patients with hard-to-heal wounds on lower extremities of different aetiologies were treated with COMS as an adjunct to standard of care (SOC). The primary endpoint was safety; secondary endpoints were wound healing, pain and wound-specific quality of life (Wound-QoL). RESULTS A total of 40 patients were enrolled in this study (intention to treat population (ITTP), n=40). Of these patients, 37 were included in the analysis of the primary endpoint (primary endpoint population, (PEP), n=37). A further subgroup of 30 patients was included in the analysis of the secondary endpoint (secondary endpoint population (SEP), n=30). Finally, the SEP was stratified regarding patients' responsiveness to SOC in an SOC non-responder subgroup (NRSG), n=21, and in an SOC responder subgroup (RSG), n=9. A total of 102 adverse events (AEs) were recorded, of which 96% were 'mild' or 'moderate', and 91% were either a singular or transient event. Only 11 AEs were serious and associated with inpatient treatments unrelated to the studied intervention. In the NRSG, reductions in wound size were found to be statistically significant within the different study periods. Additionally, an acceleration of the healing rate was detected between the baseline and the first four weeks of COMS treatment (p=0.041). The rate of near-complete and complete wound closure in the SEP after 12 weeks were 60% and 43%, respectively. Pain reduction across the treatment group was statistically significant (p≤0.002 for both the SEP and NRSG). The Wound-QoL score improved by 24% during the study (p=0.001). CONCLUSION In this study, COMS treatment for patients with hard-to-heal wounds on lower extremities was a safe and effective novel treatment option, especially for patients who did not respond to SOC.
Collapse
Affiliation(s)
| | - Jürg Traber
- Venenklinik Bellevue, Kreuzlingen, Switzerland
| | | | | | - Ulf Benecke
- Department of Dermatology, Venereology and Allergology, University Hospital, Essen, Germany
| | - Joachim Dissemond
- Department of Dermatology, Venereology and Allergology, University Hospital, Essen, Germany
| |
Collapse
|
22
|
Hlavac N, Bousalis D, Ahmad RN, Pallack E, Vela A, Li Y, Mobini S, Patrick E, Schmidt CE. Effects of Varied Stimulation Parameters on Adipose-Derived Stem Cell Response to Low-Level Electrical Fields. Ann Biomed Eng 2021; 49:3401-3411. [PMID: 34704163 PMCID: PMC10947800 DOI: 10.1007/s10439-021-02875-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 10/04/2021] [Indexed: 11/24/2022]
Abstract
Exogenous electrical fields have been explored in regenerative medicine to increase cellular expression of pro-regenerative growth factors. Adipose-derived stem cells (ASCs) are attractive for regenerative applications, specifically for neural repair. Little is known about the relationship between low-level electrical stimulation (ES) and ASC regenerative potentiation. In this work, patterns of ASC expression and secretion of growth factors (i.e., secretome) were explored across a range of ES parameters. ASCs were stimulated with low-level stimulation (20 mV/mm) at varied pulse frequencies, durations, and with alternating versus direct current. Frequency and duration had the most significant effects on growth factor expression. While a range of stimulation frequencies (1, 20, 1000 Hz) applied intermittently (1 h × 3 days) induced upregulation of general wound healing factors, neural-specific factors were only increased at 1 Hz. Moreover, the most optimal expression of neural growth factors was achieved when ASCs were exposed to 1 Hz pulses continuously for 24 h. In evaluation of secretome, apparent inconsistencies were observed across biological replications. Nonetheless, ASC secretome (from 1 Hz, 24 h ES) caused significant increase in neurite extension compared to non-stimulated control. Overall, ASCs are sensitive to ES parameters at low field strengths, notably pulse frequency and stimulation duration.
Collapse
Affiliation(s)
- Nora Hlavac
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
| | - Deanna Bousalis
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
| | - Raffae N Ahmad
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
| | - Emily Pallack
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
| | - Angelique Vela
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, USA
| | - Yuan Li
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
| | - Sahba Mobini
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
- Instituto de Micro y Nanotecnología, IMN- CNM, CSIC (CEI UAM+CSIC), Tres Cantos, Madrid, Spain
| | - Erin Patrick
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA.
| |
Collapse
|
23
|
Abe Y, Nishizawa M. Electrical aspects of skin as a pathway to engineering skin devices. APL Bioeng 2021; 5:041509. [PMID: 34849444 PMCID: PMC8604566 DOI: 10.1063/5.0064529] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/27/2021] [Indexed: 02/07/2023] Open
Abstract
Skin is one of the indispensable organs for life. The epidermis at the outermost surface provides a permeability barrier to infectious agents, chemicals, and excessive loss of water, while the dermis and subcutaneous tissue mechanically support the structure of the skin and appendages, including hairs and secretory glands. The integrity of the integumentary system is a key for general health, and many techniques have been developed to measure and control this protective function. In contrast, the effective skin barrier is the major obstacle for transdermal delivery and detection. Changes in the electrical properties of skin, such as impedance and ionic activity, is a practical indicator that reflects the structures and functions of the skin. For example, the impedance that reflects the hydration of the skin is measured for quantitative assessment in skincare, and the current generated across a wound is used for the evaluation and control of wound healing. Furthermore, the electrically charged structure of the skin enables transdermal drug delivery and chemical extraction. This paper provides an overview of the electrical aspects of the skin and summarizes current advances in the development of devices based on these features.
Collapse
Affiliation(s)
- Yuina Abe
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki-aza Aoba, Aoba-ku, Sendai 980-8579, Japan
| | - Matsuhiko Nishizawa
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki-aza Aoba, Aoba-ku, Sendai 980-8579, Japan
| |
Collapse
|
24
|
Wang D, Tan J, Zhu H, Mei Y, Liu X. Biomedical Implants with Charge-Transfer Monitoring and Regulating Abilities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004393. [PMID: 34166584 PMCID: PMC8373130 DOI: 10.1002/advs.202004393] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/12/2021] [Indexed: 05/06/2023]
Abstract
Transmembrane charge (ion/electron) transfer is essential for maintaining cellular homeostasis and is involved in many biological processes, from protein synthesis to embryonic development in organisms. Designing implant devices that can detect or regulate cellular transmembrane charge transfer is expected to sense and modulate the behaviors of host cells and tissues. Thus, charge transfer can be regarded as a bridge connecting living systems and human-made implantable devices. This review describes the mode and mechanism of charge transfer between organisms and nonliving materials, and summarizes the strategies to endow implants with charge-transfer regulating or monitoring abilities. Furthermore, three major charge-transfer controlling systems, including wired, self-activated, and stimuli-responsive biomedical implants, as well as the design principles and pivotal materials are systematically elaborated. The clinical challenges and the prospects for future development of these implant devices are also discussed.
Collapse
Affiliation(s)
- Donghui Wang
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
- School of Materials Science and EngineeringHebei University of TechnologyTianjin300130China
| | - Ji Tan
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
| | - Hongqin Zhu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
- Department of Materials ScienceFudan UniversityShanghai200433China
| | - Yongfeng Mei
- Department of Materials ScienceFudan UniversityShanghai200433China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
- School of Chemistry and Materials ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
| |
Collapse
|
25
|
Liu Z, Wan X, Wang ZL, Li L. Electroactive Biomaterials and Systems for Cell Fate Determination and Tissue Regeneration: Design and Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007429. [PMID: 34117803 DOI: 10.1002/adma.202007429] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/19/2020] [Indexed: 06/12/2023]
Abstract
During natural tissue regeneration, tissue microenvironment and stem cell niche including cell-cell interaction, soluble factors, and extracellular matrix (ECM) provide a train of biochemical and biophysical cues for modulation of cell behaviors and tissue functions. Design of functional biomaterials to mimic the tissue/cell microenvironment have great potentials for tissue regeneration applications. Recently, electroactive biomaterials have drawn increasing attentions not only as scaffolds for cell adhesion and structural support, but also as modulators to regulate cell/tissue behaviors and function, especially for electrically excitable cells and tissues. More importantly, electrostimulation can further modulate a myriad of biological processes, from cell cycle, migration, proliferation and differentiation to neural conduction, muscle contraction, embryogenesis, and tissue regeneration. In this review, endogenous bioelectricity and piezoelectricity are introduced. Then, design rationale of electroactive biomaterials is discussed for imitating dynamic cell microenvironment, as well as their mediated electrostimulation and the applying pathways. Recent advances in electroactive biomaterials are systematically overviewed for modulation of stem cell fate and tissue regeneration, mainly including nerve regeneration, bone tissue engineering, and cardiac tissue engineering. Finally, the significance for simulating the native tissue microenvironment is emphasized and the open challenges and future perspectives of electroactive biomaterials are concluded.
Collapse
Affiliation(s)
- Zhirong Liu
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xingyi Wan
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhong Lin Wang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0245, USA
| | - Linlin Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
26
|
Colciago A, Audano M, Bonalume V, Melfi V, Mohamed T, Reid AJ, Faroni A, Greer PA, Mitro N, Magnaghi V. Transcriptomic Profile Reveals Deregulation of Hearing-Loss Related Genes in Vestibular Schwannoma Cells Following Electromagnetic Field Exposure. Cells 2021; 10:cells10071840. [PMID: 34360009 PMCID: PMC8307028 DOI: 10.3390/cells10071840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 12/23/2022] Open
Abstract
Hearing loss (HL) is the most common sensory disorder in the world population. One common cause of HL is the presence of vestibular schwannoma (VS), a benign tumor of the VIII cranial nerve, arising from Schwann cell (SC) transformation. In the last decade, the increasing incidence of VS has been correlated to electromagnetic field (EMF) exposure, which might be considered a pathogenic cause of VS development and HL. Here, we explore the molecular mechanisms underlying the biologic changes of human SCs and/or their oncogenic transformation following EMF exposure. Through NGS technology and RNA-Seq transcriptomic analysis, we investigated the genomic profile and the differential display of HL-related genes after chronic EMF. We found that chronic EMF exposure modified the cell proliferation, in parallel with intracellular signaling and metabolic pathways changes, mostly related to translation and mitochondrial activities. Importantly, the expression of HL-related genes such as NEFL, TPRN, OTOGL, GJB2, and REST appeared to be deregulated in chronic EMF exposure. In conclusion, we suggest that, at a preclinical stage, EMF exposure might promote the transformation of VS cells and contribute to HL.
Collapse
Affiliation(s)
- Alessandra Colciago
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Via G. Balzaretti 9, 20133 Milan, Italy; (A.C.); (M.A.); (V.B.); (V.M.); (T.M.); (N.M.)
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Via G. Balzaretti 9, 20133 Milan, Italy; (A.C.); (M.A.); (V.B.); (V.M.); (T.M.); (N.M.)
| | - Veronica Bonalume
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Via G. Balzaretti 9, 20133 Milan, Italy; (A.C.); (M.A.); (V.B.); (V.M.); (T.M.); (N.M.)
| | - Valentina Melfi
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Via G. Balzaretti 9, 20133 Milan, Italy; (A.C.); (M.A.); (V.B.); (V.M.); (T.M.); (N.M.)
| | - Tasnim Mohamed
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Via G. Balzaretti 9, 20133 Milan, Italy; (A.C.); (M.A.); (V.B.); (V.M.); (T.M.); (N.M.)
| | - Adam J. Reid
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NQ, UK; (A.J.R.); (A.F.)
- Department of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Center, Manchester M13 9NQ, UK
| | - Alessandro Faroni
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NQ, UK; (A.J.R.); (A.F.)
| | - Peter A. Greer
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Via G. Balzaretti 9, 20133 Milan, Italy; (A.C.); (M.A.); (V.B.); (V.M.); (T.M.); (N.M.)
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi di Milano, Via G. Balzaretti 9, 20133 Milan, Italy; (A.C.); (M.A.); (V.B.); (V.M.); (T.M.); (N.M.)
- Correspondence: ; Tel.: +39-0250318414
| |
Collapse
|
27
|
Sarkar A, Messerli MA. Electrokinetic Perfusion Through Three-Dimensional Culture Reduces Cell Mortality. Tissue Eng Part A 2021; 27:1470-1479. [PMID: 33820474 DOI: 10.1089/ten.tea.2021.0008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cell proliferation and survival are dependent on mass transfer. In vivo, fluid flow promotes mass transfer through the vasculature and interstitial space, providing a continuous supply of nutrients and removal of cellular waste products. In the absence of sufficient flow, mass transfer is limited by diffusion and poses significant challenges to cell survival during tissue engineering, tissue transplantation, and treatment of degenerative diseases. Artificial perfusion may overcome these challenges. In this work, we compare the efficacy of pressure driven perfusion (PDP) with electrokinetic perfusion (EKP) toward reducing cell mortality in three-dimensional cultures of Matrigel extracellular matrix. We characterize electro-osmotic flow through Matrigel to identify conditions that generate similar interstitial flow rates to those induced by pressure. We also compare changes in cell mortality induced by continuous or pulsed EKP. We report that continuous EKP significantly reduced mortality throughout the perfusion channels more consistently than PDP at similar flow rates, and pulsed EKP decreased mortality just as effectively as continuous EKP. We conclude that EKP has significant advantages over PDP for promoting tissue survival before neovascularization and angiogenesis. Impact statement Interstitial flow helps promote mass transfer and cell survival in tissues and organs. This study generated interstitial flow using pressure driven perfusion (PDP) or electrokinetic perfusion (EKP) to promote cell viability in three-dimensional cultures. EKP through charged extracellular matrices possesses significant advantages over PDP and may promote cell survival during tissue engineering, transplantations, and treatment of degenerative diseases.
Collapse
Affiliation(s)
- Anyesha Sarkar
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Mark A Messerli
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| |
Collapse
|
28
|
Adler D, Shapira Z, Weiss S, Shainberg A, Katz A. Weak Electromagnetic Fields Accelerate Fusion of Myoblasts. Int J Mol Sci 2021; 22:ijms22094407. [PMID: 33922487 PMCID: PMC8122904 DOI: 10.3390/ijms22094407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 11/28/2022] Open
Abstract
Weak electromagnetic fields (WEF) alter Ca2+ handling in skeletal muscle myotubes. Owing to the involvement of Ca2+ in muscle development, we investigated whether WEF affects fusion of myoblasts in culture. Rat primary myoblast cultures were exposed to WEF (1.75 µT, 16 Hz) for up to six days. Under control conditions, cell fusion and creatine kinase (CK) activity increased in parallel and peaked at 4–6 days. WEF enhanced the extent of fusion after one and two days (by ~40%) vs. control, but not thereafter. Exposure to WEF also enhanced CK activity after two days (almost four-fold), but not afterwards. Incorporation of 3H-thymidine into DNA was enhanced by one-day exposure to WEF (~40%), indicating increased cell replication. Using the potentiometric fluorescent dye di-8-ANEPPS, we found that exposure of cells to 150 mM KCl resulted in depolarization of the cell membrane. However, prior exposure of cells to WEF for one day followed by addition of KCl resulted in hyperpolarization of the cell membrane. Acute exposure of cells to WEF also resulted in hyperpolarization of the cell membrane. Twenty-four hour incubation of myoblasts with gambogic acid, an inhibitor of the inward rectifying K+ channel 2.1 (Kir2.1), did not affect cell fusion, WEF-mediated acceleration of fusion or hyperpolarization. These data demonstrate that WEF accelerates fusion of myoblasts, resulting in myotube formation. The WEF effect is associated with hyperpolarization but WEF does not appear to mediate its effects on fusion by activating Kir2.1 channels.
Collapse
Affiliation(s)
- Dana Adler
- Faculty of Life Sciences, Bar Ilan University, Ramat Gan 52900, Israel; (D.A.); (A.S.)
| | - Zehavit Shapira
- Department of Physics, Bar Ilan University, Ramat Gan 52900, Israel; (Z.S.); (S.W.)
| | - Shimon Weiss
- Department of Physics, Bar Ilan University, Ramat Gan 52900, Israel; (Z.S.); (S.W.)
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Asher Shainberg
- Faculty of Life Sciences, Bar Ilan University, Ramat Gan 52900, Israel; (D.A.); (A.S.)
| | - Abram Katz
- Åstrand Laboratory of Work Physiology, The Swedish School of Sport and Health Sciences, GIH, Box 5626, SE-114 86 Stockholm, Sweden
- Correspondence:
| |
Collapse
|
29
|
Yi C, Spitters TWGM, Al-Far EADA, Wang S, Xiong T, Cai S, Yan X, Guan K, Wagner M, El-Armouche A, Antos CL. A calcineurin-mediated scaling mechanism that controls a K +-leak channel to regulate morphogen and growth factor transcription. eLife 2021; 10:e60691. [PMID: 33830014 PMCID: PMC8110307 DOI: 10.7554/elife.60691] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 04/07/2021] [Indexed: 01/10/2023] Open
Abstract
The increase in activity of the two-pore potassium-leak channel Kcnk5b maintains allometric juvenile growth of adult zebrafish appendages. However, it remains unknown how this channel maintains allometric growth and how its bioelectric activity is regulated to scale these anatomical structures. We show the activation of Kcnk5b is sufficient to activate several genes that are part of important development programs. We provide in vivo transplantation evidence that the activation of gene transcription is cell autonomous. We also show that Kcnk5b will induce the expression of different subsets of the tested developmental genes in different cultured mammalian cell lines, which may explain how one electrophysiological stimulus can coordinately regulate the allometric growth of diverse populations of cells in the fin that use different developmental signals. We also provide evidence that the post-translational modification of serine 345 in Kcnk5b by calcineurin regulates channel activity to scale the fin. Thus, we show how an endogenous bioelectric mechanism can be regulated to promote coordinated developmental signaling to generate and scale a vertebrate appendage.
Collapse
Affiliation(s)
- Chao Yi
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of SciencesShanghaiChina
| | - Tim WGM Spitters
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
| | | | - Sen Wang
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of SciencesShanghaiChina
| | - TianLong Xiong
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of SciencesShanghaiChina
| | - Simian Cai
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
| | - Xin Yan
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
| | - Kaomei Guan
- Institut für Pharmakologie und Toxikologie, Technische Universität DresdenDresdenGermany
| | - Michael Wagner
- Institut für Pharmakologie und Toxikologie, Technische Universität DresdenDresdenGermany
- Klinik für Innere Medizin und Kardiologie, Herzzentrum Dresden, Technische Universität DresdenDresdenGermany
| | - Ali El-Armouche
- Institut für Pharmakologie und Toxikologie, Technische Universität DresdenDresdenGermany
| | - Christopher L Antos
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
- Institut für Pharmakologie und Toxikologie, Technische Universität DresdenDresdenGermany
| |
Collapse
|
30
|
Montoya C, Du Y, Gianforcaro AL, Orrego S, Yang M, Lelkes PI. On the road to smart biomaterials for bone research: definitions, concepts, advances, and outlook. Bone Res 2021; 9:12. [PMID: 33574225 PMCID: PMC7878740 DOI: 10.1038/s41413-020-00131-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 11/16/2020] [Accepted: 11/20/2020] [Indexed: 01/31/2023] Open
Abstract
The demand for biomaterials that promote the repair, replacement, or restoration of hard and soft tissues continues to grow as the population ages. Traditionally, smart biomaterials have been thought as those that respond to stimuli. However, the continuous evolution of the field warrants a fresh look at the concept of smartness of biomaterials. This review presents a redefinition of the term "Smart Biomaterial" and discusses recent advances in and applications of smart biomaterials for hard tissue restoration and regeneration. To clarify the use of the term "smart biomaterials", we propose four degrees of smartness according to the level of interaction of the biomaterials with the bio-environment and the biological/cellular responses they elicit, defining these materials as inert, active, responsive, and autonomous. Then, we present an up-to-date survey of applications of smart biomaterials for hard tissues, based on the materials' responses (external and internal stimuli) and their use as immune-modulatory biomaterials. Finally, we discuss the limitations and obstacles to the translation from basic research (bench) to clinical utilization that is required for the development of clinically relevant applications of these technologies.
Collapse
Affiliation(s)
- Carolina Montoya
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA
| | - Yu Du
- Department of Endodontology, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA
- Guangdong Provincial Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatological Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Anthony L Gianforcaro
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA, 19122, USA
| | - Santiago Orrego
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA, 19122, USA
| | - Maobin Yang
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA
- Department of Endodontology, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA, 19122, USA
| | - Peter I Lelkes
- Department of Endodontology, Kornberg School of Dentistry, Temple University, Philadelphia, PA, 19140, USA.
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA, 19122, USA.
| |
Collapse
|
31
|
Caramazza L, De Angelis A, Remondini D, Castellani G, Liberti M, Apollonio F, Zironi I. Galvanotactic Phenomenon Induced by Non-Contact Electrostatic Field: Investigation in a Scratch Assay. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:2520-2523. [PMID: 33018519 DOI: 10.1109/embc44109.2020.9175695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Non-contact galvanotaxis as a way to drive the cells migration could be a promising tool for a variety of biomedical applications, such as wound healing control, avoiding the interaction between electrodes and cell cultures. To this regard, the efficacy of this electrical stimulus application has to be deeper studied to control physiological migratory phenomena in a remote way.Aim of this work is to provide an experimental investigation on the mobility of cells exposed to a static electric field in a "noncontact" mode, supported by a suitable modeling of the electric field distribution inside the experimental setup. In particular, scratch assays have been carried out placing the electrodes outside the cells medium support and changing the cells holder to study more than one configuration.Clinical Relevance- In this study the in vitro experiments on the non-contact galvanotaxis, together with the numerical simulations of the exposure setup, provide a way to investigate the effects that could affect an electrically drive cell migration.
Collapse
|
32
|
Konstantinou E, Zagoriti Z, Pyriochou A, Poulas K. Microcurrent Stimulation Triggers MAPK Signaling and TGF-β1 Release in Fibroblast and Osteoblast-Like Cell Lines. Cells 2020; 9:E1924. [PMID: 32825091 PMCID: PMC7564311 DOI: 10.3390/cells9091924] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/08/2020] [Accepted: 08/18/2020] [Indexed: 12/26/2022] Open
Abstract
Wound healing constitutes an essential process for all organisms and involves a sequence of three phases. The disruption or elongation of any of these phases can lead to a chronic or non-healing wound. Electrical stimulation accelerates wound healing by mimicking the current that is generated in the skin after any injury. Here, we sought to identify the molecular mechanisms involved in the healing process following in vitro microcurrent stimulation-a type of electrotherapy. Our results concluded that microcurrents promote cell proliferation and migration in an ERK 1/2- or p38-dependent way. Furthermore, microcurrents induce the secretion of transforming growth factor-beta-1 (TGF-β1) in fibroblasts and osteoblast-like cells. Interestingly, transcriptomic analysis uncovered that microcurrents enhance the transcriptional activation of genes implicated in Hedgehog, TGF-β1 and MAPK signaling pathways. Overall, our results demonstrate that microcurrents may enhance wound closure through a combination of signal transductions, via MAPK's phosphorylation, and the transcriptional activation of specific genes involved in the healing process. These mechanisms should be further examined in vivo, in order to verify the beneficial effects of microcurrents in wound or fracture healing.
Collapse
Affiliation(s)
| | | | | | - Konstantinos Poulas
- Laboratory of Molecular Biology and Immunology, Department of Pharmacy, University of Patras, 26504 Rio, Greece; (E.K.); (Z.Z.); (A.P.)
| |
Collapse
|
33
|
O'Clock GD, Strouse Watt W. A Model and Simulation With Therapeutic Device-Protocol Design Implications for Acute and Chronic Wounds. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:4967-4973. [PMID: 33019102 DOI: 10.1109/embc44109.2020.9175325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
An understanding of healing processes for different tissues and organs, along with the development of appropriate therapeutic devices and treatment protocols, requires an appreciation for the mechanisms-of-action and sequencing of many interconnected chemical, electrical, mechanical, and optical activities. Unfortunately, the substantial contributions that endogenous electrical mechanisms-of-action provide in healing and regulation are often overlooked, resulting in a poor transfer of knowledge from science, to engineering, and finally, to therapy. The wide variety of healing processes, their therapeutic implications, and the devices and protocol designs that are most effective cannot be understood or addressed adequately without an understanding of the endogenous electrical mechanisms-of-action associated with wound healing. Achieving this level of understanding can be enhanced by the use of appropriate models and simulations that are based on physiological/biochemical system response characteristics.
Collapse
|
34
|
Brief Electrical Stimulation Triggers an Effective Regeneration of Leech CNS. eNeuro 2020; 7:ENEURO.0030-19.2020. [PMID: 32471846 PMCID: PMC7317182 DOI: 10.1523/eneuro.0030-19.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 01/01/2023] Open
Abstract
The search for therapeutic strategies to promote neuronal regeneration following injuries toward functional recovery is of great importance. Brief low-frequency electrical stimulation (ES) has been reported as a useful method to improve neuronal regeneration in different animal models; however, the effect of ES on single neuron behavior has not been shown. Here, we study the effect of brief ES on neuronal regeneration of the CNS of adult medicinal leeches. Studying the regeneration of selected sets of identified neurons allow us to quantify the ES effect per cell type at the single-cell level. Chains of the CNS that were subjected to cut injury were observed for 3 d, and the spontaneous regeneration was compared with the electrically stimulated injured chains. We show that the ES improves the efficiency of regeneration of Retzius cells, as larger masses of the total branching tree traverse the injury site with better directed growth with no effect on the average branching tree length. No antero-posterior polarity was found along regeneration within the leech CNS. Moreover, the microglial cell distribution was examined revealing more microglial cells in proximity to the stimulation site compared with non-stimulated. Our results lay a foundation for future ES-based neuroregenerative therapies.
Collapse
|
35
|
Electric Phenomenon: A Disregarded Tool in Tissue Engineering and Regenerative Medicine. Trends Biotechnol 2020; 38:24-49. [DOI: 10.1016/j.tibtech.2019.07.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 02/08/2023]
|
36
|
Physiological strength electric fields modulate human T cell activation and polarisation. Sci Rep 2019; 9:17604. [PMID: 31772211 PMCID: PMC6879562 DOI: 10.1038/s41598-019-53898-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 10/31/2019] [Indexed: 12/24/2022] Open
Abstract
The factors and signals driving T cell activation and polarisation during immune responses have been studied mainly at the level of cells and chemical mediators. Here we describe a physical driver of these processes in the form of physiological-strength electric fields (EFs). EFs are generated at sites where epithelium is disrupted (e.g. wounded skin/bronchial epithelia) and where T cells frequently are present. Using live-cell imaging, we show human primary T cells migrate directionally to the cathode in low strength (50/150 mV/mm) EFs. Strikingly, we show for the first time that EFs significantly downregulate T cell activation following stimulation with antigen-activated APCs or anti-CD3/CD28 antibodies, as demonstrated by decreased IL-2 secretion and proliferation. These EF-induced functional changes were accompanied by a significant dampening of CD4+ T cell polarisation. Expression of critical markers of the Th17 lineage, RORγt and IL-17, and the Th17 polarisation mediator phospho-STAT3 were reduced significantly, while STAT1, ERK and c-Jun phosphorylation were comparatively unaffected suggesting STAT3 modulation by EFs as one mechanism driving effects. Overall, we identify electrical signals as important contributors to the co-ordination and regulation of human T cell functions, paving the way for a new research area into effects of naturally occurring and clinically-applied EFs in conditions where control of T cell activity is paramount.
Collapse
|
37
|
Electromigration of cell surface macromolecules in DC electric fields during cell polarization and galvanotaxis. J Theor Biol 2019; 478:58-73. [DOI: 10.1016/j.jtbi.2019.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022]
|
38
|
Wireless Direct Microampere Current in Wound Healing: Clinical and Immunohistological Data from Two Single Case Reports. BIOSENSORS-BASEL 2019; 9:bios9030107. [PMID: 31492004 PMCID: PMC6784371 DOI: 10.3390/bios9030107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/01/2019] [Accepted: 09/02/2019] [Indexed: 01/22/2023]
Abstract
Chronic pressure ulcers are hard-to-heal wounds that decrease the patient’s quality of life. Wireless Micro Current Stimulation (WMCS) is an innovative, non-invasive, similar to electrode-based electrostimulation (ES) technology, that generates and transfers ions that are negatively-charged to the injured tissue, using accessible air gases as a transfer medium. WMCS is capable of generating similar tissue potentials, as electrode-based ES, for injured tissue. Here, through immunohistochemistry, we intended to characterize the induced tissue healing biological mechanisms that occur during WMCS therapy. Two single cases of bedridden due to serious stroke white men with chronic non-healing pressure ulcers have been treated with WMCS technology. WMCS suppresses inflammatory responses by decreasing the aggregation of granulocytes, followed by stimulating myofibroblastic activity and a new formation of collagen fibers, as depicted by immunohistochemistry. As a result, WMCS provides a special adjunct or stand-alone therapy choice for chronic and non-healing injuries, similar to electrode-based ES, but with added (i.e., contactless) benefits towards its establishment as a routine clinical wound healing regime.
Collapse
|
39
|
Abstract
As the leading cause of death in cancer, there is an urgent need to develop treatments to target the dissemination of primary tumor cells to secondary organs, known as metastasis. Bioelectric signaling has emerged in the last century as an important controller of cell growth, and with the development of current molecular tools we are now beginning to identify its role in driving cell migration and metastasis in a variety of cancer types. This review summarizes the currently available research for bioelectric signaling in solid tumor metastasis. We review the steps of metastasis and discuss how these can be controlled by bioelectric cues at the level of a cell, a population of cells, and the tissue. The role of ion channel, pump, and exchanger activity and ion flux is discussed, along with the importance of the membrane potential and the relationship between ion flux and membrane potential. We also provide an overview of the evidence for control of metastasis by external electric fields (EFs) and draw from examples in embryogenesis and regeneration to discuss the implications for endogenous EFs. By increasing our understanding of the dynamic properties of bioelectric signaling, we can develop new strategies that target metastasis to be translated into the clinic.
Collapse
Affiliation(s)
- Samantha L. Payne
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, Massachusetts
| | - Madeleine J. Oudin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
40
|
Jenkins TL, Little D. Synthetic scaffolds for musculoskeletal tissue engineering: cellular responses to fiber parameters. NPJ Regen Med 2019; 4:15. [PMID: 31263573 PMCID: PMC6597555 DOI: 10.1038/s41536-019-0076-5] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Tissue engineering often uses synthetic scaffolds to direct cell responses during engineered tissue development. Since cells reside within specific niches of the extracellular matrix, it is important to understand how the matrix guides cell response and then incorporate this knowledge into scaffold design. The goal of this review is to review elements of cell-matrix interactions that are critical to informing and evaluating cellular response on synthetic scaffolds. Therefore, this review examines fibrous proteins of the extracellular matrix and their effects on cell behavior, followed by a discussion of the cellular responses elicited by fiber diameter, alignment, and scaffold porosity of two dimensional (2D) and three dimensional (3D) synthetic scaffolds. Variations in fiber diameter, alignment, and scaffold porosity guide stem cells toward different lineages. Cells generally exhibit rounded morphology on nanofibers, randomly oriented fibers, and low-porosity scaffolds. Conversely, cells exhibit elongated, spindle-shaped morphology on microfibers, aligned fibers, and high-porosity scaffolds. Cells migrate with higher velocities on nanofibers, aligned fibers, and high-porosity scaffolds but migrate greater distances on microfibers, aligned fibers, and highly porous scaffolds. Incorporating relevant biomimetic factors into synthetic scaffolds destined for specific tissue application could take advantage of and further enhance these responses.
Collapse
Affiliation(s)
- Thomas Lee Jenkins
- Department of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
| | - Dianne Little
- Department of Biomedical Engineering, Purdue University, West Lafayette, IN 47907 USA
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907 USA
| |
Collapse
|
41
|
Cunha F, Rajnicek AM, McCaig CD. Electrical Stimulation Directs Migration, Enhances and Orients Cell Division and Upregulates the Chemokine Receptors CXCR4 and CXCR2 in Endothelial Cells. J Vasc Res 2019; 56:39-53. [PMID: 30995642 DOI: 10.1159/000495311] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 11/09/2018] [Indexed: 11/19/2022] Open
Abstract
Natural direct current electric fields (DC EFs) within tissues undergoing angiogenesis have the potential to influence vessel formation, but how they affect endothelial cells is not clear. We therefore quantified behaviours of human umbilical vein endothelial cells (HUVEC) and human microvasculature endothelial cells (HMEC) stimulated by EFsin vitro. Both cell types migrated faster and toward the cathode; HUVECs responded to fields as low as 50mV/mm, but the HMEC threshold was 100 mV/mm. Mitosis was stimulated at 50 mV/mm for HMEC and at 150 mV/mm for HUVECs, but the cleavage plane was oriented orthogonal to the field vector at 200 mV/mm for both cell types. That different field strengths induced different cell responses suggests distinct underlying cellular mechanisms. A physiological electric field also upregulated expression of CXCR4 and CXCR2 chemokine receptors and upregulated phosphorylation of both chemokines in HUVEC and HMEC cells. Evidence that DC EFs direct endothelial cell migration, proliferation and upregulate chemokines involved in wound healing suggests a key role for electrical control of capillary production during healing. Our data contribute to the molecular mechanisms by which DC EFs direct endothelial cell behaviour and present a novel signalling paradigm in wound healing, tissue regeneration and angiogenesis-related diseases.
Collapse
Affiliation(s)
- Filipa Cunha
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom,
| | - Ann M Rajnicek
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Colin D McCaig
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
42
|
Mobini S, Song YH, McCrary MW, Schmidt CE. Advances in ex vivo models and lab-on-a-chip devices for neural tissue engineering. Biomaterials 2019; 198:146-166. [PMID: 29880219 PMCID: PMC6957334 DOI: 10.1016/j.biomaterials.2018.05.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/25/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023]
Abstract
The technologies related to ex vivo models and lab-on-a-chip devices for studying the regeneration of brain, spinal cord, and peripheral nerve tissues are essential tools for neural tissue engineering and regenerative medicine research. The need for ex vivo systems, lab-on-a-chip technologies and disease models for neural tissue engineering applications are emerging to overcome the shortages and drawbacks of traditional in vitro systems and animal models. Ex vivo models have evolved from traditional 2D cell culture models to 3D tissue-engineered scaffold systems, bioreactors, and recently organoid test beds. In addition to ex vivo model systems, we discuss lab-on-a-chip devices and technologies specifically for neural tissue engineering applications. Finally, we review current commercial products that mimic diseased and normal neural tissues, and discuss the future directions in this field.
Collapse
Affiliation(s)
- Sahba Mobini
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Young Hye Song
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Michaela W McCrary
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
43
|
Bacova M, Bimbova K, Fedorova J, Lukacova N, Galik J. Epidural oscillating field stimulation as an effective therapeutic approach in combination therapy for spinal cord injury. J Neurosci Methods 2019; 311:102-110. [PMID: 30339879 DOI: 10.1016/j.jneumeth.2018.10.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Traumatic spinal cord injury (SCI) causes partial or total loss of sensory and motor functions. Despite enormous efforts, there is still no effective treatment which might improve patients' neurological status.The application of electric current to the injured spinal cord is known to promote healing and tissue regeneration. The use of this modality in treating the injured spinal cord to improve neurological recovery has been introduced as a potential treatment. NEW METHOD Here we describe the method of epidural implantation of a miniature oscillating field (OF) stimulator designed in our laboratory immediately after Th9 spinal compression in Wistar rats. Three groups of animals were analyzed (intact; SCI only; OFS + SCI; n = 8 each). Histological, immunohistological and behavioral analysis were used to show the favorable effect of epidural OF stimulation on axonal regeneration and modulation of astrogliosis. RESULTS Our study revealed considerable differences in white matter integrity in animals with an implanted OF stimulator. Moreover, we detected significantly increased numbers of neurofilaments and massive reduction in activated forms of astrocytes in the group of stimulated animals compared to the animals without stimulation. COMPARISON WITH EXISTING METHOD(S) Compared with previous research, our study revealed that epidural implantation of an OF stimulator immediately after spinal compression effectively reduced the expression of inflammatory response and suppressed activated astrocyte formation. CONCLUSIONS Our finding confirms that implanting an OF stimulator is safe, stable and suitable for future combined therapy which could effectively promote and accelerate regeneration and functional restoration after spinal trauma.
Collapse
Affiliation(s)
- Maria Bacova
- Institute of Neurobiology of Biomedical Research Center Slovak Academy of Sciences, Soltesovej 4, 040 01 Kosice, Slovakia
| | - Katarina Bimbova
- Institute of Neurobiology of Biomedical Research Center Slovak Academy of Sciences, Soltesovej 4, 040 01 Kosice, Slovakia
| | - Jana Fedorova
- Institute of Neurobiology of Biomedical Research Center Slovak Academy of Sciences, Soltesovej 4, 040 01 Kosice, Slovakia
| | - Nadezda Lukacova
- Institute of Neurobiology of Biomedical Research Center Slovak Academy of Sciences, Soltesovej 4, 040 01 Kosice, Slovakia
| | - Jan Galik
- Institute of Neurobiology of Biomedical Research Center Slovak Academy of Sciences, Soltesovej 4, 040 01 Kosice, Slovakia.
| |
Collapse
|
44
|
Yan T, Jiang X, Lin G, Tang D, Zhang J, Guo X, Zhang D, Zhang Q, Jia J, Huang Y. Autophagy is required for the directed motility of keratinocytes driven by electric fields. FASEB J 2018; 33:3922-3935. [PMID: 30509146 DOI: 10.1096/fj.201801294r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Endogenous wound electric fields (EFs), an important and fundamental occurrence of wound healing, profoundly influence the directed migration of keratinocytes. Although numerous studies have unveiled the signals responsible for EF-biased direction, the mechanisms by which EFs promote keratinocyte motility remains to be elucidated. In our study, EFs enhanced the directed migratory speed of keratinocytes by inducing autophagic activity, thereby facilitating skin barrier restoration. Initially, we found that electrical signals directed keratinocytes to the cathode with enhanced motility parameters [ i.e., trajectory distance, trajectory speed, displacement distance, and displacement speed ( Td/ t)] and more efficient migration (directionality and Td/ t along the x axis, among others). Meanwhile, EFs induced a time-dependent increase in autophagic activity in keratinocytes, with constant autophagic flux, accompanied by increased transcription of numerous autophagy-related genes. Deficiency in Atg5, a key protein necessary for autophagosome formation, led to significant reduction of autophagy, which was accompanied by a substantial reduction in EF-stimulated directed motility. These results demonstrated a causal relationship between autophagy and EF-directed migratory speed. In addition, both cell migration under normal conditions and EF-biased directionality were autophagy independent. Thus, our findings define autophagy as an important functional regulator of electrically enhanced directed motility, adding to a growing understanding of EFs.-Yan, T., Jiang, X., Lin, G., Tang, D., Zhang, J., Guo, X., Zhang, D., Zhang, Q., Jia, J., Huang, Y. Autophagy is required for the directed motility of keratinocytes driven by electric fields.
Collapse
Affiliation(s)
- Tiantian Yan
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Military Burn Center, the 990th (159th) Hospital of the People's Liberation Army, Zhumadian, China
| | - Xupin Jiang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guoan Lin
- Military Burn Center, the 990th (159th) Hospital of the People's Liberation Army, Zhumadian, China
| | - Di Tang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Junhui Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaowei Guo
- Department of Burns and Plastic Surgery, the 205th Hospital of the People's Liberation Army, Jinzhou, China
| | - Dongxia Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qiong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jiezhi Jia
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuesheng Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns, and Combined Injury, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
45
|
Ashrafi M, Novak-Frazer L, Morris J, Baguneid M, Rautemaa-Richardson R, Bayat A. Electrical stimulation disrupts biofilms in a human wound model and reveals the potential for monitoring treatment response with volatile biomarkers. Wound Repair Regen 2018; 27:5-18. [DOI: 10.1111/wrr.12679] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/08/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Mohammed Ashrafi
- Plastic & Reconstructive Surgery Research, Division of Musculoskeletal & Dermatological Sciences; School of Biological Sciences, University of Manchester; Manchester United Kingdom
- Manchester University NHS Foundation Trust; Wythenshawe Hospital; Manchester United Kingdom
- Bioengineering Group, School of Materials; University of Manchester; Manchester United Kingdom
| | - Lilyann Novak-Frazer
- Manchester University NHS Foundation Trust; Wythenshawe Hospital; Manchester United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Division of Infection, Immunity and Respiratory Medicine; School of Biological Sciences, The University of Manchester and Manchester University NHS Foundation Trust; Manchester United Kingdom
| | - Julie Morris
- Honorary Reader in Medical Statistics; Manchester University NHS Foundation Trust, Wythenshawe Hospital; Manchester United Kingdom
| | - Mohamed Baguneid
- Manchester University NHS Foundation Trust; Wythenshawe Hospital; Manchester United Kingdom
| | - Riina Rautemaa-Richardson
- Manchester University NHS Foundation Trust; Wythenshawe Hospital; Manchester United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Division of Infection, Immunity and Respiratory Medicine; School of Biological Sciences, The University of Manchester and Manchester University NHS Foundation Trust; Manchester United Kingdom
| | - Ardeshir Bayat
- Plastic & Reconstructive Surgery Research, Division of Musculoskeletal & Dermatological Sciences; School of Biological Sciences, University of Manchester; Manchester United Kingdom
- Manchester University NHS Foundation Trust; Wythenshawe Hospital; Manchester United Kingdom
| |
Collapse
|
46
|
Reprogramming the Stem Cell Behavior by Shear Stress and Electric Field Stimulation: Lab-on-a-Chip Based Biomicrofluidics in Regenerative Medicine. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018. [DOI: 10.1007/s40883-018-0071-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
47
|
Growing Neural PC-12 Cell on Crosslinked Silica Aerogels Increases Neurite Extension in the Presence of an Electric Field. J Funct Biomater 2018; 9:jfb9020030. [PMID: 29677113 PMCID: PMC6023435 DOI: 10.3390/jfb9020030] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/11/2018] [Accepted: 04/18/2018] [Indexed: 12/27/2022] Open
Abstract
Externally applied electrical stimulation (ES) has been shown to enhance the nerve regeneration process and to influence the directionality of neurite outgrowth. In addition, the physical and chemical properties of the substrate used for nerve-cell regeneration is critical in fostering regeneration. Previously, we have shown that polyurea-crosslinked silica aerogels (PCSA) exert a positive influence on the extension of neurites by PC-12 cells, a cell-line model widely used to study neurite extension and electrical excitability. In this work, we have examined how an externally applied electric field (EF) influences the extension of neurites in PC-12 cells grown on two substrates: collagen-coated dishes versus collagen-coated crosslinked silica aerogels. The externally applied direct current (DC) bias was applied in vitro using a custom-designed chamber containing polydimethysiloxane (PDMS) embedded copper electrodes to create an electric field across the substrate for the cultured PC-12 cells. Results suggest orientation preference towards the anode, and, on average, longer neurites in the presence of the applied DC bias than with 0 V DC bias. In addition, neurite length was increased in cells grown on silica-crosslinked aerogel when compared to cells grown on regular petri-dishes. These results further support the notion that PCSA is a promising material for nerve regeneration.
Collapse
|
48
|
Kobylkevich BM, Sarkar A, Carlberg BR, Huang L, Ranjit S, Graham DM, Messerli MA. Reversing the direction of galvanotaxis with controlled increases in boundary layer viscosity. Phys Biol 2018; 15:036005. [PMID: 29412191 DOI: 10.1088/1478-3975/aaad91] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Weak external electric fields (EFs) polarize cellular structure and direct most migrating cells (galvanotaxis) toward the cathode, making it a useful tool during tissue engineering and for healing epidermal wounds. However, the biophysical mechanisms for sensing weak EFs remain elusive. We have reinvestigated the mechanism of cathode-directed water flow (electro-osmosis) in the boundary layer of cells, by reducing it with neutral, viscous polymers. We report that increasing viscosity with low molecular weight polymers decreases cathodal migration and promotes anodal migration in a concentration dependent manner. In contrast, increased viscosity with high molecular weight polymers does not affect directionality. We explain the contradictory results in terms of porosity and hydraulic permeability between the polymers rather than in terms of bulk viscosity. These results provide the first evidence for controlled reversal of galvanotaxis using viscous agents and position the field closer to identifying the putative electric field receptor, a fundamental, outside-in signaling receptor that controls cellular polarity for different cell types.
Collapse
Affiliation(s)
- Brian M Kobylkevich
- Department of Biology and Microbiology, South Dakota State University, Brookings, SD, United States of America. Brian Kobylkevich and Anyesha Sarkar contributed equally to this work
| | | | | | | | | | | | | |
Collapse
|
49
|
Pazmino Betancourt BA, Florczyk SJ, Simon M, Juba D, Douglas JF, Keyrouz W, Bajcsy P, Lee C, Simon CG. Effect of the scaffold microenvironment on cell polarizability and capacitance determined by probabilistic computations. Biomed Mater 2018; 13:025012. [PMID: 29072579 PMCID: PMC5815922 DOI: 10.1088/1748-605x/aa9650] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In living systems, it is frequently stated that form follows function by virtue of evolutionary pressures on organism development, but in the study of how functions emerge at the cellular level, function often follows form. We study this chicken versus egg problem of emergent structure-property relationships in living systems in the context of primary human bone marrow stromal cells cultured in a variety of microenvironments that have been shown to cause distinct patterns of cell function and differentiation. Through analysis of a publicly available catalog of three-dimensional (3D) cell shape data, we introduce a family of metrics to characterize the 'form' of the cell populations that emerge from a variety of diverse microenvironments. In particular, measures of form are considered that are expected to have direct significance for cell function, signaling and metabolic activity: dimensionality, polarizability and capacitance. Dimensionality was assessed by an intrinsic measure of cell shape obtained from the polarizability tensor. This tensor defines ellipsoids for arbitrary cell shapes and the thinnest dimension of these ellipsoids, P 1, defines a reference minimal scale for cells cultured in a 3D microenvironment. Polarizability governs the electric field generated by a cell, and determines the cell's ability to detect electric fields. Capacitance controls the shape dependence of the rate at which diffusing molecules contact the surface of the cell, and this has great significance for inter-cellular signaling. These results invite new approaches for designing scaffolds which explicitly direct cell dimensionality, polarizability and capacitance to guide the emergence of new cell functions derived from the acquired form.
Collapse
Affiliation(s)
- Beatriz A. Pazmino Betancourt
- Materials Science and Engineering Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD, 20899, USA
| | - Stephen J. Florczyk
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD, 20899, USA
- Department of Materials Science & Engineering, University of Central Florida, 12760 Pegasus Drive, Orlando, FL 32816, USA
| | - Mylene Simon
- Software and Systems Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD, 20899, USA
| | - Derek Juba
- Software and Systems Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD, 20899, USA
| | - Jack F. Douglas
- Materials Science and Engineering Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD, 20899, USA
| | - Walid Keyrouz
- Software and Systems Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD, 20899, USA
| | - Peter Bajcsy
- Software and Systems Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD, 20899, USA
| | - Christopher Lee
- Materials Science and Engineering Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD, 20899, USA
| | - Carl G. Simon
- Biosystems and Biomaterials Division, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD, 20899, USA
| |
Collapse
|
50
|
Sella S, Adami V, Amati E, Bernardi M, Chieregato K, Gatto P, Menarin M, Pozzato A, Pozzato G, Astori G. In-vitro analysis of Quantum Molecular Resonance effects on human mesenchymal stromal cells. PLoS One 2018; 13:e0190082. [PMID: 29293552 PMCID: PMC5749755 DOI: 10.1371/journal.pone.0190082] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 12/07/2017] [Indexed: 01/08/2023] Open
Abstract
Electromagnetic fields play an essential role in cellular functions interfering with cellular pathways and tissue physiology. In this context, Quantum Molecular Resonance (QMR) produces waves with a specific form at high-frequencies (4–64 MHz) and low intensity through electric fields. We evaluated the effects of QMR stimulation on bone marrow derived mesenchymal stromal cells (MSC). MSC were treated with QMR for 10 minutes for 4 consecutive days for 2 weeks at different nominal powers. Cell morphology, phenotype, multilineage differentiation, viability and proliferation were investigated. QMR effects were further investigated by cDNA microarray validated by real-time PCR. After 1 and 2 weeks of QMR treatment morphology, phenotype and multilineage differentiation were maintained and no alteration of cellular viability and proliferation were observed between treated MSC samples and controls. cDNA microarray analysis evidenced more transcriptional changes on cells treated at 40 nominal power than 80 ones. The main enrichment lists belonged to development processes, regulation of phosphorylation, regulation of cellular pathways including metabolism, kinase activity and cellular organization. Real-time PCR confirmed significant increased expression of MMP1, PLAT and ARHGAP22 genes while A2M gene showed decreased expression in treated cells compared to controls. Interestingly, differentially regulated MMP1, PLAT and A2M genes are involved in the extracellular matrix (ECM) remodelling through the fibrinolytic system that is also implicated in embryogenesis, wound healing and angiogenesis. In our model QMR-treated MSC maintained unaltered cell phenotype, viability, proliferation and the ability to differentiate into bone, cartilage and adipose tissue. Microarray analysis may suggest an involvement of QMR treatment in angiogenesis and in tissue regeneration probably through ECM remodelling.
Collapse
Affiliation(s)
- Sabrina Sella
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
| | - Valentina Adami
- High Throughput Screening Core Facility, Center for Integrative Biology, University of Trento, Trento, Italy
| | - Eliana Amati
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
| | - Martina Bernardi
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
- Hematology Project Foundation, Vicenza, Italy
| | - Katia Chieregato
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
- Hematology Project Foundation, Vicenza, Italy
| | - Pamela Gatto
- High Throughput Screening Core Facility, Center for Integrative Biology, University of Trento, Trento, Italy
| | - Martina Menarin
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
| | | | | | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Hematology Unit, Vicenza Hospital, Vicenza, Italy
- * E-mail:
| |
Collapse
|