1
|
An C, Zhao Y, Guo L, Zhang Z, Yan C, Zhang S, Zhang Y, Shao F, Qi Y, wang X, Wang H, Zhang L. Innovative approaches to boost mesenchymal stem cells efficacy in myocardial infarction therapy. Mater Today Bio 2025; 31:101476. [PMID: 39896290 PMCID: PMC11787032 DOI: 10.1016/j.mtbio.2025.101476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/25/2024] [Accepted: 01/08/2025] [Indexed: 02/04/2025] Open
Abstract
Stem cell-based therapy has emerged as a promising approach for heart repair, potentially regenerating damaged heart tissue and improving outcomes for patients with heart disease. However, the efficacy of stem cell-based therapies remains limited by several challenges, including poor cell survival, low retention rates, poor integration, and limited functional outcomes. This article reviews current enhancement strategies to optimize mesenchymal stem cell therapy for cardiac repair. Key approaches include optimizing cell delivery methods, enhancing cell engraftment, promoting cell functions through genetic and molecular modifications, enhancing the paracrine effects of stem cells, and leveraging biomaterials and tissue engineering techniques. By focusing on these enhancement techniques, the paper highlights innovative approaches that can potentially transform stem cell therapy into a more viable and effective treatment option for cardiac repair. The ongoing research and technological advancements continue to push the boundaries, hoping to make stem cell therapy a mainstream treatment for heart disease.
Collapse
Affiliation(s)
- Chuanfeng An
- Ophthalmology and Transformational Innovation Research Center, Faculty of Medicine of Dalian University of Technology&Dalian Third People's Hospital, Dalian, 116033, PR China
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116033, PR China
| | - Yuan Zhao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Liaoning, Dalian, 116024, PR China
| | - Lipeng Guo
- Ophthalmology and Transformational Innovation Research Center, Faculty of Medicine of Dalian University of Technology&Dalian Third People's Hospital, Dalian, 116033, PR China
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116033, PR China
| | - Zhijian Zhang
- Department of Ophthalmology, Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, PR China
| | - Chunxiao Yan
- Department of Ophthalmology, Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, PR China
| | - Shiying Zhang
- School of Dentistry, Shenzhen University, Shenzhen, 518060, PR China
| | - Yujie Zhang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Liaoning, Dalian, 116024, PR China
| | - Fei Shao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Liaoning, Dalian, 116024, PR China
| | - Yuanyuan Qi
- Ophthalmology and Transformational Innovation Research Center, Faculty of Medicine of Dalian University of Technology&Dalian Third People's Hospital, Dalian, 116033, PR China
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116033, PR China
| | - Xun wang
- Ophthalmology and Transformational Innovation Research Center, Faculty of Medicine of Dalian University of Technology&Dalian Third People's Hospital, Dalian, 116033, PR China
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116033, PR China
| | - Huanan Wang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Liaoning, Dalian, 116024, PR China
| | - Lijun Zhang
- Ophthalmology and Transformational Innovation Research Center, Faculty of Medicine of Dalian University of Technology&Dalian Third People's Hospital, Dalian, 116033, PR China
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116033, PR China
| |
Collapse
|
2
|
Sun Q, Li CH, Liu QS, Zhang YB, Hu BS, Feng Q, Lang Y. Research status of biomaterials based on physical signals for bone injury repair. Regen Ther 2025; 28:544-557. [PMID: 40027992 PMCID: PMC11872413 DOI: 10.1016/j.reth.2025.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/02/2025] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
Bone defects repair continues to be a significant challenge facing the world. Biological scaffolds, bioactive molecules, and cells are the three major elements of bone tissue engineering, which have been widely used in bone regeneration therapy, especially with the rise of bioactive molecules in recent years. According to their physical properties, they can be divided into force, magnetic field (MF), electric field (EF), ultrasonic wave, light, heat, etc. However, the transmission of bioactive molecules has obvious shortcomings that hinder the development of the tissue-rearing process. This paper reviews the mechanism of physical signal induction in bone tissue engineering in recent years. It summarizes the application strategies of physical signal in bone tissue engineering, including biomaterial designs, physical signal loading strategies and related pathways. Finally, the ongoing challenges and prospects for the future are discussed.
Collapse
Affiliation(s)
- Qi Sun
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Chao-Hua Li
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Qi-Shun Liu
- Department of Orthopedics, Zhejiang Medical & Health Group Hangzhou Hospital, Hangzhou, 310015, China
| | - Yuan-Bin Zhang
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Bai-Song Hu
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Qi Feng
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Yong Lang
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| |
Collapse
|
3
|
Zhou S, Wen H, He X, Han X, Li H. Pulsed electromagnetic field ameliorates the progression of osteoarthritis via the Sirt1/NF-κB pathway. Arthritis Res Ther 2025; 27:33. [PMID: 39953605 PMCID: PMC11827477 DOI: 10.1186/s13075-025-03492-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/30/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Pulsed electromagnetic field (PEMF) is a non-invasive treatment that utilizes electromagnetic fields to reduce inflammation and promote tissue repair. However, PEMFs' anti-inflammatory effect on osteoarthritis (OA) and the potential mechanism has not been fully elucidated. METHODS Human chondrocytes (C28/I2) were stimulated with interleukin (IL)-1β with or without the treatment of PEMF. CCK-8 assay Kit was used to detect cell viability. RT-qPCR, ELISA, immunofluorescent staining and western blot was used to analyze relative markers of inflammatory response and extracellular matrix (ECM) under the treatment of PEMF and related mechanism. Besides, the significance role of Sirt1 was assessed by using the Sirt1 inhibitor (EX-527). Moreover, immunohistochemistry and immunofluorescence staining were carried out to evaluate the curative effect of PEMF on OA mice induced by the destabilization of the medial meniscus (DMM). RESULTS PEMF inhibited IL-1β-mediated the expression of pro-inflammatory factors. Besides, PEMF alleviated IL-1β-induced degradation of ECM by increasing the expression of Col2a1 and ACAN, while inhibiting the expression of MMP13 and ADAMTS5. At the mechanism level, PEMF increased the expression of Sirt1 and inhibited IL-1β-induced the activation of NF-κB pathway. Furthermore, blocking Sirt1 with EX-527 attenuated the effect of PEMF on the inhibition of NF-κB pathway and the expression of ECM in IL-1β-induced chondrocytes. In vivo, PEMF-treated OA mice showed low modified mankin scores, reduced the number of osteophytes and preserved joint structure. CONCLUSIONS Our results suggest that PEMF inhibits NF-κB pathway and blocks the expression of inflammatory factors by activating the expression of Sirt1, which may be a novel strategy for OA.
Collapse
Affiliation(s)
- Siqi Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Haiyan Wen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiongwei He
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaotao Han
- Wuhan National High Magnetic Field Center, Huazhong University of Science and Technology, Wuhan, 430074, China
- State Key Laboratory of Advanced Electromagnetic Engineering and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Haohuan Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
4
|
Zeng L, Liu C, Wu Y, Liu S, Zheng Y, Hao W, Wang D, Sun L. Efficacy and safety of mesenchymal stromal cell transplantation in the treatment of autoimmune and rheumatic immune diseases: a systematic review and meta-analysis of randomized controlled trials. Stem Cell Res Ther 2025; 16:65. [PMID: 39934871 DOI: 10.1186/s13287-025-04184-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/23/2025] [Indexed: 02/13/2025] Open
Abstract
OBJECTIVE This study aims to assess the effectiveness and safety of mesenchymal stem cell (MSC) transplantation in the treatment of autoimmune and rheumatic immune diseases through randomized controlled trials (RCTs). METHODS Two researchers conducted a comprehensive search of Chinese and English databases from their inception until Dec. 2023. The literature screening and data extraction were then performed. Statistical analysis was carried out using RevMan 5.4 software. RESULTS A total of 42 relevant RCTs, involving 2,183 participants, were ultimately included in this study. These RCTs encompassed four types of rheumatic immune and bone diseases, namely rheumatoid arthritis (RA), osteoarthritis (OA), spondyloarthritis, systemic sclerosis arthritis, systemic lupus erythematosus (SLE), inflammatory bowel disease, multiple sclerosis, primary Sjögren's syndrome (PSS). The systematic review indicates that MSC transplantation may improve spondyloarthritis, RA, PSS. The meta-analysis reveals that MSC transplantation significantly improved symptoms in patients with OA [VAS (visual analogue scale): bone marrow: SMD = - 0.95, 95% CI - 1.55 to - 0.36, P = 0.002; umbilical cord: SMD = - 1.25, 95% CI - 2.04 to - 0.46, P = 0.002; adipose tissue: SMD = -1.26, 95% CI -1.99 to - 0.52, P = 0.0009)], SLE [Systemic lupus erythematosus disease activity index (SLEDAI): SMD = - 2.32, 95% CI - 3.59 to - 1.06, P = 0.0003], inflammatory bowel disease [clinical efficacy: RR = 2.02, 95% CI 1.53 to 2.67, P < 0.00001]. However, MSC transplantation may not improve the symptoms of multiple sclerosis and systemic sclerosis (Ssc). Importantly, MSC transplantation did not increase the incidence of adverse events (OA: RR = 1.23, 95% CI 0.93 to 1.65, P = 0.15; SLE: RR = 0.83, 95% CI 0.28 to 2.51, P = 0.76; Inflammatory bowel disease: RR = 0.99, 95% CI 0.81 to 1.22, P = 0.96; Multiple sclerosis: RR = 1.12, 95% CI 0.81 to 1.53, P = 0.50), supporting its safety profile across the included studies. These findings suggest that MSC transplantation holds promise for several rheumatic and autoimmune diseases while highlighting areas where further research is warranted. CONCLUSION MSC transplantation may have the potential to treat autoimmune and rheumatic immune diseases. Moreover. MSC transplantation appears to be relatively safe and could be considered as a viable alternative treatment option for autoimmune and rheumatic immune diseases.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Chang Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Yang Wu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Shuman Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Yaru Zheng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Wensa Hao
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
5
|
Abed Elrashid NA, Ali OI, Ibrahim ZM, El Sharkawy MA, Bin sheeha B, Amin WM. A Double-Blinded Randomized Controlled Trial: Can Pulsed Electromagnetic Field Therapy Be a Novel Method for Treating Chronic Rhinosinusitis? MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1868. [PMID: 39597053 PMCID: PMC11596204 DOI: 10.3390/medicina60111868] [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: 10/21/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives: Pulsed electromagnetic field (PEMF) therapy offers a promising approach to treating inflammatory diseases. Its notable anti-inflammatory and antimicrobial effects and enhancement of microcirculation in the nasal mucosa make it a valuable treatment option. Despite its potential, the use of PEMF for chronic rhinosinusitis (CRS) is still in its early stages, with limited exploration of its effectiveness. This study aimed to assess the impact of PEMF on alleviating symptoms such as fatigue, headaches, sinus opacifications, and ostiomeatal complex issues associated with CRS. Materials and Methods: Forty-seven patients of both genders with CRS, aged 19 to 40 years, were involved in this study. The participants were randomly assigned to either a magnetic or a control group. The magnetic group underwent a 10 min PEMF session with a 20-gauss magnetic field strength at 7 Hz thrice a week for a month. The control group received the same PEMF application as an inactive device. Before and after the intervention, researchers assessed fatigue levels with a visual analog fatigue scale (VAFS), headache intensity via a numerical pain-rating scale, and the status of sinus opacifications and ostiomeatal complex obstructions by computerized tomography (CT). Results: The study findings showed a significant reduction in fatigue and headache scores in the magnetic group compared to the control group (p < 0.05). Additionally, there was a notable improvement in sinus opacifications and ostiomeatal complex obstructions among participants who received PEMF therapy. Conclusions: PEMF therapy effectively reduces fatigue, headaches, and sinus opacifications in CRS patients, suggesting its potential for inclusion in CRS management guidelines to improve patient outcomes and quality of life. The results of this study indicate that PEMF represents a noninvasive and cost-effective approach for treating adults with mild-to-moderate CRS.
Collapse
Affiliation(s)
| | - Olfat Ibrahim Ali
- Physical Therapy Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia;
- Department of Basic Science for Physical Therapy, Faculty of Physical Therapy, Cairo University, Giza 12613, Egypt
| | - Zizi M. Ibrahim
- Department of Rehabilitation Sciences, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (Z.M.I.); (B.B.s.)
| | - Mohammed A. El Sharkawy
- Department of Otorhinolaryngology, Faculty of Medicine, Al-Azhar University, Cairo 11884, Egypt;
| | - Bodor Bin sheeha
- Department of Rehabilitation Sciences, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (Z.M.I.); (B.B.s.)
| | - Wafaa Mahmoud Amin
- Department of Physical Therapy, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
- Basic Science Department for Physical Therapy, Faculty of Physical Therapy, Cairo University, Giza 12613, Egypt
| |
Collapse
|
6
|
Caliogna L, Berni M, Torriani C, Mancuso ME, Di Minno MND, Brancato AM, Jannelli E, Mosconi M, Pasta G. Pathogenesis of osteoarthritis, rheumatoid arthritis, and hemophilic arthropathy: The role of angiogenesis. Haemophilia 2024; 30:1256-1264. [PMID: 39297375 PMCID: PMC11659485 DOI: 10.1111/hae.15097] [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: 04/19/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 12/21/2024]
Abstract
INTRODUCTION The term 'chronic inflammatory arthritis' (IA) can be used to define a group of heterogeneous diseases in which inflammation of the synovium is the common feature while having different pathogenesis and clinical outcomes. This condition can be found in osteoarthritis (OA), rheumatoid arthritis (RA), and hemophilic arthropathy (HA). AIM The objective is to try to highlight similarities and differences in the three pathological conditions and understand both molecular and physiological mechanisms. METHOD We have carried out a systematic review of the available literature following the guidelines Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA). RESULTS By comparing the data in the literature on OA, RA, and HA we have shown that the three pathologies differ in initial etiology but they motivate the same molecular pathways. CONCLUSION In this review we highlighted the similarities and differences between these diseases, creating ideas for future studies both in vivo and in vitro to develop new therapeutic agents and suggest possible biomarkers to follow the evolution and severity of the disease.
Collapse
Affiliation(s)
- Laura Caliogna
- Orthopedics and Traumatology ClinicIRCCS Policlinico San Matteo FoundationPaviaItaly
| | - Micaela Berni
- Orthopedics and Traumatology ClinicIRCCS Policlinico San Matteo FoundationPaviaItaly
| | - Camilla Torriani
- Orthopedics and Traumatology ClinicIRCCS Policlinico San Matteo FoundationPaviaItaly
| | - Maria Elisa Mancuso
- Center for Thrombosis and Hemorrhagic DiseasesIRCCS Humanitas Research HospitalRozzanoMilanItaly
| | | | - Alice Maria Brancato
- Orthopedics and Traumatology ClinicIRCCS Policlinico San Matteo FoundationPaviaItaly
| | - Eugenio Jannelli
- Orthopedics and Traumatology ClinicIRCCS Policlinico San Matteo FoundationPaviaItaly
- Department of Clinical, Surgical, Diagnostic and Pediatric SciencesUniversity of PaviaPaviaItaly
| | - Mario Mosconi
- Orthopedics and Traumatology ClinicIRCCS Policlinico San Matteo FoundationPaviaItaly
- Department of Clinical, Surgical, Diagnostic and Pediatric SciencesUniversity of PaviaPaviaItaly
| | - Gianluigi Pasta
- Orthopedics and Traumatology ClinicIRCCS Policlinico San Matteo FoundationPaviaItaly
| |
Collapse
|
7
|
Trentini M, D’Amora U, Ronca A, Lovatti L, Calvo-Guirado JL, Licastro D, Monego SD, Delogu LG, Wieckowski MR, Barak S, Dolkart O, Zavan B. Bone Regeneration Revolution: Pulsed Electromagnetic Field Modulates Macrophage-Derived Exosomes to Attenuate Osteoclastogenesis. Int J Nanomedicine 2024; 19:8695-8707. [PMID: 39205866 PMCID: PMC11352519 DOI: 10.2147/ijn.s470901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction In the process of bone regeneration, a prominent role is played by macrophages involved in both the initial inflammation and the regeneration/vascularization phases, due to their M2 anti-inflammatory phenotype. Together with osteoclasts, they participate in the degradation of the bone matrix if the inflammatory process does not end. In this complex scenario, recently, much attention has been paid to extracellular communication mediated by nanometer-sized vesicles, with high information content, called exosomes (EVs). Considering these considerations, the purpose of the present work is to demonstrate how the presence of a pulsed electromagnetic field (PEMF) can positively affect communication through EVs. Methods To this aim, macrophages and osteoclasts were treated in vitro with PEMF and analyzed through molecular biology analysis and by electron microscopy. Moreover, EVs produced by macrophages were characterized and used to verify their activity onto osteoclasts. Results The results confirmed that PEMF not only reduces the inflammatory activity of macrophages and the degradative activity of osteoclasts but that the EVS produced by macrophages, obtained from PEMF treatment, positively affect osteoclasts by reducing their activity. Discussion The co-treatment of PEMF with M2 macrophage-derived EVs (M2-EVs) decreased osteoclastogenesis to a greater degree than separate treatments.
Collapse
Affiliation(s)
- Martina Trentini
- Translational Medicine Department, University of Ferrara, Ferrara, 44121, Italy
| | - Ugo D’Amora
- Institute of Polymers, Composites and Biomaterials - National Research Council (IPCB-CNR), Naples, 80125, Italy
| | - Alfredo Ronca
- Institute of Polymers, Composites and Biomaterials - National Research Council (IPCB-CNR), Naples, 80125, Italy
| | - Luca Lovatti
- Institute of Polymers, Composites and Biomaterials - National Research Council (IPCB-CNR), Naples, 80125, Italy
| | - José Luis Calvo-Guirado
- Faculty of Health Sciences, Universidad Autonoma de Chile, Santiago de Chile, 7500912, Chile
| | | | | | | | - Mariusz R Wieckowski
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | | | | | - Barbara Zavan
- Translational Medicine Department, University of Ferrara, Ferrara, 44121, Italy
| |
Collapse
|
8
|
Song K, Hu J, Yang M, Xia Y, He C, Yang Y, Zhu S. Pulsed electromagnetic fields potentiate bone marrow mesenchymal stem cell chondrogenesis by regulating the Wnt/β-catenin signaling pathway. J Transl Med 2024; 22:741. [PMID: 39107784 PMCID: PMC11301989 DOI: 10.1186/s12967-024-05470-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Pulsed electromagnetic fields (PEMFs) show promise as a treatment for knee osteoarthritis (KOA) by reducing inflammation and promoting chondrogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). PURPOSE To identify the efficacy window of PEMFs to induce BMSCs chondrogenic differentiation and explore the cellular mechanism under chondrogenesis of BMSCs in regular and inflammatory microenvironments. METHODS BMSCs were exposed to PEMFs (75 Hz, 1.6/2/3/3.8 mT) for 7 and 14 days. The histology, proliferation, migration and chondrogenesis of BMSCs were assessed to identify the optimal parameters. Using these optimal parameters, transcriptome analysis was performed to identify target genes and signaling pathways, validated through immunohistochemical assays, western blotting, and qRT-PCR, with or without the presence of IL-1β. The therapeutic effects of PEMFs and the effective cellular signaling pathways were evaluated in vivo. RESULTS BMSCs treated with 3 mT PEMFs showed the optimal chondrogenesis on day 7, indicated by increased expression of ACAN, COL2A, and SOX9, and decreased levels of MMP3 and MMP13 at both transcriptional and protein levels. The advantages of 3 mT PEMFs diminished in the 14-day culture groups. Transcriptome analysis identified sFRP3 as a key molecule targeted by PEMF treatment, which competitively inhibited Wnt/β-catenin signaling, regardless of IL-1β presence or duration of exposure. This inhibition of the Wnt/β-catenin pathway was also confirmed in a KOA mouse model following PEMF exposure. CONCLUSIONS PEMFs at 75 Hz and 3 mT are optimal in inducing early-stage chondrogenic differentiation of BMSCs. The induction and chondroprotective effects of PEMFs are mediated by sFRP3 and Wnt/β-catenin signaling, irrespective of inflammatory conditions.
Collapse
Affiliation(s)
- Kangping Song
- Rehabilitation Medicine Center, Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, #37 Guoxue Alley, Wuhou strict, Chengdu, Sichuan, 610041, PR China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, China
| | - Jing Hu
- Rehabilitation Medicine Center, Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, #37 Guoxue Alley, Wuhou strict, Chengdu, Sichuan, 610041, PR China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, China
| | - Ming Yang
- The Lab of Aging Research, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, Chengdu, China
| | - Yong Xia
- Rehabilitation Medicine Center, Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, #37 Guoxue Alley, Wuhou strict, Chengdu, Sichuan, 610041, PR China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, China
| | - Chengqi He
- Rehabilitation Medicine Center, Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, #37 Guoxue Alley, Wuhou strict, Chengdu, Sichuan, 610041, PR China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, China
| | - Yonghong Yang
- Rehabilitation Medicine Center, Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, #37 Guoxue Alley, Wuhou strict, Chengdu, Sichuan, 610041, PR China.
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, China.
| | - Siyi Zhu
- Rehabilitation Medicine Center, Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, #37 Guoxue Alley, Wuhou strict, Chengdu, Sichuan, 610041, PR China.
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Qi J, Li H, Du Y, Liu Y, He W, Meng J, Wei L, Zhang K, Lu Y, Zhu X. Circulating Autoantibody Profiling Identifies LIMS1 as a Potential Target for Pathogenic Autoimmunity in pathologic Myopia. Mol Cell Proteomics 2024; 23:100783. [PMID: 38729610 PMCID: PMC11215957 DOI: 10.1016/j.mcpro.2024.100783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
High myopia is a leading cause of blindness worldwide, among which pathologic myopia, characterized by typical myopic macular degeneration, is the most detrimental. However, its pathogenesis remains largely unknown. Here, using a HuProt array, we first initiated a serological autoantibody profiling of high myopia and identified 18 potential autoantibodies, of which anti-LIMS1 autoantibody was validated by a customized focused microarray. Further subgroup analysis revealed its actual relevance to pathologic myopia, rather than simple high myopia without myopic macular degeneration. Mechanistically, anti-LIMS1 autoantibody predominantly belonged to IgG1/IgG2/IgG3 subclasses. Serum IgG obtained from patients with pathologic myopia could disrupt the barrier function of retinal pigment epithelial cells via cytoskeleton disorganization and tight junction component reduction, and also trigger a pro-inflammatory mediator cascade in retinal pigment epithelial cells, which were all attenuated by depletion of anti-LIMS1 autoantibody. Together, these data uncover a previously unrecognized autoimmune etiology of myopic macular degeneration in pathologic myopia.
Collapse
Affiliation(s)
- Jiao Qi
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, People's Republic of China; NHC Key Laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, People's Republic of China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People's Republic of China
| | - Hao Li
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yu Du
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, People's Republic of China; NHC Key Laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, People's Republic of China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People's Republic of China
| | - Yun Liu
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Wenwen He
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, People's Republic of China; NHC Key Laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, People's Republic of China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People's Republic of China
| | - Jiaqi Meng
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, People's Republic of China; NHC Key Laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, People's Republic of China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People's Republic of China
| | - Ling Wei
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, People's Republic of China; NHC Key Laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, People's Republic of China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People's Republic of China
| | - Keke Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, People's Republic of China; NHC Key Laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, People's Republic of China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People's Republic of China
| | - Yi Lu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, People's Republic of China; NHC Key Laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, People's Republic of China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People's Republic of China.
| | - Xiangjia Zhu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, People's Republic of China; NHC Key Laboratory of Myopia and Related Eye Diseases, Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China; Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, People's Republic of China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
10
|
Gerdesmeyer L, Burgkart R, Saxena A. Clavicle fracture and triathlon performance: a case report. J Med Case Rep 2024; 18:197. [PMID: 38566165 PMCID: PMC10988895 DOI: 10.1186/s13256-024-04482-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Collarbone fracture is a common injury, particularly among athletes involved in contact sports and participating in endurance activities. Conventional treatment requires surgery and postoperative immobilization, resulting in an average return-to-sport timeframe of approximately 13 weeks. This case challenges the established treatment protocols, aiming to expedite recovery and enable a quicker resumption of high-intensity athletic activities. CASE PRESENTATION A 24-year-old Caucasian athlete completed a Half-Ironman Triathlon (70.3) merely three weeks post-collarbone fracture. Utilizing Extracorporeal Magneto-Transduction Therapy (EMTT) alongside surgical intervention, the patient achieved accelerated healing and remarkable performance outcomes without encountering any adverse effects. CONCLUSIONS The integration of EMTT into the treatment paradigm for bone fractures alters the traditional understanding of recovery timelines and rehabilitation strategies. This case highlights the potential benefits of electromagnetic wave therapy in expediting the healing process and enabling athletes to resume high-level sports activities at an earlier stage.
Collapse
Affiliation(s)
- Lennart Gerdesmeyer
- Department of Orthopaedics and Sports Orthopaedics, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Rainer Burgkart
- Department of Orthopaedics and Sports Orthopaedics, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Amol Saxena
- PAMF-Sutter Department of Sports Medicine, 795 El Camino Real, Clark Building, Level 3, Palo Alto, CA, 94301, USA
| |
Collapse
|
11
|
Xie W, Song C, Guo R, Zhang X. Static magnetic fields in regenerative medicine. APL Bioeng 2024; 8:011503. [PMID: 38486824 PMCID: PMC10939708 DOI: 10.1063/5.0191803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
All organisms on Earth live in the weak but ubiquitous geomagnetic field. Human beings are also exposed to magnetic fields generated by multiple sources, ranging from permanent magnets to magnetic resonance imaging (MRI) in hospitals. It has been shown that different magnetic fields can generate various effects on different tissues and cells. Among them, stem cells appear to be one of the most sensitive cell types to magnetic fields, which are the fundamental units of regenerative therapies. In this review, we focus on the bioeffects of static magnetic fields (SMFs), which are related to regenerative medicine. Most reports in the literature focus on the influence of SMF on bone regeneration, wound healing, and stem cell production. Multiple aspects of the cellular events, including gene expression, cell signaling pathways, reactive oxygen species, inflammation, and cytoskeleton, have been shown to be affected by SMFs. Although no consensus yet, current evidence indicates that moderate and high SMFs could serve as a promising physical tool to promote bone regeneration, wound healing, neural differentiation, and dental regeneration. All in vivo studies of SMFs on bone regeneration and wound healing have shown beneficial effects, which unravel the great potential of SMFs in these aspects. More mechanistic studies, magnetic field parameter optimization, and clinical investigations on human bodies will be imperative for the successful clinical applications of SMFs in regenerative medicine.
Collapse
Affiliation(s)
| | - Chao Song
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, HFIPS, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Ruowen Guo
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, HFIPS, Chinese Academy of Sciences, Hefei, Anhui 230031, China
| | - Xin Zhang
- Author to whom correspondence should be addressed:. Tel.: 86–551-65593356
| |
Collapse
|
12
|
Zhou J, Jia F, Qu M, Ning P, Huang X, Tan L, Liu D, Zhong P, Wu Q. The prevention effect of pulsed electromagnetic fields treatment on senile osteoporosis in vivo via improving the inflammatory bone microenvironment. Electromagn Biol Med 2024:1-15. [PMID: 38329038 DOI: 10.1080/15368378.2024.2314093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/26/2023] [Indexed: 02/09/2024]
Abstract
This study aimed to assess PEMF in a rat model of senile osteoporosis and its relationship with NLRP3-mediated low-grade inflammation in the bone marrow microenvironment. A total of 24 Sprague Dawley (SD) rats were included in this study. Sixteen of them were 24-month natural-aged male SD rats, which were randomly distributed into the Aged group and the PEMF group (n = 8 per group). The remaining 8 3-month -old rats were used as the Young positive control group (n = 8). Rats in the PEMF group received 12 weeks of PEMF with 40 min/day, five days per week, while the other rats received placebo PEMF intervention. Bone mineral density/microarchitecture, serum levels of CTX-1 and P1CP, and NLRP3-related signaling genes and proteins in rat bone marrow were then analyzed. The 12-week of PEMF showed significant mitigation of aging-induced bone loss and bone microarchitecture deterioration, i.e. PEMF increased the bone mineral density of the proximal femur and L5 vertebral body and improved parameters of the proximal tibia and L4 vertebral body. Further analysis showed that PEMF reversed aging-induced bone turnover, specifically, decreased serum CTX-1 and elevated serum P1CP. Furthermore, PEMF also dramatically inhibited NLRP3-mediated low-grade inflammation in the bone marrow, i.e. PEMF inhibited the levels of NLRP3, proCaspase1, cleaved Caspase1, IL-1β, and GSDMD-N. The study demonstrated that PEMF could mitigate the aging-induced bone loss and reverses the deterioration of bone microarchitecture probably through inhibiting NLRP3-mediated low-grade chronic inflammation to improve the inflammatory bone microenvironment in aged rats.
Collapse
Affiliation(s)
- Jun Zhou
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Acupuncture/Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Feiyang Jia
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Acupuncture/Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Mengjian Qu
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Acupuncture/Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Pengyun Ning
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Acupuncture/Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiarong Huang
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Acupuncture/Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lu Tan
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Acupuncture/Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Danni Liu
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Acupuncture/Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Peirui Zhong
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Acupuncture/Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qi Wu
- The First Affiliated Hospital, Rehabilitation Medicine Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Acupuncture/Rehabilitation Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- School of Rehabilitation Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
13
|
Ejma-Multański A, Wajda A, Paradowska-Gorycka A. Cell Cultures as a Versatile Tool in the Research and Treatment of Autoimmune Connective Tissue Diseases. Cells 2023; 12:2489. [PMID: 37887333 PMCID: PMC10605903 DOI: 10.3390/cells12202489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023] Open
Abstract
Cell cultures are an important part of the research and treatment of autoimmune connective tissue diseases. By culturing the various cell types involved in ACTDs, researchers are able to broaden the knowledge about these diseases that, in the near future, may lead to finding cures. Fibroblast cultures and chondrocyte cultures allow scientists to study the behavior, physiology and intracellular interactions of these cells. This helps in understanding the underlying mechanisms of ACTDs, including inflammation, immune dysregulation and tissue damage. Through the analysis of gene expression patterns, surface proteins and cytokine profiles in peripheral blood mononuclear cell cultures and endothelial cell cultures researchers can identify potential biomarkers that can help in diagnosing, monitoring disease activity and predicting patient's response to treatment. Moreover, cell culturing of mesenchymal stem cells and skin modelling in ACTD research and treatment help to evaluate the effects of potential drugs or therapeutics on specific cell types relevant to the disease. Culturing cells in 3D allows us to assess safety, efficacy and the mechanisms of action, thereby aiding in the screening of potential drug candidates and the development of novel therapies. Nowadays, personalized medicine is increasingly mentioned as a future way of dealing with complex diseases such as ACTD. By culturing cells from individual patients and studying patient-specific cells, researchers can gain insights into the unique characteristics of the patient's disease, identify personalized treatment targets, and develop tailored therapeutic strategies for better outcomes. Cell culturing can help in the evaluation of the effects of these therapies on patient-specific cell populations, as well as in predicting overall treatment response. By analyzing changes in response or behavior of patient-derived cells to a treatment, researchers can assess the response effectiveness to specific therapies, thus enabling more informed treatment decisions. This literature review was created as a form of guidance for researchers and clinicians, and it was written with the use of the NCBI database.
Collapse
Affiliation(s)
- Adam Ejma-Multański
- Department of Molecular Biology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (A.W.); (A.P.-G.)
| | | | | |
Collapse
|
14
|
Zhang W, Zha K, Hu W, Xiong Y, Knoedler S, Obed D, Panayi AC, Lin Z, Cao F, Mi B, Liu G. Multifunctional hydrogels: advanced therapeutic tools for osteochondral regeneration. Biomater Res 2023; 27:76. [PMID: 37542353 PMCID: PMC10403923 DOI: 10.1186/s40824-023-00411-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/05/2023] [Indexed: 08/06/2023] Open
Abstract
Various joint pathologies such as osteochondritis dissecans, osteonecrosis, rheumatic disease, and trauma, may result in severe damage of articular cartilage and other joint structures, ranging from focal defects to osteoarthritis (OA). The osteochondral unit is one of the critical actors in this pathophysiological process. New approaches and applications in tissue engineering and regenerative medicine continue to drive the development of OA treatment. Hydrogel scaffolds, a component of tissue engineering, play an indispensable role in osteochondral regeneration. In this review, tissue engineering strategies regarding osteochondral regeneration were highlighted and summarized. The application of hydrogels for osteochondral regeneration within the last five years was evaluated with an emphasis on functionalized physical and chemical properties of hydrogel scaffolds, functionalized delivery hydrogel scaffolds as well as functionalized intelligent response hydrogel scaffolds. Lastly, to serve as guidance for future efforts in the creation of bioinspired hydrogel scaffolds, a succinct summary and new views for specific mechanisms, applications, and existing limitations of the newly designed functionalized hydrogel scaffolds were offered.
Collapse
Affiliation(s)
- Wenqian Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Kangkang Zha
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Weixian Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yuan Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Samuel Knoedler
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
| | - Doha Obed
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
- Department of Plastic, Aesthetic, Hand and Reconstructive Surgery, Hannover Medical School, Hannover, Germany
| | - Adriana C Panayi
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, 67071, Ludwigshafen/Rhine, Germany
| | - Ze Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Faqi Cao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore.
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
15
|
Flatscher J, Pavez Loriè E, Mittermayr R, Meznik P, Slezak P, Redl H, Slezak C. Pulsed Electromagnetic Fields (PEMF)-Physiological Response and Its Potential in Trauma Treatment. Int J Mol Sci 2023; 24:11239. [PMID: 37510998 PMCID: PMC10379303 DOI: 10.3390/ijms241411239] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Environmental biophysical interactions are recognized to play an essential part in the human biological processes associated with trauma recovery. Many studies over several decades have furthered our understanding of the effects that Pulsed Electromagnetic Fields (PEMF) have on the human body, as well as on cellular and biophysical systems. These investigations have been driven by the observed positive clinical effects of this non-invasive treatment on patients, mainly in orthopedics. Unfortunately, the diversity of the various study setups, with regard to physical parameters, molecular and cellular response, and clinical outcomes, has made it difficult to interpret and evaluate commonalities, which could, in turn, lead to finding an underlying mechanistic understanding of this treatment modality. In this review, we give a birds-eye view of the vast landscape of studies that have been published on PEMF, presenting the reader with a scaffolded summary of relevant literature starting from categorical literature reviews down to individual studies for future research studies and clinical use. We also highlight discrepancies within the many diverse study setups to find common reporting parameters that can lead to a better universal understanding of PEMF effects.
Collapse
Affiliation(s)
- Jonas Flatscher
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
| | - Elizabeth Pavez Loriè
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
| | | | - Paul Meznik
- AUVA Trauma Center Vienna-Meidling, 1120 Vienna, Austria
| | - Paul Slezak
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
| | - Cyrill Slezak
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Department of Physics, Utah Valley University, Orem, UT 84058, USA
| |
Collapse
|
16
|
Zhai M, Zhang C, Cui J, Liu J, Li Y, Xie K, Luo E, Tang C. Electromagnetic fields ameliorate hepatic lipid accumulation and oxidative stress: potential role of CaMKKβ/AMPK/SREBP-1c and Nrf2 pathways. Biomed Eng Online 2023; 22:51. [PMID: 37217972 DOI: 10.1186/s12938-023-01114-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/13/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide, and is related to disturbed lipid metabolism and redox homeostasis. However, a definitive drug treatment has not been approved for this disease. Studies have found that electromagnetic fields (EMF) can ameliorate hepatic steatosis and oxidative stress. Nevertheless, the mechanism remains unclear. METHODS NAFLD models were established by feeding mice a high-fat diet. Simultaneously, EMF exposure is performed. The effects of the EMF on hepatic lipid deposition and oxidative stress were investigated. Additionally, the AMPK and Nrf2 pathways were analysed to confirm whether they were activated by the EMF. RESULTS Exposure to EMF decreased the body weight, liver weight and serum triglyceride (TG) levels and restrained the excessive hepatic lipid accumulation caused by feeding the HFD. The EMF boosted CaMKKβ protein expression, activated AMPK phosphorylation and suppressed mature SREBP-1c protein expression. Meanwhile, the activity of GSH-Px was enhanced following an increase in nuclear Nrf2 protein expression by PEMF. However, no change was observed in the activities of SOD and CAT. Consequently, EMF reduced hepatic reactive oxygen species (ROS) and MDA levels, which means that EMF relieved liver damage caused by oxidative stress in HFD-fed mice. CONCLUSIONS EMF may activate the CaMKKβ/AMPK/SREBP-1c and Nrf2 pathways to control hepatic lipid deposition and oxidative stress. This investigation indicates that EMF may be a novel therapeutic method for NAFLD.
Collapse
Affiliation(s)
- Mingming Zhai
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
- Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Xi'an, China
| | - Chenxu Zhang
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Jinxiu Cui
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Juan Liu
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Yuanzhe Li
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Kangning Xie
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Erping Luo
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China.
| | - Chi Tang
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China.
- Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Xi'an, China.
| |
Collapse
|
17
|
Liu FY, Shi M, Li X, Yuan HJ, Tian XM, Xia YM, Zhou M, Wang FS. MRI/PAI Dual-modal Imaging-guided Precise Tracking of Bone Marrow-derived Mesenchymal Stem Cells Labeled with Nanoparticles for Treating Liver Cirrhosis. J Clin Transl Hepatol 2023; 11:382-392. [PMID: 36643042 PMCID: PMC9817052 DOI: 10.14218/jcth.2021.00580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/24/2022] [Accepted: 05/05/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND AND AIMS Stem cell transplantation is a potential treatment option for liver cirrhosis (LC). Accurately and noninvasively monitoring the distribution, migration, and prognosis of transplanted stem cells using imaging methods is important for in-depth study of the treatment mechanisms. Our study aimed to develop Au-Fe3O4 silica nanoparticles (NPs) as tracking nanoplatforms for dual-modal stem cell imaging. METHODS Au-Fe3O4 silica NPs were synthesized by seed-mediated growth method and co-precipitation. The efficiency and cytotoxicity of the NPs-labeled bone marrow-derived mesenchymal stem cells (BM-MSCs) were evaluated by Cell Counting Kit-8 assays, ICP-MS, phenotypic characterization, and histological staining. The biodistribution of labeled BM-MSCs injected through different routes (the hepatic artery or tail vein) into rats with LC was detected by magnetic resonance imaging (MRI), photoacoustic imaging (PAI), and Prussian blue staining. RESULTS Synthesized Au-Fe3O4 silica NPs consisted of a core (star-shaped Au NPs) and an outside silica layer doped with Fe3O4 NPs. After 24 h coincubation with 2.0 OD concentration of NPs, the viability of BM-MSCs was 77.91%±5.86% and the uptake of Au and Fe were (22.65±1.82) µg/mL and (234.03±11.47) µg/mL, respectively. The surface markers of labeled BM-MSCs unchanged significantly. Labeled BM-MSCs have osteogenic and adipogenic differentiation potential. Post injection in vivo, rat livers were hypointense on MRI and hyperintense on PAI. Prussian blue staining showed that more labeled BM-MSCs accumulated in the liver of the hepatic artery group. The severity of LC of the rats in the hepatic artery group was significantly alleviated. CONCLUSIONS Au-Fe3O4 silica NPs were suitable MRI/PAI dual-modal imaging nanoplatforms for stem cell tracking in regenerative medicine. Transhepatic arterial infusion of BM-MSCs was the optimal route for the treatment of LC.
Collapse
Affiliation(s)
- Feng-Yong Liu
- Chinese PLA Medical School, Beijing, China
- Senior Department of Infectious Diseases, Fifth Medical Center of PLA General Hospital, Beijing, China
- Department of Interventional Radiology, Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Ming Shi
- Senior Department of Infectious Diseases, Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Xin Li
- Department of Interventional Radiology, Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Hong-Jun Yuan
- Department of Interventional Radiology, Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Xiao-Mei Tian
- Chinese PLA Medical School, Beijing, China
- Department of Interventional Radiology, Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yi-Mao Xia
- Chinese PLA Medical School, Beijing, China
- Department of Interventional Radiology, Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Min Zhou
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fu-Sheng Wang
- Chinese PLA Medical School, Beijing, China
- Senior Department of Infectious Diseases, Fifth Medical Center of PLA General Hospital, Beijing, China
- Correspondence to: Fu-Sheng Wang, Senior Department of Infectious Diseases, Fifth Medical Center of PLA General Hospital, Beijing 100039, China. ORCID: https://orcid.org/0000-0002-8043-6685. Tel: +86-15801502008, Fax: +86-10-66933332, E-mail:
| |
Collapse
|
18
|
Hack SJ, Beane WS, Tseng KAS. Biophysics at the edge of life and death: radical control of apoptotic mechanisms. FRONTIERS IN CELL DEATH 2023; 2:1147605. [PMID: 39897412 PMCID: PMC11784940 DOI: 10.3389/fceld.2023.1147605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Recent studies have furthered our understanding of how dying and living cells interact in different physiological contexts, however the signaling that initiates and mediates apoptosis and apoptosis-induced proliferation are more complex than previously thought. One increasingly important area of study is the biophysical control of apoptosis. In addition to biochemical regulation, biophysical signals (including redox chemistry, bioelectric gradients, acoustic and magnetic stimuli) are also known yet understudied regulators of both cell death and apoptosis-induced proliferation. Mounting evidence suggests biophysical signals may be key targets for therapeutic interventions. This review highlights what is known about the role of biophysical signals in controlling cell death mechanisms during development, regeneration, and carcinogenesis. Since biophysical signals can be controlled spatiotemporally, bypassing the need for genetic manipulation, further investigation may lead to fine-tuned modulation of apoptotic pathways to direct desired therapeutic outcomes.
Collapse
Affiliation(s)
- Samantha J. Hack
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI, USA
| | - Wendy S. Beane
- Western Michigan University, Department of Biological Sciences, Kalamazoo, MI, USA
| | - Kelly Ai-Sun Tseng
- University of Nevada, Las Vegas, School of Life Sciences, Las Vegas, NV, USA
| |
Collapse
|
19
|
Zhao H, Liu C, Liu Y, Ding Q, Wang T, Li H, Wu H, Ma T. Harnessing electromagnetic fields to assist bone tissue engineering. Stem Cell Res Ther 2023; 14:7. [PMID: 36631880 PMCID: PMC9835389 DOI: 10.1186/s13287-022-03217-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/08/2022] [Indexed: 01/13/2023] Open
Abstract
Bone tissue engineering (BTE) emerged as one of the exceptional means for bone defects owing to it providing mechanical supports to guide bone tissue regeneration. Great advances have been made to facilitate the success of BTE in regenerating bone within defects. The use of externally applied fields has been regarded as an alternative strategy for BTE. Electromagnetic fields (EMFs), known as a simple and non-invasive therapy, can remotely provide electric and magnetic stimulation to cells and biomaterials, thus applying EMFs to assist BTE would be a promising strategy for bone regeneration. When combined with BTE, EMFs improve cell adhesion to the material surface by promoting protein adsorption. Additionally, EMFs have positive effects on mesenchymal stem cells and show capabilities of pro-angiogenesis and macrophage polarization manipulation. These advantages of EMFs indicate that it is perfectly suitable for representing the adjuvant treatment of BTE. We also summarize studies concerning combinations of EMFs and diverse biomaterial types. The strategy of combining EMFs and BTE receives encouraging outcomes and holds a promising future for effectively treating bone defects.
Collapse
Affiliation(s)
- Hongqi Zhao
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Chaoxu Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Yang Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Qing Ding
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Tianqi Wang
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Hao Li
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Tian Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
20
|
Hong JE, Lee CG, Hwang S, Kim J, Jo M, Kang DH, Yoo SH, Kim WS, Lee Y, Rhee KJ. Pulsed Electromagnetic Field (PEMF) Treatment Ameliorates Murine Model of Collagen-Induced Arthritis. Int J Mol Sci 2023; 24:ijms24021137. [PMID: 36674651 PMCID: PMC9862561 DOI: 10.3390/ijms24021137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/24/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease of the joint synovial membranes. RA is difficult to prevent or treat; however, blocking proinflammatory cytokines is a general therapeutic strategy. Pulsed electromagnetic field (PEMF) is reported to alleviate RA's inflammatory response and is being studied as a non-invasive physical therapy. In this current study, PEMF decreased paw inflammation in a collagen-induced arthritis (CIA) murine model. PEMF treatment at 10 Hz was more effective in ameliorating arthritis than at 75 Hz. In the PEMF-treated CIA group, the gross inflammation score and cartilage destruction were lower than in the untreated CIA group. The CIA group treated with PEMF also showed lower serum levels of IL-1β but not IL-6, IL-17, or TNF-α. Serum levels of total anti-type II collagen IgG and IgG subclasses (IgG1, IgG2a, and IgG2b) remained unchanged. In contrast, tissue protein levels of IL-1β, IL-6, TNF-α, receptor activator of nuclear factor kappa-Β (RANK), RANK ligand (RANKL), IL-6 receptor (IL-6R), and TNF-α receptor1 (TNFR1) were all lower in the ankle joints of the PEMF-treated CIA group compared with the CIA group. The results of this study suggest that PEMF treatment can preserve joint morphology cartilage and delay the occurrence of CIA. PEMF has potential as an effective adjuvant therapy that can suppress the progression of RA.
Collapse
Affiliation(s)
- Ju-Eun Hong
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Republic of Korea
| | - Chang-Gun Lee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Republic of Korea
- Department of Medical Genetics, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Soonjae Hwang
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Republic of Korea
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, GAIHST, Gachon University College of Medicine, Incheon 21999, Republic of Korea
| | - Junyoung Kim
- Department of Biomedical Engineering, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Republic of Korea
| | - Minjeong Jo
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Republic of Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Da-Hye Kang
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Republic of Korea
| | - Sang-Hyeon Yoo
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Republic of Korea
| | - Woo-Seung Kim
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Republic of Korea
| | - Yongheum Lee
- Department of Biomedical Engineering, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Republic of Korea
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Republic of Korea
- Correspondence: ; Tel.: +82-33-760-2445; Fax: +82-33-760-2195
| |
Collapse
|
21
|
Zhang T, Zhao Z, Wang T. Pulsed electromagnetic fields as a promising therapy for glucocorticoid-induced osteoporosis. Front Bioeng Biotechnol 2023; 11:1103515. [PMID: 36937753 PMCID: PMC10020513 DOI: 10.3389/fbioe.2023.1103515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is considered the third type of osteoporosis and is accompanied by high morbidity and mortality. Long-term usage of glucocorticoids (GCs) causes worsened bone quality and low bone mass via their effects on bone cells. Currently, there are various clinical pharmacological treatments to regulate bone mass and skeletal health. Pulsed electromagnetic fields (PEMFs) are applied to treat patients suffering from delayed fracture healing and non-unions. PEMFs may be considered a potential and side-effect-free therapy for GIOP. PEMFs inhibit osteoclastogenesis, stimulate osteoblastogenesis, and affect the activity of bone marrow mesenchymal stem cells (BMSCs), osteocytes and blood vessels, ultimately leading to the retention of bone mass and strength. However, the underlying signaling pathways via which PEMFs influence GIOP remain unclear. This review attempts to summarize the underlying cellular mechanisms of GIOP. Furthermore, recent advances showing that PEMFs affect bone cells are discussed. Finally, we discuss the possibility of using PEMFs as therapy for GIOP.
Collapse
Affiliation(s)
- Tianxiao Zhang
- Innovation Center for Wound Repair, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiliang Zhao
- Innovation Center for Wound Repair, West China Hospital, Sichuan University, Chengdu, China
| | - Tiantian Wang
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Tiantian Wang,
| |
Collapse
|
22
|
Pulsed Electromagnetic Fields Disrupt Staphylococcus epidermidis Biofilms and Enhance the Antibiofilm Efficacy of Antibiotics. Microbiol Spectr 2022; 10:e0194922. [PMID: 36314923 PMCID: PMC9769884 DOI: 10.1128/spectrum.01949-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Staphylococcus epidermidis is implicated in a multitude of human infections and is one of the major causes of clinical infections in hospitals, especially at surgical sites and on indwelling medical devices, such as orthopedic implants. These infections are especially dangerous because of the S. epidermidis propensity to form biofilms, which increases resistance to antibiotics and the natural immune response. This study investigated pulsed electromagnetic fields (PEMF) as a potential treatment to combat such infections, as PEMF exposure was expected to disrupt the electrostatic forces that adhere staphylococcal cells to surfaces and to one another. To test the effect of PEMF on biofilms, S. epidermidis cultures were exposed to PEMF at various durations either during the growth phase or after a full biofilm had formed. In addition, cells were exposed to PEMF and concomitant antibiotic treatment. Biofilm viability was quantified by both crystal violet and alamarBlue assays and scanning electron microscopy. The results demonstrated that PEMF significantly inhibited biofilm formation and disrupted preformed biofilms in vitro while also showing synergistic biofilm inhibition when combined with antibiotics. These combined results indicate that PEMF should be considered a promising novel technique for treating S. epidermidis biofilm infections and undergo further testing in vivo. IMPORTANCE Antibiotic resistance and biofilm infections are major issues in health care because of the lack of a successful treatment modality and poor patient outcomes. These infections are a particular issue following orthopedic surgery or trauma wherein an infection may form on an orthopedic implant or patient's bone. The presented study demonstrates that pulsed electromagnetic fields may be a promising novel treatment for such infections and can overcome the medical challenges presented by biofilm formation. Furthermore, the effects demonstrated are even greater when combining pulsed electromagnetic field therapy with traditional antibiotics.
Collapse
|
23
|
Xu Y, Wang Q, Wang XX, Xiang XN, Peng JL, He CQ, He HC. The Effect of Different Frequencies of Pulsed Electromagnetic Fields on Cartilage Repair of Adipose Mesenchymal Stem Cell-Derived Exosomes in Osteoarthritis. Cartilage 2022; 13:200-212. [PMID: 36377077 PMCID: PMC9924977 DOI: 10.1177/19476035221137726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The intra-articular injection of mesenchymal stem cell (MSC)-derived exosomes has already been proved to reverse osteoarthritic cartilage degeneration. Pulsed electromagnetic field (PEMF) has been found to regulate the biogenic function of MSCs. However, the effect of PEMF on MSC-derived exosomes has not yet been characterized. The aim of this study was to elucidate the regulatory role of different frequencies of PEMF in promoting the osteoarthritic cartilage regeneration of MSC-derived exosomes. METHODS The adipose tissue-derived MSCs (AMSCs) were extracted from the epididymal fat of healthy rats and further exposed to the PEMF at 1 mT amplitude and a frequency of 15, 45, and 75 Hz, respectively, in an incubator. The chondrocytes were treated with interlukin-1β (IL-1β) and the regenerative effect of co-culturing with PEMF-exposed AMSC-derived exosomes was assessed via Western blot, quantitative polymerase chain reaction, and ELISA assays. A rat model of osteoarthritis was established by anterior cruciate ligament transection (ACLT) surgery and received 4 times intra-articular injection of PEMF-exposed AMSC-derived exosomes once a week. After 8 weeks, the knee joint specimens of rats were collected for micro-computed tomography and histologic analyses. RESULTS PEMF-exposed AMSC-derived exosomes could be endocytosed with IL-1β-induced chondrocytes. Compared with the AMSC-derived exosomes alone, the PEMF-exposed AMSC-derived exosomes substantially suppressed the inflammation and extracellular matrix degeneration of IL-1β-induced chondrocytes as shown by higher expression of transcripts and proteins of COL2A1, SOX9, and ACAN and lower expression of MMP13 and caspase-1. Of these, the 75-Hz PEMF presented a more significant inhibitive effect than the 15-Hz and 45-Hz PEMFs. Furthermore, the intra-articular injection of 75-Hz PEMF-exposed exosomes could obviously increase the number of tibial epiphyseal trabeculae, lead to a remarkable decrease in Osteoarthritis Research Society International score, and upregulate the COL2A1 and ACAN protein level of the degenerated cartilage. CONCLUSION The present study demonstrated that PEMF stimulation could effectively promote the regeneration effects of AMSC-derived exosomes on osteoarthritic cartilage. Compared with other frequency parameters, the PEMF at a frequency of 75 Hz showed a superior positive effect on AMSC-derived exosomes in suppressing the IL-1β-induced chondrocyte inflammation and extracellular matrix catabolism, as well as the osteoarthritic cartilage degeneration.
Collapse
Affiliation(s)
- Yang Xu
- Rehabilitation Medicine Centre, West
China Hospital, Sichuan University, Chengdu, P.R. China,School of Rehabilitation Sciences, West
China School of Medicine, Sichuan University, Chengdu, P.R. China,Rehabilitation Medicine Key Laboratory
of Sichuan Province, Chengdu, P.R. China
| | - Qian Wang
- Rehabilitation Medicine Centre, West
China Hospital, Sichuan University, Chengdu, P.R. China,School of Rehabilitation Sciences, West
China School of Medicine, Sichuan University, Chengdu, P.R. China,Rehabilitation Medicine Key Laboratory
of Sichuan Province, Chengdu, P.R. China
| | - Xiang-Xiu Wang
- Rehabilitation Medicine Centre, West
China Hospital, Sichuan University, Chengdu, P.R. China,School of Rehabilitation Sciences, West
China School of Medicine, Sichuan University, Chengdu, P.R. China,Rehabilitation Medicine Key Laboratory
of Sichuan Province, Chengdu, P.R. China
| | - Xiao-Na Xiang
- Rehabilitation Medicine Centre, West
China Hospital, Sichuan University, Chengdu, P.R. China,School of Rehabilitation Sciences, West
China School of Medicine, Sichuan University, Chengdu, P.R. China,Rehabilitation Medicine Key Laboratory
of Sichuan Province, Chengdu, P.R. China
| | - Jia-Lei Peng
- Rehabilitation Medicine Centre, West
China Hospital, Sichuan University, Chengdu, P.R. China,School of Rehabilitation Sciences, West
China School of Medicine, Sichuan University, Chengdu, P.R. China,Rehabilitation Medicine Key Laboratory
of Sichuan Province, Chengdu, P.R. China
| | - Cheng-Qi He
- Rehabilitation Medicine Centre, West
China Hospital, Sichuan University, Chengdu, P.R. China,School of Rehabilitation Sciences, West
China School of Medicine, Sichuan University, Chengdu, P.R. China,Rehabilitation Medicine Key Laboratory
of Sichuan Province, Chengdu, P.R. China
| | - Hong-Chen He
- Rehabilitation Medicine Centre, West
China Hospital, Sichuan University, Chengdu, P.R. China,School of Rehabilitation Sciences, West
China School of Medicine, Sichuan University, Chengdu, P.R. China,Rehabilitation Medicine Key Laboratory
of Sichuan Province, Chengdu, P.R. China,Hong-Chen He, Rehabilitation Medicine
Centre, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R.
China.
| |
Collapse
|
24
|
Sakhrani N, Stefani RM, Setti S, Cadossi R, Ateshian GA, Hung CT. Pulsed Electromagnetic Field Therapy and Direct Current Electric Field Modulation Promote the Migration of Fibroblast-like Synoviocytes to Accelerate Cartilage Repair In Vitro. APPLIED SCIENCES (BASEL, SWITZERLAND) 2022; 12:12406. [PMID: 36970107 PMCID: PMC10035757 DOI: 10.3390/app122312406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Articular cartilage injuries are a common source of joint pain and dysfunction. As articular cartilage is avascular, it exhibits a poor intrinsic healing capacity for self-repair. Clinically, osteochondral grafts are used to surgically restore the articular surface following injury. A significant challenge remains with the repair properties at the graft-host tissue interface as proper integration is critical toward restoring normal load distribution across the joint. A key to addressing poor tissue integration may involve optimizing mobilization of fibroblast-like synoviocytes (FLS) that exhibit chondrogenic potential and are derived from the adjacent synovium, the specialized connective tissue membrane that envelops the diarthrodial joint. Synovium-derived cells have been directly implicated in the native repair response of articular cartilage. Electrotherapeutics hold potential as low-cost, low-risk, non-invasive adjunctive therapies for promoting cartilage healing via cell-mediated repair. Pulsed electromagnetic fields (PEMFs) and applied direct current (DC) electric fields (EFs) via galvanotaxis are two potential therapeutic strategies to promote cartilage repair by stimulating the migration of FLS within a wound or defect site. PEMF chambers were calibrated to recapitulate clinical standards (1.5 ± 0.2 mT, 75 Hz, 1.3 ms duration). PEMF stimulation promoted bovine FLS migration using a 2D in vitro scratch assay to assess the rate of wound closure following cruciform injury. Galvanotaxis DC EF stimulation assisted FLS migration within a collagen hydrogel matrix in order to promote cartilage repair. A novel tissue-scale bioreactor capable of applying DC EFs in sterile culture conditions to 3D constructs was designed in order to track the increased recruitment of synovial repair cells via galvanotaxis from intact bovine synovium explants to the site of a cartilage wound injury. PEMF stimulation further modulated FLS migration into the bovine cartilage defect region. Biochemical composition, histological analysis, and gene expression revealed elevated GAG and collagen levels following PEMF treatment, indicative of its pro-anabolic effect. Together, PEMF and galvanotaxis DC EF modulation are electrotherapeutic strategies with complementary repair properties. Both procedures may enable direct migration or selective homing of target cells to defect sites, thus augmenting natural repair processes for improving cartilage repair and healing.
Collapse
Affiliation(s)
- Neeraj Sakhrani
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Robert M. Stefani
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | | | | | - Gerard A. Ateshian
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Mechanical Engineering, Columbia University, New York, NY 10027, USA
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| |
Collapse
|
25
|
Abstract
Abstract
The pathogenesis of connective tissue diseases (CTDs), represented by systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), systemic sclerosis (SSc), primary Sjögren’s syndrome (pSS), and idiopathic inflammatory myopathies (IIM), includes various immune cells involved in both innate and adaptive immunity. The mesenchymal stem cells (MSCs) are unique due to their regulatory effect on immunity. This makes them a promising therapeutic approach for patients with immune-mediated disorders such as CTD. The safety and clinical efficacy of MSC treatment in CTD have been tested in a growing number of preclinical and clinical studies. Administration of MSCs has consistently shown benefits with both symptomatic and histologic improvement in CTD animal models. MSC therapies in severe and drug-resistant CTD patients have shown promise in a number of the pilot studies, cohort studies, and randomized controlled trials in SLE, RA, and SSc, but some problems still need to be resolved in the transition from the bench to the bedside. The relevant studies in pSS and IIM are still in their infancy, but have displayed encouraging outcomes. Considerable efficacy variations have been observed in terms of the route of delivery, time of MSC injection, origin of the MSCs and dosage. Furthermore, the optimization of conventional drugs combined with MSC therapies and the applications of novel cell engineering approaches requires additional research. In this review, we summarize the current evidence about the immunoregulatory mechanism of MSCs, as well as the preclinical and clinical studies of MSC-based therapy for the treatment of CTDs.
Collapse
|
26
|
Ronniger M, Aguida B, Stacke C, Chen Y, Ehnert S, Erdmann N, Eschenburg G, Falldorf K, Pooam M, Wing A, Ahmad M. A Novel Method to Achieve Precision and Reproducibility in Exposure Parameters for Low-Frequency Pulsed Magnetic Fields in Human Cell Cultures. Bioengineering (Basel) 2022; 9:bioengineering9100595. [PMID: 36290562 PMCID: PMC9598188 DOI: 10.3390/bioengineering9100595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/30/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
The effects of extremely low-frequency electromagnetic field (ELF-MF) exposure on living systems have been widely studied at the fundamental level and also claimed as beneficial for the treatment of diseases for over 50 years. However, the underlying mechanisms and cellular targets of ELF-MF exposure remain poorly understood and the field has been plagued with controversy stemming from an endemic lack of reproducibility of published findings. To address this problem, we here demonstrate a technically simple and reproducible EMF exposure protocol to achieve a standardized experimental approach which can be readily adopted in any lab. As an assay system, we chose a commercially available inflammatory model human cell line; its response to magnetic fields involves changes in gene expression which can be monitored by a simple colorimetric reporter gene assay. The cells were seeded and cultured in microplates and inserted into a custom-built, semi-automated incubation and exposure system which accurately controls the incubation (temperature, humidity, CO2) and magnetic-field exposure conditions. A specific alternating magnetic field (<1.0% spatial variance) including far-field reduction provided defined exposure conditions at the position of each well of the microplate. To avoid artifacts, all environmental and magnetic-field exposure parameters were logged in real time throughout the duration of the experiment. Under these extensively controlled conditions, the effect of the magnetic field on the cell cultures as assayed by the standardized operating procedure was highly reproducible between experiments. As we could fully define the characteristics (frequency, intensity, duration) of the pulsed magnetic field signals at the position of the sample well, we were, for the first time, able to accurately determine the effect of changing single ELF-MF parameters such as signal shape, frequency, intensity and duty cycle on the biological response. One signal in particular (10 Hz, 50% duty cycle, rectangular, bipolar, 39.6μT) provided a significant reduction in cytokine reporter gene expression by 37% in our model cell culture line. In sum, the accuracy, environmental control and data-logging capacity of the semi-automated exposure system should greatly facilitate research into fundamental cellular response mechanisms and achieve the consistency necessary to bring ELF-MF/PEMF research results into the scientific mainstream.
Collapse
Affiliation(s)
- Michael Ronniger
- Sachtleben GmbH, 20251 Hamburg, Germany
- Correspondence: (M.R.); (M.A.); Tel.: +49-408-060-961-25 (M.R.); +33-014-427-2916 (M.A.)
| | - Blanche Aguida
- Photobiology Research Group, Sorbonne Université CNRS, 75005 Paris, France
| | | | - Yangmengfan Chen
- Department of Biology, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Sabrina Ehnert
- Department of Biology, Faculty of Science, Naresuan University, Phitsanulok 65000, Thailand
| | | | | | | | - Marootpong Pooam
- Siegfried Weller Institute for Trauma Research, Department of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
| | | | - Margaret Ahmad
- Photobiology Research Group, Sorbonne Université CNRS, 75005 Paris, France
- Correspondence: (M.R.); (M.A.); Tel.: +49-408-060-961-25 (M.R.); +33-014-427-2916 (M.A.)
| |
Collapse
|
27
|
Huang M, Li P, Chen F, Cai Z, Yang S, Zheng X, Li W. Is extremely low frequency pulsed electromagnetic fields applicable to gliomas? A literature review of the underlying mechanisms and application of extremely low frequency pulsed electromagnetic fields. Cancer Med 2022; 12:2187-2198. [PMID: 35929424 PMCID: PMC9939155 DOI: 10.1002/cam4.5112] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/07/2022] [Accepted: 07/19/2022] [Indexed: 11/07/2022] Open
Abstract
Gliomas refer to a group of complicated human brain tumors with a low 5-year survival rate and limited therapeutic options. Extremely low-frequency pulsed electromagnetic field (ELF-PEMF) is a specific magnetic field featuring almost no side effects. However, the application of ELF-PEMF in the treatment of gliomas is rare. This review summarizes five significant underlying mechanisms including calcium ions, autophagy, apoptosis, angiogenesis, and reactive oxygen species, and applications of ELF-PEMF in glioma treatment from a clinical practice perspective. In addition, the prospects of ELF-PEMF in combination with conventional therapy for the treatment of gliomas are reviewed. This review benefits any specialists, especially oncologists, interested in this new therapy because it can help treat patients with gliomas properly.
Collapse
Affiliation(s)
- Mengqian Huang
- Cancer Center, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Parker Li
- Clinical MedicineShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Feng Chen
- Cancer Center, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Zehao Cai
- Cancer Center, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Shoubo Yang
- Cancer Center, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Xiaohong Zheng
- Cancer Center, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Wenbin Li
- Cancer Center, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
28
|
Fang YY, Tu Q, Zhang YT, Liu J, Liu HG, Zhao ZH, Wu H, Yin TJ. Effect of Occupational Extremely Low-Frequency Electromagnetic Field Exposure on the Thyroid Gland of Workers: A Prospective Study. Curr Med Sci 2022; 42:817-823. [PMID: 35963949 DOI: 10.1007/s11596-022-2610-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/19/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the biological effects of occupational extremely low-frequency electromagnetic field (ELF-EMF) exposure on the thyroid gland. METHODS We conducted a prospective analysis of 85 workers (exposure group) exposed to an ELF-EMF (100 μT, 10-100 Hz) produced by the electromagnetic aircraft launch system and followed up on thyroid function indices, immunological indices, and color Doppler images for 3 years. Additionally, 116 healthy volunteers were randomly selected as controls (control group), the thyroid function of whom was compared to the exposure group. RESULTS No significant difference was observed in thyroid function between the exposure and control groups. During the follow-up of the exposure group, the serum free triiodothyronine (FT3) level was found to slowly decrease and free thyroxine (FT4) level slowly increase with increasing exposure time. However, no significant difference was found in thyroid-stimulating hormone (TSH) over the three years, and no significant difference was observed in the FT3, FT4 and TSH levels between different exposure subgroups. Furthermore, no significant changes were observed in thyroid autoantibody levels and ultrasound images between subgroups or over time. CONCLUSION Long-term exposure to ELF-EMF may promote thyroid secretion of T4 and inhibit deiodination of T4 to T3. ELF-EMF has no significant effect on thyroid immune function and morphology.
Collapse
Affiliation(s)
- Yuan-Yuan Fang
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Qian Tu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu-Ting Zhang
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian Liu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui-Guo Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhi-Hua Zhao
- Naval University of Engineering, Wuhan, 430030, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Tie-Jun Yin
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
29
|
Wang M, Li Y, Feng L, Zhang X, Wang H, Zhang N, Viohl I, Li G. Pulsed Electromagnetic Field Enhances Healing of a Meniscal Tear and Mitigates Posttraumatic Osteoarthritis in a Rat Model. Am J Sports Med 2022; 50:2722-2732. [PMID: 35834942 DOI: 10.1177/03635465221105874] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Meniscal tears in the avascular region are thought to rarely heal and are a considerable challenge to treat. Although the therapeutic effects of a pulsed electromagnetic field (PEMF) have been extensively studied in a variety of orthopaedic disorders, the effect of a PEMF on meniscal healing has not been reported. HYPOTHESIS PEMF treatment would promote meniscal healing and prevent osteoarthritis progression. STUDY DESIGN Controlled laboratory study. METHODS A total of 72 twelve-week-old male Sprague-Dawley rats with full-thickness longitudinal medial meniscal tears in the avascular region were divided into 3 groups: control (Gcon), treatment with a classic signal PEMF (Gclassic), and treatment with a high-slew rate signal PEMF (GHSR). Macroscopic observation and histological analysis of the meniscus and articular cartilage were performed to evaluate the meniscal healing and progression of osteoarthritis. The synovium was harvested for histological and immunofluorescent analysis to evaluate the intra-articular inflammation. Meniscal healing, articular cartilage degeneration, and synovitis were quantitatively evaluated according to their scoring systems. RESULTS Dramatic degenerative changes of the meniscus and articular cartilage were noticed during gross observation and histological evaluation in Gcon at 8 weeks. However, the menisci in the 2 treatment groups were restored to normal morphology, with a smooth surface and shiny white color. Particularly, the HSR signal remarkably enhanced the fibrochondrogenesis and accelerated the remodeling process of the regenerated tissue. The meniscal healing scores of the PEMF treatment groups were significantly higher than those in Gcon at 8 weeks. Specifically, the HSR signal showed a significantly higher meniscal repair score than did the classic signal at week 8 (P < .01). Additionally, the HSR signal significantly downregulated the secretion levels of interleukin 1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) in the meniscus and synovium as compared with the control group. When compared with the 2 treatment groups, Gcon had significantly higher degeneration scores (Gcon vs Gclassic, P < .0001; Gcon vs GHSR, P < .0001). The HSR signal also exhibited significantly lower synovitis scores compared with the other two groups (Gcon vs Gclassic, P < .0001; Gclassic vs GHSR, P = .0002). CONCLUSION A PEMF promoted the healing of meniscal tears in the avascular region and restored the injured meniscus to its structural integrity in a rat model. As compared with the classic signal, the HSR signal showed increased capability to promote fibrocartilaginous tissue formation and modulate the inflammatory environment, therefore protecting the knee joint from posttraumatic osteoarthritis development. CLINICAL RELEVANCE Adjuvant PEMF therapy may offer a new approach for the treatment of meniscal tears attributed to the enhanced meniscal repair and ameliorated osteoarthritis progression.
Collapse
Affiliation(s)
- Ming Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Yucong Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Lu Feng
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Xiaoting Zhang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Haixing Wang
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Nianli Zhang
- Research and Clinical Affairs, Orthofix Medical Inc, Lewisville, Texas, USA
| | - Ingmar Viohl
- Research and Clinical Affairs, Orthofix Medical Inc, Lewisville, Texas, USA
| | - Gang Li
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| |
Collapse
|
30
|
Interaction between Mesenchymal Stem Cells and the Immune System in Rheumatoid Arthritis. Pharmaceuticals (Basel) 2022; 15:ph15080941. [PMID: 36015088 PMCID: PMC9416102 DOI: 10.3390/ph15080941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that causes damage to joints. This review focuses on the possibility of influencing the disease through immunomodulation by mesenchymal stem cells (MSCs). There is an occurrence of rheumatoid factor and RA-specific autoantibodies to citrullinated proteins in most patients. Citrulline proteins have been identified in the joints of RA patients, and are considered to be the most suitable candidates for the stimulation of anti-citrulline protein antibodies production. Fibroblast-like proliferating active synoviocytes actively promote inflammation and destruction in the RA joint, in association with pro-inflammatory cells. The inflammatory process may be suppressed by MSCs, which are a population of adherent cells with the following characteristic phenotype: CD105+, CD73+, CD90+, CD45−, CD34− and HLA DR−. Following the stimulation process, MSCs are capable of immunomodulatory action through the release of bioactive molecules, as well as direct contact with the cells of the immune system. Furthermore, MSCs show the ability to suppress natural killer cell activation and dendritic cells maturation, inhibit T cell proliferation and function, and induce T regulatory cell formation. MSCs produce factors that suppress inflammatory processes, such as PGE2, TGF-β, HLA-G5, IDO, and IL-10. These properties suggest that MSCs may affect and suppress the excessive inflammation that occurs in RA. The effect of MSCs on rheumatoid arthritis has been proven to be a suitable alternative treatment thanks to successful experiments and clinical studies.
Collapse
|
31
|
Huang J, Zhao L, Xiong J, Liang Y. miRNA Expression Profile of Magnetic Field Combined with Nano-Scaffolds Enhance Chondrogenesis. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The differentiation of mesenchymal stem cells (MSC) into cartilage is a critical step in the cartilage damage repair. Pulsed electromagnetic fields may provide a solution to promote the differentiation and proliferation of MSC, with a promisingly strong synergistic effect apparent when
MSC subjected to magnetic nanoparticles and pulsed electromagnetic fields simultaneously. Previously, we developed a magnetic nanoparticles hydrogel (MagGel) that, when combined with magnetic fields, substantially enhanced cartilage tissue repair; however, we still know little about the mechanisms
underlying its effectiveness. Here, we examined the role of magnetic fields and nano-scaffolds in promoting differentiation of MSCs into chondrocytes. Our results suggest that the mechanism causing MSC differentiation under these conditions resulted from the activation of multiple miRNAs and
signaling pathways. These results can be built upon by future molecular studies to identify which miRNAs are involved in to chondrogenesis. By identifying these miRNAs and their targeted pathways, we can better manipulate the expression of the specific miRNAs that promote MSC differentiation
and cartilage repair. Our study provides a starting point for the study of magnetic field-induced differentiation of the magnetic nanoparticles within stem cells.
Collapse
Affiliation(s)
- Jianghong Huang
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen
University Health Science Center, Shenzhen, 518035, China
| | - Ling Zhao
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen
University Health Science Center, Shenzhen, 518035, China
| | - Jianyi Xiong
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen
University Health Science Center, Shenzhen, 518035, China
| | - Yujie Liang
- Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen Key Laboratory for Psychological Healthcare and Shenzhen Institute of Mental Health, Shenzhen, Guangdong, 518020, China
| |
Collapse
|
32
|
Diamagnetic Therapy in a Patient with Complex Regional Pain Syndrome Type I and Multiple Drug Intolerance: A Case Report. REPORTS 2022. [DOI: 10.3390/reports5020018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Complex regional pain syndrome (CRPS) is a neurologic chronic pain condition hard to diagnose and treat, and able to significantly impact the quality of life. Currently, the available multimodal, individualized treatments (i.e., pharmacological and non-pharmacological therapies including invasive procedures) are aimed only at symptom control. Herein, we report a 69-year-old Caucasian female who came to our attention due to a 3-year history of severe (10/10) burning pain in her right ankle, along with oedema and local changes in skin color and temperature, which occurred after the ankle sprain. Previous pharmacological attempts failed due to multiple drug intolerance. Clinical examination confirmed the CRPS type I diagnosis, and a weekly diamagnetic therapy protocol was started since the patient refused further medications and interventional procedures. After 10 weeks of treatment, a significant (p < 0.01) reduction in pain severity and absence of oedema (difference in ankles’ circumference: from 3 cm to 0) were observed, with consequent improvements in quality of life and no adverse events. Although high-quality clinical evidence is still lacking, our case report suggests further investigating the potential use of diamagnetic therapy as a non-invasive and safe adjunctive treatment for CRPS, and as an alternative when patients did not benefit from drugs and/or refuse invasive procedures.
Collapse
|
33
|
Lee CG, Park C, Hwang S, Hong JE, Jo M, Eom M, Lee Y, Rhee KJ. Pulsed Electromagnetic Field (PEMF) Treatment Reduces Lipopolysaccharide-Induced Septic Shock in Mice. Int J Mol Sci 2022; 23:ijms23105661. [PMID: 35628471 PMCID: PMC9147061 DOI: 10.3390/ijms23105661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 12/04/2022] Open
Abstract
Despite advances in medicine, mortality due to sepsis has not decreased. Pulsed electromagnetic field (PEMF) therapy is emerging as an alternative treatment in many inflammation-related diseases. However, there are few studies on the application of PEMF therapy to sepsis. In the current study, we examined the effect of PEMF therapy on a mouse model of lipopolysaccharide (LPS)-induced septic shock. Mice injected with LPS and treated with PEMF showed higher survival rates compared with the LPS group. The increased survival was correlated with decreased levels of pro-inflammatory cytokine mRNA expression and lower serum nitric oxide levels and nitric oxide synthase 2 mRNA expression in the liver compared with the LPS group. In the PEMF + LPS group, there was less organ damage in the liver, lungs, spleen, and kidneys compared to the LPS group. To identify potential gene targets of PEMF treatment, microarray analysis was performed, and the results showed that 136 genes were up-regulated, and 267 genes were down-regulated in the PEMF + LPS group compared to the LPS group. These results suggest that PEMF treatment can dramatically decrease septic shock through the reduction of pro-inflammatory cytokine gene expression. In a clinical setting, PEMF may provide a beneficial effect for patients with bacteria-induced sepsis and reduce septic shock-induced mortality.
Collapse
Affiliation(s)
- Chang-Gun Lee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
- Department of Medical Genetics, School of Medicine, Ajou University, Suwon 16499, Korea
| | - Chanoh Park
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
| | - Soonjae Hwang
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, GAIST, College of Medicine, Gachon University, Incheon 21999, Korea
| | - Ju-Eun Hong
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
| | - Minjeong Jo
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
| | - Minseob Eom
- Department of Pathology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
| | - Yongheum Lee
- Department of Biomedical Engineering, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea
- Correspondence: (Y.L.); (K.-J.R.); Tel.: +82-33-760-2863 (Y.L.); +82-33-760-2445 (K.-J.R.); Fax: +82-33-760-2561 (Y.L.); +82-33-760-2195 (K.-J.R.)
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University MIRAE Campus, Wonju 26493, Korea; (C.-G.L.); (C.P.); (S.H.); (J.-E.H.); (M.J.)
- Correspondence: (Y.L.); (K.-J.R.); Tel.: +82-33-760-2863 (Y.L.); +82-33-760-2445 (K.-J.R.); Fax: +82-33-760-2561 (Y.L.); +82-33-760-2195 (K.-J.R.)
| |
Collapse
|
34
|
Raman N, Imran SAM, Ahmad Amin Noordin KB, Zaman WSWK, Nordin F. Mechanotransduction in Mesenchymal Stem Cells (MSCs) Differentiation: A Review. Int J Mol Sci 2022; 23:4580. [PMID: 35562971 PMCID: PMC9105508 DOI: 10.3390/ijms23094580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/13/2022] [Accepted: 04/13/2022] [Indexed: 02/04/2023] Open
Abstract
Mechanotransduction is the process by which physical force is converted into a biochemical signal that is used in development and physiology; meanwhile, it is intended for the ability of cells to sense and respond to mechanical forces by activating intracellular signals transduction pathways and the relative phenotypic adaptation. It encompasses the role of mechanical stimuli for developmental, morphological characteristics, and biological processes in different organs; the response of cells to mechanically induced force is now also emerging as a major determinant of disease. Due to fluid shear stress caused by blood flowing tangentially across the lumen surface, cells of the cardiovascular system are typically exposed to a variety of mechanotransduction. In the body, tissues are continuously exposed to physical forces ranging from compression to strain, which is caused by fluid pressure and compressive forces. Only lately, though, has the importance of how forces shape stem cell differentiation into lineage-committed cells and how mechanical forces can cause or exacerbate disease besides organizing cells into tissues been acknowledged. Mesenchymal stem cells (MSCs) are potent mediators of cardiac repair which can secret a large array of soluble factors that have been shown to play a huge role in tissue repair. Differentiation of MSCs is required to regulate mechanical factors such as fluid shear stress, mechanical strain, and the rigidity of the extracellular matrix through various signaling pathways for their use in regenerative medicine. In the present review, we highlighted mechanical influences on the differentiation of MSCs and the general factors involved in MSCs differentiation. The purpose of this study is to demonstrate the progress that has been achieved in understanding how MSCs perceive and react to their mechanical environment, as well as to highlight areas where more research has been performed in previous studies to fill in the gaps.
Collapse
Affiliation(s)
- Narmadaa Raman
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (N.R.); (S.A.M.I.)
- Department of Microbiology, Faculty of Science, Universiti Tunku Abdul Rahman, Kampar 31900, Malaysia
| | - Siti A. M. Imran
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (N.R.); (S.A.M.I.)
| | | | | | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (N.R.); (S.A.M.I.)
| |
Collapse
|
35
|
Babaniamansour P, Salimi M, Dorkoosh F, Mohammadi M. Magnetic Hydrogel for Cartilage Tissue Regeneration as well as a Review on Advantages and Disadvantages of Different Cartilage Repair Strategies. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7230354. [PMID: 35434125 PMCID: PMC9012656 DOI: 10.1155/2022/7230354] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/06/2022] [Accepted: 03/11/2022] [Indexed: 01/21/2023]
Abstract
There is a clear clinical need for efficient cartilage healing strategies for treating cartilage defects which burdens millions of patients physically and financially. Different strategies including microfracture technique, osteochondral transfer, and scaffold-based treatments have been suggested for curing cartilage injuries. Although some improvements have been achieved in several facets, current treatments are still less than satisfactory. Recently, different hydrogel-based biomaterials have been suggested as a therapeutic candidate for cartilage tissue regeneration due to their biocompatibility, high water content, and tunability. Specifically, magnetic hydrogels are becoming more attractive due to their smart response to magnetic fields remotely. We seek to outline the context-specific regenerative potential of magnetic hydrogels for cartilage tissue repair. In this review, first, we explained conventional techniques for cartilage repair and then compared them with new scaffold-based approaches. We illustrated various hydrogels used for cartilage regeneration by highlighting the magnetic hydrogels. Also, we gathered in vitro and in vivo studies of how magnetic hydrogels promote chondrogenesis as well as studied the biological mechanism which is responsible for cartilage repair due to the application of magnetic hydrogel.
Collapse
Affiliation(s)
- Parto Babaniamansour
- Department of Biomedical Engineering, AmirKabir University of Technology, Tehran, Iran
| | - Maryam Salimi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Bone and Joint Diseases Research Center, Department of Orthopedic Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farid Dorkoosh
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutics, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Mohammadi
- Department of Biomedical Engineering, University of Isfahan, Isfahan, Iran
| |
Collapse
|
36
|
Bioengineered Living Bone Grafts-A Concise Review on Bioreactors and Production Techniques In Vitro. Int J Mol Sci 2022; 23:ijms23031765. [PMID: 35163687 PMCID: PMC8836415 DOI: 10.3390/ijms23031765] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/29/2022] [Accepted: 01/30/2022] [Indexed: 12/26/2022] Open
Abstract
It has been observed that bone fractures carry a risk of high mortality and morbidity. The deployment of a proper bone healing method is essential to achieve the desired success. Over the years, bone tissue engineering (BTE) has appeared to be a very promising approach aimed at restoring bone defects. The main role of the BTE is to apply new, efficient, and functional bone regeneration therapy via a combination of bone scaffolds with cells and/or healing promotive factors (e.g., growth factors and bioactive agents). The modern approach involves also the production of living bone grafts in vitro by long-term culture of cell-seeded biomaterials, often with the use of bioreactors. This review presents the most recent findings concerning biomaterials, cells, and techniques used for the production of living bone grafts under in vitro conditions. Particular attention has been given to features of known bioreactor systems currently used in BTE: perfusion bioreactors, rotating bioreactors, and spinner flask bioreactors. Although bioreactor systems are still characterized by some limitations, they are excellent platforms to form bioengineered living bone grafts in vitro for bone fracture regeneration. Moreover, the review article also describes the types of biomaterials and sources of cells that can be used in BTE as well as the role of three-dimensional bioprinting and pulsed electromagnetic fields in both bone healing and BTE.
Collapse
|
37
|
Mohamed NA, Marei I, Crovella S, Abou-Saleh H. Recent Developments in Nanomaterials-Based Drug Delivery and Upgrading Treatment of Cardiovascular Diseases. Int J Mol Sci 2022; 23:1404. [PMID: 35163328 PMCID: PMC8836006 DOI: 10.3390/ijms23031404] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 01/27/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading causes of morbidity and mortality worldwide. However, despite the recent developments in the management of CVDs, the early and long outcomes vary considerably in patients, especially with the current challenges facing the detection and treatment of CVDs. This disparity is due to a lack of advanced diagnostic tools and targeted therapies, requiring innovative and alternative methods. Nanotechnology offers the opportunity to use nanomaterials in improving health and controlling diseases. Notably, nanotechnologies have recognized potential applicability in managing chronic diseases in the past few years, especially cancer and CVDs. Of particular interest is the use of nanoparticles as drug carriers to increase the pharmaco-efficacy and safety of conventional therapies. Different strategies have been proposed to use nanoparticles as drug carriers in CVDs; however, controversies regarding the selection of nanomaterials and nanoformulation are slowing their clinical translation. Therefore, this review focuses on nanotechnology for drug delivery and the application of nanomedicine in CVDs.
Collapse
Affiliation(s)
- Nura A. Mohamed
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Isra Marei
- Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London SW7 2AZ, UK;
- Department of Pharmacology, Weill Cornell Medicine in Qatar, Doha P.O. Box 24144, Qatar
| | - Sergio Crovella
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Haissam Abou-Saleh
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
- Biomedical Research Center (BRC), Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
38
|
Abstract
In 2001, the concept of the neurovascular unit was introduced at the Stroke Progress Review Group meeting. The neurovascular unit is an important element of the health and disease status of blood vessels and nerves in the central nervous system. Since then, the neurovascular unit has attracted increasing interest from research teams, who have contributed greatly to the prevention, treatment, and prognosis of stroke and neurodegenerative diseases. However, additional research is needed to establish an efficient, low-cost, and low-energy in vitro model of the neurovascular unit, as well as enable noninvasive observation of neurovascular units in vivo and in vitro. In this review, we first summarize the composition of neurovascular units, then investigate the efficacy of different types of stem cells and cell culture methods in the construction of neurovascular unit models, and finally assess the progress of imaging methods used to observe neurovascular units in recent years and their positive role in the monitoring and investigation of the mechanisms of a variety of central nervous system diseases.
Collapse
Affiliation(s)
- Taiwei Dong
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Min Li
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Feng Gao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Peifeng Wei
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Jian Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Provinve, China
| |
Collapse
|
39
|
Zhu H, Zhuang Y, Li D, Dong N, Ma H, Liu L, Shi Q, Ju X. Cryo-Temperature Pretreatment Increases the Pro-Angiogenic Capacity of Three-Dimensional Mesenchymal Stem Cells via the PI3K-AKT Pathway. Cell Transplant 2022; 31:9636897221106996. [PMID: 35727010 PMCID: PMC9218451 DOI: 10.1177/09636897221106996] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
To increase the potential and effectiveness of three-dimensional (3D) mesenchymal stem cells (MSCs) for clinical applications, this study explored the effects of short cryo-temperature pretreatment on MSC function. Adipose-derived MSCs (A-MSCs) were cultured via the ordinary monolayer method and 3D hanging drop spheroid method. When the cells adhered to the wall or formed a spheroid, they were subjected to hypothermic stress at 4°C for 1 h and then divided into three recovery periods at 37°C, specifically 0, 12, and 24 h. The control group was not subjected to any treatment throughout the study. Monolayer and 3D spheroid A-MSCs were analyzed via RNA sequencing after hypothermic stress at 4°C for 1 h. Subsequently, each group of cells was collected and subjected to phenotype identification via flow cytometry, and mRNA expression was detected via reverse transcription-quantitative polymerase chain reaction analysis. Western blot analysis was performed to analyze the PI3K-AKT signaling pathway in A-MSCs. The effects of A-MSCs on angiogenesis in vivo were examined using a chick chorioallantoic membrane assay. Transwell assays were performed to determine whether the culture supernatant from each group could induce the chemotaxis of human umbilical vein endothelial cells (HUVECs). Three-dimensional spheroid culture did not change the phenotype of A-MSCs. The expression of fibroblast growth factors, hepatocyte growth factors, and other angiogenesis-related factors in A-MSCs was upregulated. A-MSCs subjected to hypothermic stress promoted angiogenesis under both monolayer and 3D spheroid cultures. Moreover, the chemotaxis of HUVECs to the 3D spheroid culture supernatant increased substantially. Short cryo-temperature pretreatment could stimulate 3D spheroid A-MSCs and activate the PI3K-AKT pathway. This approach has the advantages of promoting angiogenesis and maintaining cell viability.
Collapse
Affiliation(s)
- Huasu Zhu
- Department of Pediatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Yong Zhuang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Dong Li
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Na Dong
- Department of Pediatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Huixian Ma
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Linghong Liu
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Qing Shi
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan, China
| | - Xiuli Ju
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan, China
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
- Xiuli Ju, Department of Pediatrics, Qilu Hospital of Shandong University, No. 107 Wenhua West Road, Jinan 250012, Shandong, China.
| |
Collapse
|
40
|
Weaver JS, Vina ER, Munk PL, Klauser AS, Elifritz JM, Taljanovic MS. Gouty Arthropathy: Review of Clinical Manifestations and Treatment, with Emphasis on Imaging. J Clin Med 2021; 11:jcm11010166. [PMID: 35011907 PMCID: PMC8745871 DOI: 10.3390/jcm11010166] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/16/2021] [Accepted: 12/26/2021] [Indexed: 12/22/2022] Open
Abstract
Gout, a crystalline arthropathy caused by the deposition of monosodium urate crystals in the articular and periarticular soft tissues, is a frequent cause of painful arthropathy. Imaging has an important role in the initial evaluation as well as the treatment and follow up of gouty arthropathy. The imaging findings of gouty arthropathy on radiography, ultrasonography, computed tomography, dual energy computed tomography, and magnetic resonance imaging are described to include findings of the early, acute and chronic phases of gout. These findings include early monosodium urate deposits, osseous erosions, and tophi, which may involve periarticular tissues, tendons, and bursae. Treatment of gout includes non-steroidal anti-inflammatories, colchicine, glucocorticoids, interleukin-1 inhibitors, xanthine oxidase inhibitors, uricosuric drugs, and recombinant uricase. Imaging is critical in monitoring response to therapy; clinical management can be modulated based on imaging findings. This review article describes the current standard of care in imaging and treatment of gouty arthropathy.
Collapse
Affiliation(s)
- Jennifer S. Weaver
- Department of Radiology, University of New Mexico, Albuquerque, NM 87131, USA;
- Correspondence:
| | - Ernest R. Vina
- Department of Medicine, University of Arizona Arthritis Center, Tucson, AZ 85724, USA;
| | - Peter L. Munk
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
- Department of Radiology, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
| | - Andrea S. Klauser
- Radiology Department, Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria;
| | - Jamie M. Elifritz
- Departments of Radiology and Pathology, University of New Mexico, Albuquerque, NM 87131, USA;
- New Mexico Office of the Medical Investigator, Albuquerque, NM 87131, USA
| | - Mihra S. Taljanovic
- Department of Radiology, University of New Mexico, Albuquerque, NM 87131, USA;
- Departments of Medical Imaging and Orthopaedic Surgery, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
41
|
Yan J, Liu C, Tu C, Zhang R, Tang X, Li H, Wang H, Ma Y, Zhang Y, Wu H, Sheng G. Hydrogel-hydroxyapatite-monomeric collagen type-I scaffold with low-frequency electromagnetic field treatment enhances osteochondral repair in rabbits. Stem Cell Res Ther 2021; 12:572. [PMID: 34774092 PMCID: PMC8590294 DOI: 10.1186/s13287-021-02638-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/25/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cartilage damage is a common medical issue in clinical practice. Complete cartilage repair remains a significant challenge owing to the inferior quality of regenerative tissue. Safe and non-invasive magnetic therapy combined with tissue engineering to repair cartilage may be a promising breakthrough. METHODS In this study, a composite scaffold made of Hydroxyapatite-Collagen type-I (HAC) and PLGA-PEG-PLGA thermogel was produced to match the cartilage and subchondral layers in osteochondral defects, respectively. Bone marrow mesenchymal stem cells (BMSC) encapsulated in the thermogel were stimulated by an electromagnetic field (EMF). Effect of EMF on the proliferation and chondrogenic differentiation potential was evaluated in vitro. 4 mm femoral condyle defect was constructed in rabbits. The scaffolds loaded with BMSCs were implanted into the defects with or without EMF treatment. Effects of the combination treatment of the EMF and composite scaffold on rabbit osteochondral defect was detected in vivo. RESULTS In vitro experiments showed that EMF could promote proliferation and chondrogenic differentiation of BMSCs partly by activating the PI3K/AKT/mTOR and Wnt1/LRP6/β-catenin signaling pathway. In vivo results further confirmed that the scaffold with EMF enhances the repair of osteochondral defects in rabbits, and, in particular, cartilage repair. CONCLUSION Hydrogel-Hydroxyapatite-Monomeric Collagen type-I scaffold with low-frequency EMF treatment has the potential to enhance osteochondral repair.
Collapse
Affiliation(s)
- Jiyuan Yan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China
| | - Chaoxu Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China
| | - Chang Tu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Ruizhuo Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China
| | - Xiangyu Tang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Hao Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China
| | - Huaixi Wang
- Department of Spine and Spinal Cord Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan, Zhengzhou, People's Republic of China
| | - Yongzhuang Ma
- Department of Orthopedics, Shanxi Bethune Hospital, Taiyuan, Shanxi, People's Republic of China
| | - Yingchi Zhang
- Department of Traumatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China.
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China.
| | - Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
42
|
Lv X, Wang L, Zou X, Huang S. Umbilical Cord Mesenchymal Stem Cell Therapy for Regenerative Treatment of Rheumatoid Arthritis: Opportunities and Challenges. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3927-3936. [PMID: 34584402 PMCID: PMC8462093 DOI: 10.2147/dddt.s323107] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/26/2021] [Indexed: 12/25/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease of unknown etiology with a high rate of disability. Traditional treatments for RA remain a challenging issue. For example, nonsteroidal anti-inflammatory drugs (NSAIDs) have no therapeutic effects on joint destruction, and the prominent side effects include gastrointestinal symptoms. RA is characterized by recurrence and bone attrition. Therefore, regenerative medicine and the use of umbilical cord mesenchymal stem cell (UC-MSC) therapies have recently emerged as potential options. UC-MSCs are multifunctional stem cells that are present in neonatal umbilical cord tissue and can differentiate into many kinds of cells, which have broad clinical application prospects in the tissue engineering of bone, cartilage, muscle, tendon, ligament, nerve, liver, endothelium, and myocardium. Moreover, UC-MSCs have advantages, such as convenient collection of materials and no ethical disputes; thus, these cells have attracted increasing attention from researchers. However, there are few clinical studies regarding UC-MSC therapy for RA. In this paper, we will review traditional drugs for RA treatment and then focus on UC-MSC therapy for RA, including preclinical and clinical UC-MSC applications for RA patients in the context of regenerative medicine. Finally, we will summarize the challenges and perspectives of UC-MSCs as a potential therapeutic strategy for RA. This review will help to design and discover more potent and efficacious treatments for RA patients and aid in advancing this class of cell therapy.
Collapse
Affiliation(s)
- Xiaolan Lv
- Department of Laboratory Medicine, Liuzhou Maternity and Child Healthcare Hospital, Liu Zhou, Guang Xi, People's Republic of China
| | - Liming Wang
- Shaanxi Jiuzhou Biomedical Science and Technology Group, Xi'an, Shaan Xi, People's Republic of China
| | - XiaoRong Zou
- Department of Hematology, 986 Hospital of Fourth Military Medical University, Xi'an, Shaan Xi, People's Republic of China
| | - Shigao Huang
- Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| |
Collapse
|
43
|
Luo X, Cui J, Long X, Chen Z. TLRs Play Crucial Roles in Regulating RA Synoviocyte. Endocr Metab Immune Disord Drug Targets 2021; 20:1156-1165. [PMID: 32338225 DOI: 10.2174/1871530320666200427115225] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/23/2020] [Accepted: 01/30/2020] [Indexed: 12/13/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease comparing the inflammation of synovium. Macrophage-like synoviocytes and fibroblast-like synoviocytes (synoviocytes) are crucial ingredients of synovium. Therein, a lot of research has focused on synoviocytes. Researches demonstrated that TLR1, TLR2, TLR3, TLR4, TLR5, TLR6 TLR7 and TLR9 are expressed in synoviocyte. Additionally, the expression of TLR2, TLR3, TLR4 and TLR5 is increased in RA synoviocyte. In this paper, we review the exact role of TLR2, TLR3, TLR4 and TLR5 participate in regulating the production of inflammatory factors in RA synoviocyte. Furthermore, we discuss the role of vasoactive intestinal peptide (VIP), MicroRNA, Monome of Chinese herb and other cells (Monocyte and T cell) influence the function of synoviocyte by regulating TLRs. The activation of toll-like receptors (TLRs) in synoviocyte leads to the aggravation of arthritis, comparing with angiogenesis and bone destruction. Above all, TLRs are promising targets for managing RA.
Collapse
Affiliation(s)
- Xuling Luo
- Department of Orthopaedics, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Juncheng Cui
- Department of Orthopaedics, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Xin Long
- Department of Orthopaedics, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Zhiwei Chen
- Department of Orthopaedics, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| |
Collapse
|
44
|
Mansourian M, Shanei A. Evaluation of Pulsed Electromagnetic Field Effects: A Systematic Review and Meta-Analysis on Highlights of Two Decades of Research In Vitro Studies. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6647497. [PMID: 34368353 PMCID: PMC8342182 DOI: 10.1155/2021/6647497] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 05/30/2021] [Accepted: 06/27/2021] [Indexed: 12/13/2022]
Abstract
Pulsed electromagnetic field (PEMF) therapy is a type of physical stimulation that affects biological systems by producing interfering or coherent fields. Given that cell types are significantly distinct, which represents an important factor in stimulation, and that PEMFs can have different effects in terms of frequency and intensity, time of exposure, and waveform. This study is aimed at investigating if distinct positive and negative responses would correspond to specific characteristics of cells, frequency and flux density, time of exposure, and waveform. Necessary data were abstracted from the experimental observations of cell-based in vitro models. The observations were obtained from 92 publications between the years 1999 and 2019, which are available on PubMed and Web of Science databases. From each of the included studies, type of cells, pulse frequency of exposure, exposure flux density, and assayed cell responses were extracted. According to the obtained data, most of the experiments were carried out on human cells, and out of 2421 human cell experiments, cell changes were observed only in 51.05% of the data. In addition, the results pointed out the potential effects of PEMFs on some human cell types such as MG-63 human osteosarcoma cells (p value < 0.001) and bone marrow mesenchymal stem cells. However, human osteogenic sarcoma SaOS-2 (p < 0.001) and human adipose-derived mesenchymal stem cells (AD-MSCs) showed less sensitivity to PEMFs. Nevertheless, the evidence suggests that frequencies higher than 100 Hz, flux densities between 1 and 10 mT, and chronic exposure more than 10 days would be more effective in establishing a cellular response. This study successfully reported useful information about the role of cell type and signal characteristic parameters, which were of high importance for targeted therapies using PEMFs. Our findings would provide a deeper understanding about the effect of PEMFs in vitro, which could be useful as a reference for many in vivo experiments or preclinical trials.
Collapse
Affiliation(s)
- Mahsa Mansourian
- Department of Medical Physics, Faculty of Medicine, Isfahan University of Medical Science, Isfahan, Hezar Jerib Avenue, Isfahan, Iran
| | - Ahmad Shanei
- Department of Medical Physics, Faculty of Medical Science, Isfahan University of Medical Science, Isfahan, Hezar Jerib Street, Isfahan, Iran
| |
Collapse
|
45
|
Ross C, Overholt T, Xu R, Badlani G, Evans RJ, Matthews CA, Walker SJ. Pulsed electromagnetic field (PEMF) as an adjunct therapy for pain management in interstitial cystitis/bladder pain syndrome. Int Urogynecol J 2021; 33:487-491. [PMID: 34100976 DOI: 10.1007/s00192-021-04862-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION AND HYPOTHESIS Patients with interstitial cystitis/bladder pain syndrome (IC/BPS) often experience chronic pelvic and even systemic pain that can be difficult to clinically manage. Pulsed electromagnetic field (PEMF) therapy, a non-invasive strategy that has shown significant efficacy for pain reduction in other chronic pain conditions, may provide benefit for pain management in patients with IC/BPS. METHODS PEMF delivery to patients occurs via a bio-electromagnetic-energy device which consists of a flexible mat (180 × 50 cm) that the patient lies on for systemic, full-body delivery and/or a flexible pad (50 × 15 cm) for targeted delivery to a specific body region (e.g., pelvic area). The duration of individual sessions, number of sessions per day, total number of sessions, and follow-up observation period vary between previously published studies. Positive outcomes are typically reported as a significant reduction in visual analog scale (VAS) pain score and functional improvement assessed using validated questionnaires specific to the condition under study. RESULTS AND CONCLUSIONS The use of PEMF has been evaluated as a therapeutic strategy for pain management in several clinical scenarios. Randomized, double-blinded, placebo-controlled trials have reported positive efficacy and safety profiles when PEMF was used to treat non-specific low back pain, patellofemoral pain syndrome, chronic post-operative pain, osteoarthritis-related pain, rheumatoid arthritis-related pain, and fibromyalgia-related pain. Based on these positive outcomes in a variety of pain conditions, clinical trials to evaluate whether PEMF can provide a safe, non-invasive therapeutic approach to improve symptoms of chronic pain and fatigue in patients with IC/BPS are warranted.
Collapse
Affiliation(s)
- Christina Ross
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, 391 Technology Way, Winston Salem, NC, 27101, USA
| | - Tyler Overholt
- Department of Urology/Female Pelvic Health, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Raymond Xu
- Department of Urology/Female Pelvic Health, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Gopal Badlani
- Department of Urology/Female Pelvic Health, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Robert J Evans
- Department of Urology/Female Pelvic Health, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Catherine A Matthews
- Department of Urology/Female Pelvic Health, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA
| | - Stephen J Walker
- Wake Forest Institute for Regenerative Medicine, Wake Forest Baptist Medical Center, 391 Technology Way, Winston Salem, NC, 27101, USA. .,Department of Urology/Female Pelvic Health, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA.
| |
Collapse
|
46
|
Kaszuba-Zwoińska J, Novak P, Nowak B, Furgała A, Wójcik-Piotrowicz K, Piszczek P, Guzdek P, Pytko-Polończyk J. Low-frequency electromagnetic field influences human oral mucosa keratinocyte viability in response to lipopolysaccharide or minocycline treatment in cell culture conditions. Biomed Pharmacother 2021; 137:111340. [DOI: 10.1016/j.biopha.2021.111340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 01/09/2023] Open
|
47
|
Pooam M, Aguida B, Drahy S, Jourdan N, Ahmad M. Therapeutic application of light and electromagnetic fields to reduce hyper-inflammation triggered by COVID-19. Commun Integr Biol 2021; 14:66-77. [PMID: 33995820 PMCID: PMC8096326 DOI: 10.1080/19420889.2021.1911413] [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: 03/05/2021] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 12/17/2022] Open
Abstract
COVID-19 - related morbidity is associated with exaggerated inflammation and cytokine production in the lungs, leading to acute respiratory failure. The cellular mechanisms underlying these so-called 'cytokine storms' are regulated through the Toll-like receptor 4 (TLR4) signaling pathway and by ROS (Reactive Oxygen Species). Both light (Photobiomodulation) and magnetic fields (e.g., Pulsed Electro Magnetic Field) stimulation are noninvasive therapies known to confer anti-inflammatory effects and regulate ROS signaling pathways. Here we show that daily exposure to two 10-minute intervals of moderate intensity infra-red light significantly lowered the inflammatory response induced via the TLR4 receptor signaling pathway in human cell cultures. Anti-inflammatory effects were likewise achieved by electromagnetic field exposure of cells to daily 10-minute intervals of either Pulsed Electromagnetic Fields (PEMF), or to Low-Level static magnetic fields. Because current illumination and electromagnetic field therapies have no known side effects, and are already approved for some medical uses, we have here developed protocols for verification in clinical trials of COVID-19 infection. These treatments are affordable, simple to implement, and may help to resolve the acute respiratory distress of COVID-19 patients both in the home and in the hospital.
Collapse
Affiliation(s)
- Marootpong Pooam
- Photobiology Research Group, Sorbonne Université - CNRS, Paris, France
- Department of Biology, Faculty of Science, Naresuan University, Phitsanulok, Thailand
| | - Blanche Aguida
- Photobiology Research Group, Sorbonne Université - CNRS, Paris, France
| | - Soria Drahy
- Photobiology Research Group, Sorbonne Université - CNRS, Paris, France
| | - Nathalie Jourdan
- Photobiology Research Group, Sorbonne Université - CNRS, Paris, France
| | - Margaret Ahmad
- Photobiology Research Group, Sorbonne Université - CNRS, Paris, France
- Xavier University, Cincinnati, Ohio, U.S.A
| |
Collapse
|
48
|
Markov A, Thangavelu L, Aravindhan S, Zekiy AO, Jarahian M, Chartrand MS, Pathak Y, Marofi F, Shamlou S, Hassanzadeh A. Mesenchymal stem/stromal cells as a valuable source for the treatment of immune-mediated disorders. Stem Cell Res Ther 2021; 12:192. [PMID: 33736695 PMCID: PMC7971361 DOI: 10.1186/s13287-021-02265-1] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Over recent years, mesenchymal stem/stromal cells (MSCs) and their potential biomedical applications have received much attention from the global scientific community in an increasing manner. Firstly, MSCs were successfully isolated from human bone marrow (BM), but in the next steps, they were also extracted from other sources, mostly from the umbilical cord (UC) and adipose tissue (AT). The International Society for Cellular Therapy (ISCT) has suggested minimum criteria to identify and characterize MSCs as follows: plastic adherence, surface expression of CD73, D90, CD105 in the lack of expression of CD14, CD34, CD45, and human leucocyte antigen-DR (HLA-DR), and also the capability to differentiate to multiple cell types including adipocyte, chondrocyte, or osteoblast in vitro depends on culture conditions. However, these distinct properties, including self-renewability, multipotency, and easy accessibility are just one side of the coin; another side is their huge secretome which is comprised of hundreds of mediators, cytokines, and signaling molecules and can effectively modulate the inflammatory responses and control the infiltration process that finally leads to a regulated tissue repair/healing or regeneration process. MSC-mediated immunomodulation is a direct result of a harmonic synergy of MSC-released signaling molecules (i.e., mediators, cytokines, and chemokines), the reaction of immune cells and other target cells to those molecules, and also feedback in the MSC-molecule-target cell axis. These features make MSCs a respectable and eligible therapeutic candidate to be evaluated in immune-mediated disorders, such as graft versus host diseases (GVHD), multiple sclerosis (MS), Crohn's disease (CD), and osteoarthritis (OA), and even in immune-dysregulating infectious diseases such as the novel coronavirus disease 2019 (COVID-19). This paper discussed the therapeutic applications of MSC secretome and its biomedical aspects related to immune-mediated conditions. Sources for MSC extraction, their migration and homing properties, therapeutic molecules released by MSCs, and the pathways and molecular mechanisms possibly involved in the exceptional immunoregulatory competence of MSCs were discussed. Besides, the novel discoveries and recent findings on immunomodulatory plasticity of MSCs, clinical applications, and the methods required for their use as an effective therapeutic option in patients with immune-mediated/immune-dysregulating diseases were highlighted.
Collapse
Affiliation(s)
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Surendar Aravindhan
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Angelina Olegovna Zekiy
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit (G401), 69120 Heidelberg, Germany
| | | | - Yashwant Pathak
- Professor and Associate Dean for Faculty Affairs, Taneja College of Pharmacy, University of South Florida, Tampa, FL USA
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Shamlou
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Regenerative Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Benya PD, Kavanaugh A, Zakarian M, Söderlind P, Jashashvili T, Zhang N, Waldorff EI, Ryaby JT, Billi F. Pulsed electromagnetic field (PEMF) transiently stimulates the rate of mineralization in a 3-dimensional ring culture model of osteogenesis. PLoS One 2021; 16:e0244223. [PMID: 33539401 PMCID: PMC7861434 DOI: 10.1371/journal.pone.0244223] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/04/2020] [Indexed: 11/25/2022] Open
Abstract
Pulsed Electromagnetic Field (PEMF) has shown efficacy in bone repair and yet the optimum characteristics of this modality and its molecular mechanism remain unclear. To determine the effects of timing of PEMF treatment, we present a novel three-dimensional culture model of osteogenesis that demonstrates strong de novo generation of collagen and mineral matrix and exhibits stimulation by PEMF in multiple stages over 62 days of culture. Mouse postnatal day 2 calvarial pre-osteoblasts were cast within and around Teflon rings by polymerization of fibrinogen and cultured suspended without contact with tissue culture plastic. Ring constructs were exposed to PEMF for 4h/day for the entire culture (Daily), or just during Day1-Day10, Day11-Day 27, or Day28-Day63 and cultured without PEMF for the preceding or remaining days, and compared to no-PEMF controls. PEMF was conducted as HF Physio, 40.85 kHz frequency with a 67 ms burst period and an amplitude of 1.19 mT. Osteogenesis was kinetically monitored by repeated fluorescence measurements of continuously present Alizarin Red S (ARS) and periodically confirmed by micro-CT. PEMF treatment induced early-onset and statistically significant transient stimulation (~4-fold) of the mineralization rate when PEMF was applied Daily, or during D1-D10 and D11-D27. Stimulation was apparent but not significant between D28-D63 by ARS but was significant at D63 by micro-CT. PEMF also shifted the micro-CT density profiles to higher densities in each PEMF treatment group. Ring culture generated tissue with a mineral:matrix ratio of 2.0 by thermogravimetric analysis (80% of the calvaria control), and the deposited crystal structure was 50% hydroxyapatite by X-ray diffraction (63% of the calvaria and femur controls), independent of PEMF. These results were consistent with backscatter, secondary electron, and elemental analysis by scanning electron microscopy. Thus, in a defined, strong osteogenic environment, PEMF applied at different times was capable of further stimulation of osteogenesis with the potential to enhance bone repair.
Collapse
Affiliation(s)
- Paul D. Benya
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Aaron Kavanaugh
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Martin Zakarian
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Philip Söderlind
- Department of Architecture and Urban Design, University of California Los Angeles, Los Angeles, California, United States of America
| | - Tea Jashashvili
- Department of Radiology, Molecular Imaging Center, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Nianli Zhang
- Orthofix Medical Inc., Lewisville, Texas, United States of America
| | - Erik I. Waldorff
- Orthofix Medical Inc., Lewisville, Texas, United States of America
| | - James T. Ryaby
- Orthofix Medical Inc., Lewisville, Texas, United States of America
| | - Fabrizio Billi
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
50
|
Emelianov VY, Preobrazhenskaia EV, Nikolaev NS. Evaluating the Effectiveness of Biophysical Methods of Osteogenesis Stimulation: Review. TRAUMATOLOGY AND ORTHOPEDICS OF RUSSIA 2021; 27:86-96. [DOI: https:/doi.org/10.21823/2311-2905-2021-27-1-86-96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Background. Stimulation of osteogenesis (SO) by biophysical methods has been widely used in practice to accelerate healing or stimulate the healing of fractures with non-unions, since the middle of the XIX century. SO can be carried out by direct current electrostimulation, or indirectly by low-intensity pulsed ultrasound, capacitive electrical coupling stimulation, and pulsed electromagnetic field stimulation. SO simulates natural physiological processes: in the case of electrical stimulation, it changes the electromagnetic potential of damaged cell tissues in a manner similar to normal healing processes, or in the case of low-intensity pulsed ultrasound, it produces weak mechanical effects on the fracture area. SO increases the expression of factors and signaling pathways responsible for tissue regeneration and bone mineralization and ultimately accelerates bone union.The purpose of this review was to present the most up-to-date data from laboratory and clinical studies of the effectiveness of SO.Material and Methods. The results of laboratory studies and the final results of metaanalyses for each of the four SO methods published from 1959 to 2020 in the PubMed, EMBASE, and eLibrary databases are reviewed.Conclusion. The use of SO effectively stimulates the healing of fractures with the correct location of the sensors, compliance with the intensity and time of exposure, as well as the timing of use for certain types of fractures. In case of non-union or delayed union of fractures, spondylodesis, arthrodesis, preference should be given to non-invasive methods of SO. Invasive direct current stimulation can be useful for non-union of long bones, spondylodesis with the risk of developing pseudoarthrosis.
Collapse
|