1
|
Wang HY, Chen YJ, Huang IC, Lin CR, Lin KL, Chen CH. The effectiveness of pulsed electromagnetic field therapy in patients with shoulder impingement syndrome: A systematic review and meta-analysis of randomized controlled trials. PLoS One 2025; 20:e0323837. [PMID: 40388433 PMCID: PMC12088032 DOI: 10.1371/journal.pone.0323837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 04/16/2025] [Indexed: 05/21/2025] Open
Abstract
We performed a systematic review and meta-analysis to assess the efficacy of pulsed electromagnetic field (PEMF) therapy in treating patients with shoulder impingement syndrome. We sourced data from PubMed, the Cochrane Library, and Embase databases up until June 19, 2024. Our analysis included randomized controlled trials (RCTs) that evaluated the impact of PEMF therapy on pain levels and functional capacity in these patients. In total, four RCTs involving 252 participants were included. The pooled data indicated that PEMF therapy significantly reduced short-term pain (standardized mean difference [SMD] = -0.34, 95% confidence interval [CI] = -0.66 to -0.01, three RCTs, n = 166) and improved both short-term (SMD = 0.4, 95% CI = 0.08 to 0.73, three RCTs, n = 166) and long-term functional capacity (SMD = 0.6, 95% CI = 0.33 to 0.88, three RCTs, n = 212). The aforementioned results all achieved clinical significance. The observed low heterogeneity for short-term pain, along with short term and long-term functional capacity, highlights the sustained robustness and consistency of the effect on functional capacity over time. These results suggest that PEMF therapy is statistically effective in enhancing short-term pain relief and improving both short-term and long-term functional capacity in patients with shoulder impingement syndrome, with clinically significant benefits. However, the study limitations include a small sample size and variability in PEMF protocols, highlighting the necessity for standardized methodologies in future research.
Collapse
Affiliation(s)
- Hsun-Yi Wang
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Jen Chen
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - I-Ching Huang
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Gangshan Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chao-Ruei Lin
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ko-Long Lin
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Hsin Chen
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Neurotechanology and Assistive Technology Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
2
|
Ibrahim A, Gupton M, Schroeder F. Regenerative Medicine in Orthopedic Surgery: Expanding Our Toolbox. Cureus 2024; 16:e68487. [PMID: 39364457 PMCID: PMC11447103 DOI: 10.7759/cureus.68487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
Regenerative medicine leverages the body's inherent regenerative capabilities to repair damaged tissues and address organ dysfunction. In orthopedics, this approach includes a variety of treatments collectively known as orthoregeneration, encompassing modalities such as prolotherapy, extracorporeal shockwave therapy, pulsed electromagnetic field therapy, therapeutic ultrasound, and photobiomodulation therapy, and orthobiologics like platelet-rich plasma and cell-based therapies. These minimally invasive techniques are becoming prominent due to their potential for fewer complications in orthopedic surgery. As regenerative medicine continues to advance, surgeons must stay informed about these developments. This paper highlights the current state of regenerative medicine in orthopedics and advocates for further clinical research to validate and expand these treatments to enhance patient outcomes.
Collapse
Affiliation(s)
- Ayah Ibrahim
- Orthopedic Surgery, Burrell College of Osteopathic Medicine, Las Cruces, USA
| | - Marco Gupton
- Orthopedic Surgery, Mountainview Regional Medical Center, Las Cruces, USA
| | - Frederick Schroeder
- Orthopedic Surgery, Burrell College of Osteopathic Medicine, Las Cruces, USA
| |
Collapse
|
3
|
Iwasaki N, Roldo M, Karali A, Blunn G. In vitro development of a muscle-tendon junction construct using decellularised extracellular matrix: Effect of cyclic tensile loading. BIOMATERIALS ADVANCES 2024; 161:213873. [PMID: 38692180 DOI: 10.1016/j.bioadv.2024.213873] [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: 01/03/2024] [Revised: 04/10/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
The muscle tendon junction (MTJ) plays a crucial role in transmitting the force generated by muscles to the tendon and then to the bone. Injuries such as tears and strains frequently happen at the MTJ, where the regenerative process is limited due to poor vascularization and the complex structure of the tissue. Current solutions for a complete tear at the MTJ have not been successful and therefore, the development of a tissue-engineered MTJ may provide a more effective treatment. In this study, decellularised extracellular matrix (DECM) derived from sheep MTJ was used to provide a scaffold for the MTJ with the relevant mechanical properties and differentiation cues such as the relase of growth factors. Human mesenchymal stem cells (MSCs) were seeded on DECM and 10 % cyclic strain was applied using a bioreactor. MSCs cultured on DECM showed significantly higher gene and protein expression of MTJ markers such as collagen 22, paxillin and talin, than MSCs in 2D culture. Although collagen 22 protein expression was higher in the cells with strain than without strain, reduced gene expression of other MTJ markers was observed when the strain was applied. DECM combined with 10 % strain enhanced myogenic differentiation, while tenogenic differentiation was reduced when compared to static cultures of MSCs on DECM. For the first time, these results showed that DECM derived from the MTJ can induce MTJ marker gene and protein expression by MSCs, however, the effect of strain on the MTJ development in DECM culture needs further investigation.
Collapse
Affiliation(s)
- Nodoka Iwasaki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK.
| | - Marta Roldo
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Aikaterina Karali
- School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth, UK
| | - Gordon Blunn
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| |
Collapse
|
4
|
Su DB, Zhao ZX, Yin DC, Ye YJ. Promising application of pulsed electromagnetic fields on tissue repair and regeneration. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 187:36-50. [PMID: 38280492 DOI: 10.1016/j.pbiomolbio.2024.01.003] [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: 07/11/2023] [Revised: 12/14/2023] [Accepted: 01/19/2024] [Indexed: 01/29/2024]
Abstract
Tissue repair and regeneration is a vital biological process in organisms, which is influenced by various internal mechanisms and microenvironments. Pulsed electromagnetic fields (PEMFs) are becoming a potential medical technology due to its advantages of effectiveness and non-invasiveness. Numerous studies have demonstrated that PEMFs can stimulate stem cell proliferation and differentiation, regulate inflammatory reactions, accelerate wound healing, which is of great significance for tissue regeneration and repair, providing a solid basis for enlarging its clinical application. However, some important issues such as optimal parameter system and potential deep mechanisms remain to be resolved due to PEMFs window effect and biological complexity. Thus, it is of great importance to comprehensively summarizing and analyzing the literature related to the biological effects of PEMFs in tissue regeneration and repair. This review expounded the biological effects of PEMFs on stem cells, inflammation response, wound healing and musculoskeletal disorders in order to improve the application value of PEMFs in medicine. It is believed that with the continuous exploration of biological effects of PEMFs, it will be applied increasingly widely to tissue repair and other diseases.
Collapse
Affiliation(s)
- Dan-Bo Su
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Zi-Xu Zhao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Da-Chuan Yin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ya-Jing Ye
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
5
|
Chen C, Zhou H, Yin Y, Hu H, Jiang B, Zhang K, Wu S, Shen M, Wang Z. Rotator cuff muscle degeneration in a mouse model of glenohumeral osteoarthritis induced by monoiodoacetic acid. J Shoulder Elbow Surg 2023; 32:500-511. [PMID: 36442828 DOI: 10.1016/j.jse.2022.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/06/2022] [Accepted: 10/20/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Osteoarthritis (OA) is a disease of joint degeneration and impaired function. Muscle atrophy, fatty infiltration, and fibrosis are degenerative features of muscle injury and predict poor outcomes in some degenerative and exercise-related injuries. Patients with glenohumeral joint OA usually have rotator cuff muscle degeneration, even though the rotator cuff is intact. However, the mechanism and correlation between OA and degeneration of muscles around joints are still unknown. METHODS Forty-five 12-month-old C57BL/6J mice received a single injection of monoiodoacetic acid into the right glenohumeral joint. The sham group was injected with saline on the same day in the right glenohumeral joint. Three and 6 weeks after the operation, gait analysis was conducted to evaluate the function of the forelimb. Then, the shoulder joint and supraspinatus muscle were collected for histologic staining, reverse transcription quantitative polymerase chain reaction, and biomechanics test. Correlations between fat area fraction in muscle, percentage wet muscle weight change or Osteoarthritis Research Society International score, and gait analysis/muscle mechanics tests were assessed using Pearson's correlation coefficient or Spearman's correlation coefficient. RESULTS Compared with the sham group, the monoiodoacetic acid group developed significant glenohumeral joint OA and the supraspinatus muscle developed significant fatty infiltration and muscle atrophy. Shoulder function correlated with glenohumeral joint OA/rotator cuff muscle severity, weight loss, and fatty infiltration. CONCLUSION In mice, glenohumeral joint OA can lead to rotator cuff degeneration and inferior limb function. The small animal model could be a powerful tool to further study the potential mechanisms between glenohumeral OA and rotator cuff muscle degeneration.
Collapse
Affiliation(s)
- Chuanshun Chen
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Clinical Medicine Eight-Year Program, Xiangya Medical School of Central South University, Changsha, Hunan, China
| | - Hecheng Zhou
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Clinical Medicine Eight-Year Program, Xiangya Medical School of Central South University, Changsha, Hunan, China
| | - Yuesong Yin
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Clinical Medicine Eight-Year Program, Xiangya Medical School of Central South University, Changsha, Hunan, China
| | - Hai Hu
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Binbin Jiang
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Kexiang Zhang
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Song Wu
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Minren Shen
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Zili Wang
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
6
|
Cicek F, Tastekin B, Baldan I, Tokus M, Pelit A, Ocal I, Gunay I, Ogur HU, Cicek H. Effect of 40 Hz Magnetic Field Application in Posttraumatic Muscular Atrophy Development on Muscle Mass and Contractions in Rats. Bioelectromagnetics 2022; 43:453-461. [PMID: 36477897 DOI: 10.1002/bem.22429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/06/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
Muscle atrophy refers to the deterioration of muscle tissue due to a long-term decrease in muscle function. In the present study, we simulated rectus femoris muscle atrophy experimentally and investigated the effect of pulsed electromagnetic field (PEMF) application on the atrophy development through muscle mass, maximal contraction force, and contraction-relaxation time. A quadriceps tendon rupture with a total tenotomy was created on the rats' hind limbs, inhibiting knee extension for 6 weeks, and this restriction of the movement led to the development of disuse atrophy, while the control group underwent no surgery. The operated and control groups were divided into subgroups according to PEMF application (1.5 mT for 45 days) or no PEMF. All groups were sacrificed after 6 weeks and had their entire rectus femoris removed. To measure the contraction force, the muscles were placed in an organ bath connected to a transducer. As a result of the atrophy, muscle mass and strength were reduced in the operated group, while no muscle mass loss was observed in the operated PEMF group. Furthermore, measurements of single, incomplete and full tetanic contraction force and contraction time (CT) did not change significantly in the operated group that received the PEMF application. The PEMF application prevented atrophy resulting from 6 weeks of immobility, according to the contraction parameters. The effects of PEMF on contraction force and CT provide a basis for further studies in which PEMF is investigated as a noninvasive therapy for disuse atrophy development. © 2022 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Figen Cicek
- Department of Biophysics, Cukurova University, Adana, Turkey
| | - Bora Tastekin
- Department of Biophysics, Cukurova University, Adana, Turkey
| | - Ilknur Baldan
- Department of Biophysics, Cukurova University, Adana, Turkey
| | - Murat Tokus
- Department of Biophysics, Cukurova University, Adana, Turkey
| | - Aykut Pelit
- Department of Biophysics, Cukurova University, Adana, Turkey
| | - Isil Ocal
- Department of Biophysics, Cukurova University, Adana, Turkey
| | - Ismail Gunay
- Department of Biophysics, Cukurova University, Adana, Turkey
| | - Hasan U Ogur
- Adana City Hospital, Orthopedics and Traumatology Clinics, Adana, Turkey
| | - Hakan Cicek
- Adana City Hospital, Orthopedics and Traumatology Clinics, Adana, Turkey
| |
Collapse
|
7
|
Ong MTY, Man GCW, Lau LCM, He X, Qiu J, Wang Q, Chow MCS, Choi BCY, Yu M, Yung PSH. Effect of pulsed electromagnetic field as an intervention for patients with quadriceps weakness after anterior cruciate ligament reconstruction: a double-blinded, randomized-controlled trial. Trials 2022; 23:771. [PMID: 36096886 PMCID: PMC9465849 DOI: 10.1186/s13063-022-06674-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/18/2022] [Indexed: 11/18/2022] Open
Abstract
Background The ultimate goal of anterior cruciate ligament reconstructions (ACLR) is to fulfil the return-to-play (RTP) criteria. Quadriceps muscle strength is one of the key determinants for a patient’s successful return-to-play after ACLR. Quadriceps muscle atrophy can persist beyond the completion of the rehabilitation program in almost half the patients and the reason behind this is still unknown. There are emerging evidences showing that pulsed electromagnetic field (PEMF) can modulate mitochondrial activities for muscle gain. PEMF exposure on top of regular exercise training may promote muscle regeneration and tissue healing. Methods This is a double-blinded, randomized controlled trial to investigate the effects of PEMF treatment during the postoperative period on quadriceps muscle strength in ACL injured patient. Adult patients (aged 18–30) with a unilateral ACL injury, total quadriceps muscle volume is equal or more than 7% deficit on involved leg compared with uninvolved leg, sporting injury with a Tegner score of 7+, and both knees without a history of injury/prior surgery will be recruited. To estimate the improvement of patients, isokinetic muscle assessment, ultrasound imaging and MRI for quadriceps muscle thickness, self-reported outcomes with questionnaires, KT-1000 for knee laxity and biomechanical analysis, and Xtreme CT for bone mineral density will be performed. To investigate the mechanism of PEMF therapy on increasing quadriceps strength, samples of blood serum will be drawn before and after intervention. Discussion This is the first trial evaluating the effects of PEMF on quadriceps muscle recovery after ACLR. The proposed study addresses a huge research gap by evaluating practical use of PEMF as part of rehabilitation. The proposed study will provide much needed scientific support in the use of this noninvasive treatment modality to facilitate recovery of quadriceps strength after PEMF. Trial registration ClinicalTrials.gov NCT05184023. Registered on 5 January 2022 Supplementary Information The online version contains supplementary material available at 10.1186/s13063-022-06674-2.
Collapse
|
8
|
Han F, Yin S, Wu H, Zhou C, Wang X. Effect on myoblast differentiation by extremely low frequency pulsed electromagnetic fields. J MECH MED BIOL 2022. [DOI: 10.1142/s0219519422400267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
9
|
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
|
10
|
Terrie L, Burattini M, Van Vlierberghe S, Fassina L, Thorrez L. Enhancing Myoblast Fusion and Myotube Diameter in Human 3D Skeletal Muscle Constructs by Electromagnetic Stimulation. Front Bioeng Biotechnol 2022; 10:892287. [PMID: 35814025 PMCID: PMC9256958 DOI: 10.3389/fbioe.2022.892287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/06/2022] [Indexed: 11/25/2022] Open
Abstract
Skeletal muscle tissue engineering (SMTE) aims at the in vitro generation of 3D skeletal muscle engineered constructs which mimic the native muscle structure and function. Although native skeletal muscle is a highly dynamic tissue, most research approaches still focus on static cell culture methods, while research on stimulation protocols indicates a positive effect, especially on myogenesis. A more mature muscle construct may be needed especially for the potential applications for regenerative medicine purposes, disease or drug disposition models. Most efforts towards dynamic cell or tissue culture methods have been geared towards mechanical or electrical stimulation or a combination of those. In the context of dynamic methods, pulsed electromagnetic field (PEMF) stimulation has been extensively used in bone tissue engineering, but the impact of PEMF on skeletal muscle development is poorly explored. Here, we evaluated the effects of PEMF stimulation on human skeletal muscle cells both in 2D and 3D experiments. First, PEMF was applied on 2D cultures of human myoblasts during differentiation. In 2D, enhanced myogenesis was observed, as evidenced by an increased myotube diameter and fusion index. Second, 2D results were translated towards 3D bioartificial muscles (BAMs). BAMs were subjected to PEMF for varying exposure times, where a 2-h daily stimulation was found to be effective in enhancing 3D myotube formation. Third, applying this protocol for the entire 16-days culture period was compared to a stimulation starting at day 8, once the myotubes were formed. The latter was found to result in significantly higher myotube diameter, fusion index, and increased myosin heavy chain 1 expression. This work shows the potential of electromagnetic stimulation for enhancing myotube formation both in 2D and 3D, warranting its further consideration in dynamic culturing techniques.
Collapse
Affiliation(s)
- Lisanne Terrie
- Tissue Engineering Lab, Dep. Development and Regeneration, KU Leuven Kulak, Kortrijk, Belgium
| | - Margherita Burattini
- Tissue Engineering Lab, Dep. Development and Regeneration, KU Leuven Kulak, Kortrijk, Belgium
- Dept. of Surgical Sciences, Dentistry and Maternity, University of Verona, Verona, Italy
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Dep. of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Lorenzo Fassina
- Dept. of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Lieven Thorrez
- Tissue Engineering Lab, Dep. Development and Regeneration, KU Leuven Kulak, Kortrijk, Belgium
- *Correspondence: Lieven Thorrez,
| |
Collapse
|
11
|
Bi J, Jing H, Zhou C, Gao P, Han F, Li G, Zhang S. Regulation of skeletal myogenesis in C2C12 cells through modulation of Pax7, MyoD, and myogenin via different low-frequency electromagnetic field energies. Technol Health Care 2022; 30:371-382. [PMID: 35124612 PMCID: PMC9028610 DOI: 10.3233/thc-thc228034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND: A low-frequency electromagnetic field (LF-EMF) exerts important biological effects on the human body. OBJECTIVE: We previously studied the immunity and atrophy of gastrocnemius muscles in rats with spinal cord injuries and found that LF-EMF with a magnetic flux density of 1.5 mT exerted excellent therapeutic and preventive effects on reducing myotubes and increasing spatium intermusculare. However, the effects of LF-EMF on all stages of skeletal myogenesis, such as activation, proliferation, differentiation, and fusion of satellite cells to myotubes as stimulated by myogenic regulatoryfactors (MRFs), have not been fully elucidated. METHODS: This study investigated the optimal LF-EMF magnetic flux density that exerted maximal effects on all stages of C2C12 cell skeletal myogenesis as well as its impact on regulatory MRFs. RESULTS: The results showed that an LF-EMF with a magnetic flux density of 2.0 mT could activate C2C12 cells and upregulate the proliferation-promoting transcription factor PAX7. On the other hand, 1.5 mT EMF could upregulate the expression of MyoD and myogenin. CONCLUSION: LF-EMF could prevent the disappearance of myotubes, with different magnetic flux densities of LF-EMF exerting independent and positive effects on skeletal myogenesis such as satellite cell activation and proliferation, muscle cell differentiation, and myocyte fusion.
Collapse
Affiliation(s)
- Jiaqi Bi
- Harbin Children’s Hospital, Harbin, Heilongjiang, China
- Emergency Department, SongBei Hospital of The Fourth Hospital Affiliated of Harbin Medical University, Harbin, Heilongjiang, China
- Harbin Children’s Hospital, Harbin, Heilongjiang, China
| | - Hong Jing
- Harbin Children’s Hospital, Harbin, Heilongjiang, China
- Harbin Children’s Hospital, Harbin, Heilongjiang, China
| | - ChenLiang Zhou
- Emergency Department, SongBei Hospital of The Fourth Hospital Affiliated of Harbin Medical University, Harbin, Heilongjiang, China
| | - Peng Gao
- The First Department of General Surgery, Harbin Children’s Hospital, Harbin, Heilongjiang, China
| | - Fujun Han
- Emergency Department, SongBei Hospital of The Fourth Hospital Affiliated of Harbin Medical University, Harbin, Heilongjiang, China
| | - Gang Li
- The Second Department of Orthopedics, The First Hospital of Yichun, Yichun, Heilongjiang, China
| | - Shiwei Zhang
- Harbin Children’s Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
12
|
BI JIAQI, JING HONG, ZHOU CHENLIANG, GAO PENG, HAN FUJUN, LI GANG, SHI DONGFANG. EFFECT OF LOW-FREQUENCY ELECTROMAGNETICS (LFE) ON MUSCLE SATELLITE CELLS DIFFERENTIATION AND IMMUNE SYSTEM IN RAT. J MECH MED BIOL 2021. [DOI: 10.1142/s0219519421400546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Spinal cord injury (SCI) is a severe neurological disease. Although surgery within 8[Formula: see text]h after SCI can substantially reduce paraplegia, most patients still suffer from hypomusculariasis after neuron recovery, which results in insufficient lower limb muscles to support bodyweight. Currently, there is no effective method to prevent muscle atrophy. Previous studies have shown that low-frequency electromagnetics (LFE) can stimulate the differentiation, proliferation and fusion of muscle satellite cells, however, the optimal electromagnetic strength and effects on the immune system have not been established. Here, we investigated the influence of LFE at different electromagnetic strengths on muscle cell recovery and assessed the impact of chronic LFE on the immune system of SCI rats. The rat immune system was rapidly activated after SCI. High-energy LFE provoked intensive immune responses, while low-energy LFE did not affect immune responses. Simultaneously, LFE effectively prevented myotube reduction and atrophy in SCI rats. The mRNA and protein levels of Pax7 and MyoD were increased after LFE at both high and low electromagnetic strengths, with the latter leading to more robust increases. Indeed, LFE remarkably induced muscle cell fusion. Together, our results demonstrated that LFE activates muscle satellite cells via stimulating myogenic factors. Chronic low-energy LFE is a safe therapy with no adverse impact on the immune system of SCI rats. LFE with 1.5 mT energy should be considered as an optimal therapeutic strategy.
Collapse
Affiliation(s)
- JIAQI BI
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, Heilongjiang, P. R. China
- Harbin Children’s Hospital, Harbin 150010, Heilongjiang, P. R. China
| | - HONG JING
- Harbin Children’s Hospital, Harbin 150010, Heilongjiang, P. R. China
| | - CHENLIANG ZHOU
- Emergency Department, SongBei Hospital of The Fourth Hospital, Affiliated of Harbin Medical University, Harbin 150021, Heilongjiang, P. R. China
| | - PENG GAO
- The First Department of General Surgery, Harbin Children’s Hospital, Harbin 150010, Heilongjiang, P. R. China
| | - FUJUN HAN
- Emergency Department, SongBei Hospital of The Fourth Hospital, Affiliated of Harbin Medical University, Harbin 150021, Heilongjiang, P. R. China
| | - GANG LI
- The Second Department of Orthopedics, The First Hospital of Yichun Yichun, 153000, Heilongjiang, P. R. China
| | - DONGFANG SHI
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, Heilongjiang, P. R. China
| |
Collapse
|
13
|
DNMT1 and miRNAs: possible epigenetics footprints in electromagnetic fields utilization in oncology. Med Oncol 2021; 38:125. [PMID: 34495398 DOI: 10.1007/s12032-021-01574-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/01/2021] [Indexed: 10/20/2022]
Abstract
Many studies were performed to unravel the effects of different types of Electromagnetic fields (EMFs) on biological systems. Some studies were conducted to exploit EMFs for medical purposes mainly in cancer therapy. Although many studies suggest that the EMFs exposures can be effective in pre-clinical cancer issues, the treatment outcomes of these exposures on the cancer cells, especially at the molecular level, are challenging and overwhelmingly complicated yet. This article aims to review the epigenetic mechanisms that can be altered by EMFs exposures with the main emphasis on Extremely low frequency electromagnetic field (ELF-EMF). The epigenetic mechanisms are reversible and affected by environmental factors, thus, EMFs exposures can modulate these mechanisms. According to the reports, ELF-EMF exposures affect epigenetic machinery directly or through the molecular signaling pathways. ELF-EMF in association with DNA methylation, histone modification, miRNAs, and nucleosome remodeling could affect the homeostasis of cancer cells and play a role in DNA damage repairing, apoptosis induction, prevention of metastasis, differentiation, and cell cycle regulation. In general, the result of this study shows that ELF-EMF exposure probably can be effective in cancer epigenetic therapy, but more molecular and clinical investigations are needed to clarify the safe and specific dosimetric characteristics of ELF-EMF in practice.
Collapse
|
14
|
Mu JK, Zi L, Li YQ, Yu LP, Cui ZG, Shi TT, Zhang F, Gu W, Hao JJ, Yu J, Yang XX. Jiuzhuan Huangjing Pills relieve mitochondrial dysfunction and attenuate high-fat diet-induced metabolic dysfunction-associated fatty liver disease. Biomed Pharmacother 2021; 142:112092. [PMID: 34449316 DOI: 10.1016/j.biopha.2021.112092] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/14/2021] [Accepted: 08/19/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD) is a common global chronic liver disease. Jiuzhuan Huangjing Pills (JHP) have been used for the treatment of human disease for over a thousand years, but their efficacy and underlying mechanism(s) of action against MAFLD are unknown. We investigated the alleviating effects of JHP on high-fat diet (HFD)-induced MAFLD. METHODS In vitro and in vivo methods were used to evaluate the effects of JHP on MAFLD. L02 adipocyte models were induced by fat emulsion and adipocytes were treated with JHP for 24 h. MAFLD rat models were induced by HFD-feeding and were intragastrically administered JHP for 12 weeks. Changes in fat accumulation, L02 cell damage, body weight, food intake, histological parameters, organ indexes, biochemical parameters, and mitochondrial indicators including ultrastructure, oxidative stress, energy metabolism, and fatty acid metabolism were investigated. RESULTS JHP attenuated the increase in levels of total cholesterol, triglyceride, low density lipoprotein cholesterol, alanine transaminase, and aspartate transaminase levels, and significantly increased high density lipoprotein cholesterol. JHP up-regulated levels of glutathione (GSH) and superoxide dismutase (SOD), and down-regulated malondialdehyde (MDA). JHP afforded protection to the mitochondrial ultrastructure, and inhibited the HFD-induced increase in MDA and the reduction of SOD, GSH, ATP synthase, and complex I and II, in liver mitochondria. JHP regulated the expression of β-oxidation genes, including acyl-CoA dehydrogenase, cyl-CoA dehydrogenase long chain, carnitine palmitoyltransferase 1A, carnitine palmitoyltransferase 1B, peroxisomal proliferator-activated receptor-gamma coactivator-1α and peroxide proliferator activated receptor α. CONCLUSION JHP alleviates HFD-induced MAFLD through the protection of mitochondrial function.
Collapse
Affiliation(s)
- Jian-Kang Mu
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Southern Medicine Utilization, 1076 Yuhua Road, Kunming 650500, China
| | - Lei Zi
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Southern Medicine Utilization, 1076 Yuhua Road, Kunming 650500, China
| | - Yan-Qin Li
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Southern Medicine Utilization, 1076 Yuhua Road, Kunming 650500, China
| | - Li-Ping Yu
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Southern Medicine Utilization, 1076 Yuhua Road, Kunming 650500, China
| | - Zheng-Guo Cui
- Department of Environmental Health, University of Fukui School of Medical Science, 23-3 Matsuoka Shimoaizuki, Eiheiji 910-1193, Japan
| | - Ting-Ting Shi
- Department of Pharmaceutical Preparation, The Xixi Hospital of Hangzhou Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou 310023, China
| | - Fan Zhang
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Southern Medicine Utilization, 1076 Yuhua Road, Kunming 650500, China
| | - Wen Gu
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Southern Medicine Utilization, 1076 Yuhua Road, Kunming 650500, China
| | - Jun-Jie Hao
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Southern Medicine Utilization, 1076 Yuhua Road, Kunming 650500, China
| | - Jie Yu
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Southern Medicine Utilization, 1076 Yuhua Road, Kunming 650500, China.
| | - Xing-Xin Yang
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Southern Medicine Utilization, 1076 Yuhua Road, Kunming 650500, China.
| |
Collapse
|
15
|
Liu X, Liu M, Lee L, Davies M, Wang Z, Kim H, Feeley BT. Trichostatin A regulates fibro/adipogenic progenitor adipogenesis epigenetically and reduces rotator cuff muscle fatty infiltration. J Orthop Res 2021; 39:1452-1462. [PMID: 32970360 PMCID: PMC7987912 DOI: 10.1002/jor.24865] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 02/04/2023]
Abstract
Rotator cuff (RC) muscle fatty infiltration (FI) is an important factor that determines the clinical outcome of patients with RC repair. There is no effective treatment for RC muscle FI at this time. The goal of this study is to define the role Trichostatin A (TSA), a histone deacetylase (HDAC) inhibitor in regulating muscle fibro/adipogenic progenitors (FAPs) adipogenesis and treating muscle fatty degeneration after massive RC tears in a mouse model. We hypothesize that TSA reduces muscle FI after massive RC tears. HDAC activity was measured in FAPs in RC muscle after tendon/nerve transection or sham surgery. FAPs were treated with TSA for 2 weeks and FAP adipogenesis was evaluated with perilipin and Oil Red O staining, as well as reverse transcript-polymerase chain reaction for adipogenesis-related genes. About 0.5 mg/kg TSA or dimethyl sulfoxide was administered to C57B/L6 mice with massive rotator cuff tears through daily intraperitoneal injection for 6 weeks. Supraspinatus muscles were harvested for biochemical and histology analysis. We found that FAPs showed significantly higher HDAC activity after RC tendon/nerve transection. TSA treatment significantly reduced HDAC activity and inhibited adipogenesis of FAPs. TSA also abolished the role of bone morphogenetic protein-7 in inducing FAP adipogenesis and promoted FAP brown/beige adipose tissue (BAT) differentiation. TSA injection significantly increased histone H3 acetylation and reduced FI of rotator cuff muscles after massive tendon tears. Results from this study showed that TSA can regulate FAP adipogenesis and promote FAP BAT differentiation epigenetically. HDAC inhibition may be a new treatment strategy to reduce muscle FI after RC tears and repair.
Collapse
Affiliation(s)
- Xuhui Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
- Department of Orthopaedic Surgery, University of California, San Francisco, California, USA
| | - Mengyao Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
- Department of Orthopaedic Surgery, University of California, San Francisco, California, USA
| | - Lawrence Lee
- Department of Veterans Affairs, San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
| | - Michael Davies
- Department of Orthopaedic Surgery, University of California, San Francisco, California, USA
| | - Zili Wang
- Department of Veterans Affairs, San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
- Department of Orthopaedic Surgery, University of California, San Francisco, California, USA
- Department of Orthopaedic Surgery, Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Hubert Kim
- Department of Veterans Affairs, San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
- Department of Orthopaedic Surgery, University of California, San Francisco, California, USA
| | - Brian T. Feeley
- Department of Veterans Affairs, San Francisco Veterans Affairs Health Care System, San Francisco, California, USA
- Department of Orthopaedic Surgery, University of California, San Francisco, California, USA
| |
Collapse
|
16
|
Dolkart O, Kazum E, Rosenthal Y, Sher O, Morag G, Yakobson E, Chechik O, Maman E. Effects of focused continuous pulsed electromagnetic field therapy on early tendon-to-bone healing. Bone Joint Res 2021; 10:298-306. [PMID: 33934605 PMCID: PMC8160030 DOI: 10.1302/2046-3758.105.bjr-2020-0253.r2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aims Rotator cuff (RC) tears are common musculoskeletal injuries which often require surgical intervention. Noninvasive pulsed electromagnetic field (PEMF) devices have been approved for treatment of long-bone fracture nonunions and as an adjunct to lumbar and cervical spine fusion surgery. This study aimed to assess the effect of continuous PEMF on postoperative RC healing in a rat RC repair model. Methods A total of 30 Wistar rats underwent acute bilateral supraspinatus tear and repair. A miniaturized electromagnetic device (MED) was implanted at the right shoulder and generated focused PEMF therapy. The animals’ left shoulders served as controls. Biomechanical, histological, and bone properties were assessed at three and six weeks. Results Extension of the tendon from preload to the maximum load to failure was significantly better in the PEMF-treated shoulders at three weeks compared to controls (p = 0.038). The percentage strain was significantly higher in the PEMF group at both timepoints (p = 0.037). Collagen organization was significantly better (p = 0.034) as was tissue mineral density in the PEMF-treated group at three weeks (p = 0.028). Tendon immunohistochemistry revealed a prominent increase in type I collagen at the repair site at three weeks following continuous PEMF treatment compared with controls. None of the other tested parameters differed between the groups. Conclusion MED-generated PEMF may enhance early postoperative tendon-to-bone healing in an acute rat supraspinatus detachment and repair model. Superior biomechanical elasticity parameters together with better collagen organization suggest improved RC healing. Cite this article: Bone Joint Res 2021;10(5):298–306.
Collapse
Affiliation(s)
- Oleg Dolkart
- Shoulder Unit, Division of Orthopaedic Surgery, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Efi Kazum
- Shoulder Unit, Division of Orthopaedic Surgery, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yoav Rosenthal
- Shoulder Unit, Division of Orthopaedic Surgery, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Sher
- Pathology Institute, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Guy Morag
- Shoulder Unit, Division of Orthopaedic Surgery, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Ofir Chechik
- Shoulder Unit, Division of Orthopaedic Surgery, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eran Maman
- Shoulder Unit, Division of Orthopaedic Surgery, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
17
|
Vinhas A, Rodrigues MT, Gonçalves AI, Reis RL, Gomes ME. Magnetic responsive materials modulate the inflammatory profile of IL-1β conditioned tendon cells. Acta Biomater 2020; 117:235-245. [PMID: 32966921 DOI: 10.1016/j.actbio.2020.09.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/10/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022]
Abstract
Tendinopathies represent half of all musculoskeletal injuries worldwide. Inflammatory events contribute to both tendon healing and to tendinopathy conditions but the cellular triggers leading to one or the other are unknown. In previous studies, we showed that magnetic field actuation modulates human tendon cells (hTDCs) behavior in pro-inflammatory environments, and that magnetic responsive membranes could positively influence inflammation responses in a rat ectopic model. Herein, we propose to investigate the potential synergistic action of the magnetic responsive membranes, made of a polymer blend of starch with polycaprolactone incorporating magnetic nanoparticles (magSPCL), and the actuation of pulsed electromagnetic field (PEMF): 5 Hz, 4mT of intensity and 50% of duty cycle, in IL-1β-treated-hTDCs, and in the immunomodulatory response of macrophages. It was found that the expression of pro-inflammatory (TNFα, IL-6, IL-8, COX-2) and ECM remodeling (MMP-1,-2,-3) markers tend to decrease in cells cultured onto magSPCL membranes under PEMF, while the expression of TIMP-1 and anti-inflammatory genes (IL-4, IL-10) increases. Also, CD16++ and CD206+ macrophages were only found on magSPCL membranes with PEMF application. Magnetic responsive membranes show a modulatory effect on the inflammatory profile of hTDCs favoring anti-inflammatory cues which is also supported by the anti-inflammatory/repair markers expressed in macrophages. These results suggest that magnetic responsive magSPCL membranes can contribute for inflammation resolution acting on both resident cell populations and inflammatory cells, and thus significantly contribute to tendon regenerative strategies. Statement of significance Magnetically-assisted strategies have received great attention in recent years to remotely trigger and guide cell responses. Inflammation plays a key role in tendon healing but persistent pro-inflammatory molecules can contribute to tendon disorders, and therefore provide a therapeutic target for advanced treatments. We have previously reported that magnetic fields modulate the response of human tendon cells (hTDCs) conditioned to pro-inflammatory environments (IL-1β-treated-hTDCs), and that magnetic responsive membranes positively influence immune responses. In the present work, we combined pulsed electromagnetic field (PEMF) and magnetic responsive membranes to guide the inflammatory profile of IL-1β-treated-hTDCs and of macrophages. The results showed that the synergistic action of PEMF and magnetic membranes supports the applicability of magnetically actuated systems to regulate inflammatory events and stimulate tendon regeneration.
Collapse
Affiliation(s)
- A Vinhas
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - M T Rodrigues
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - A I Gonçalves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - R L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - M E Gomes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
18
|
Perucca Orfei C, Lovati AB, Lugano G, Viganò M, Bottagisio M, D'Arrigo D, Sansone V, Setti S, de Girolamo L. Pulsed electromagnetic fields improve the healing process of Achilles tendinopathy: a pilot study in a rat model. Bone Joint Res 2020; 9:613-622. [PMID: 33072305 PMCID: PMC7533373 DOI: 10.1302/2046-3758.99.bjr-2020-0113.r1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aims In the context of tendon degenerative disorders, the need for innovative conservative treatments that can improve the intrinsic healing potential of tendon tissue is progressively increasing. In this study, the role of pulsed electromagnetic fields (PEMFs) in improving the tendon healing process was evaluated in a rat model of collagenase-induced Achilles tendinopathy. Methods A total of 68 Sprague Dawley rats received a single injection of type I collagenase in Achilles tendons to induce the tendinopathy and then were daily exposed to PEMFs (1.5 mT and 75 Hz) for up to 14 days - starting 1, 7, or 15 days after the injection - to identify the best treatment option with respect to the phase of the disease. Then, 7 and 14 days of PEMF exposure were compared to identify the most effective protocol. Results The daily exposure to PEMFs generally provided an improvement in the fibre organization, a decrease in cell density, vascularity, and fat deposition, and a restoration of the physiological cell morphology compared to untreated tendons. These improvements were more evident when the tendons were exposed to PEMFs during the mid-acute phase of the pathology (7 days after induction) rather than during the early (1 day after induction) or the late acute phase (15 days after induction). Moreover, the exposure to PEMFs for 14 days during the mid-acute phase was more effective than for 7 days. Conclusion PEMFs exerted a positive role in the tendon healing process, thus representing a promising conservative treatment for tendinopathy, although further investigations regarding the clinical evaluation are needed. Cite this article: Bone Joint Res 2020;9(9):613–622.
Collapse
Affiliation(s)
| | | | - Gaia Lugano
- IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | - Marco Viganò
- IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | | | - Daniele D'Arrigo
- IRCCS Orthopedic Institute Galeazzi, Milan, Italy.,Regenerative Medicine Technologies Laboratory, Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
| | - Valerio Sansone
- IRCCS Orthopedic Institute Galeazzi, Milan, Italy.,Faculty of Medicine and Surgery, University of Milan, Milan, Italy
| | | | | |
Collapse
|
19
|
Colombini A, Perucca Orfei C, Vincenzi F, De Luca P, Ragni E, Viganò M, Setti S, Varani K, de Girolamo L. A2A adenosine receptors are involved in the reparative response of tendon cells to pulsed electromagnetic fields. PLoS One 2020; 15:e0239807. [PMID: 32998161 PMCID: PMC7527253 DOI: 10.1371/journal.pone.0239807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/14/2020] [Indexed: 11/19/2022] Open
Abstract
Tendinopathy is a degenerative disease in which inflammatory mediators have been found to be sometimes present. The interaction between inflammation and matrix remodeling in human tendon cells (TCs) is supported by the secretion of cytokines such as IL-1β, IL-6 and IL-33. In this context, it has been demonstrated that pulsed electromagnetic fields (PEMFs) were able to reduce inflammation and promote tendon marker synthesis. The aim of this study was to evaluate the anabolic and anti-inflammatory PEMF-mediated response on TCs in an in vitro model of inflammation. Moreover, since PEMFs enhance the anti-inflammatory efficacy of adenosine through the adenosine receptors (ARs), the study also focused on the role of A2AARs. Human TCs were exposed to PEMFs for 48 hours. After stimulation, A2AAR saturation binding experiments were performed. Along with 48 hours PEMF stimulation, TCs were treated with IL-1β and A2AAR agonist CGS-21680. IL-1Ra, IL-6, IL-8, IL-10, IL-33, VEGF, TGF-β1, PGE2 release and SCX, COL1A1, COL3A1, ADORA2A expression were quantified. PEMFs exerted A2AAR modulation on TCs and promoted COL3A1 upregulation and IL-33 secretion. In presence of IL-1β, TCs showed an upregulation of ADORA2A, SCX and COL3A1 expression and an increase of IL-6, IL-8, PGE2 and VEGF secretion. After PEMF and IL-1β exposure, IL-33 was upregulated, whereas IL-6, PGE2 and ADORA2A were downregulated. These findings demonstrated that A2AARs have a role in the promotion of the TC anabolic/reparative response to PEMFs and to IL-1β.
Collapse
Affiliation(s)
- Alessandra Colombini
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- * E-mail:
| | | | - Fabrizio Vincenzi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Paola De Luca
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Enrico Ragni
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Marco Viganò
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | | | - Katia Varani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Laura de Girolamo
- Orthopaedic Biotechnology Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|
20
|
Hu H, Yang W, Zeng Q, Chen W, Zhu Y, Liu W, Wang S, Wang B, Shao Z, Zhang Y. Promising application of Pulsed Electromagnetic Fields (PEMFs) in musculoskeletal disorders. Biomed Pharmacother 2020; 131:110767. [PMID: 33152929 DOI: 10.1016/j.biopha.2020.110767] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/06/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence suggests that an exogenous electromagnetic field might be involved in many biologic processes which are of great importance for therapeutic interventions. Pulsed electromagnetic fields (PEMFs) are known to be a noninvasive, safe and effective therapy agent without apparent side effects. Numerous studies have shown that PEMFs possess the potential to become a stand-alone or adjunctive treatment modality for treating musculoskeletal disorders. However, several issues remain unresolved. Prior to their widely clinical application, further researches from well-designed, high-quality studies are still required to standardize the treatment parameters and derive the optimal protocol for health-care decision making. In this review, we aim to provide current evidence on the mechanism of action, clinical applications, and controversies of PEMFs in musculoskeletal disorders.
Collapse
Affiliation(s)
- Hongzhi Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China.
| | - Wenbo Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Qianwen Zeng
- Department of Pediatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Wei Chen
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - YanBin Zhu
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Weijian Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shangyu Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Baichuan Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yingze Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China.
| |
Collapse
|
21
|
Citeroni MR, Ciardulli MC, Russo V, Della Porta G, Mauro A, El Khatib M, Di Mattia M, Galesso D, Barbera C, Forsyth NR, Maffulli N, Barboni B. In Vitro Innovation of Tendon Tissue Engineering Strategies. Int J Mol Sci 2020; 21:E6726. [PMID: 32937830 PMCID: PMC7555358 DOI: 10.3390/ijms21186726] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Tendinopathy is the term used to refer to tendon disorders. Spontaneous adult tendon healing results in scar tissue formation and fibrosis with suboptimal biomechanical properties, often resulting in poor and painful mobility. The biomechanical properties of the tissue are negatively affected. Adult tendons have a limited natural healing capacity, and often respond poorly to current treatments that frequently are focused on exercise, drug delivery, and surgical procedures. Therefore, it is of great importance to identify key molecular and cellular processes involved in the progression of tendinopathies to develop effective therapeutic strategies and drive the tissue toward regeneration. To treat tendon diseases and support tendon regeneration, cell-based therapy as well as tissue engineering approaches are considered options, though none can yet be considered conclusive in their reproduction of a safe and successful long-term solution for full microarchitecture and biomechanical tissue recovery. In vitro differentiation techniques are not yet fully validated. This review aims to compare different available tendon in vitro differentiation strategies to clarify the state of art regarding the differentiation process.
Collapse
Affiliation(s)
- Maria Rita Citeroni
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Maria Camilla Ciardulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (G.D.P.); (N.M.)
| | - Valentina Russo
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (G.D.P.); (N.M.)
- Interdepartment Centre BIONAM, Università di Salerno, via Giovanni Paolo I, 84084 Fisciano (SA), Italy
| | - Annunziata Mauro
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Mohammad El Khatib
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Miriam Di Mattia
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| | - Devis Galesso
- Fidia Farmaceutici S.p.A., via Ponte della Fabbrica 3/A, 35031 Abano Terme (PD), Italy; (D.G.); (C.B.)
| | - Carlo Barbera
- Fidia Farmaceutici S.p.A., via Ponte della Fabbrica 3/A, 35031 Abano Terme (PD), Italy; (D.G.); (C.B.)
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Thornburrow Drive, Stoke on Trent ST4 7QB, UK;
| | - Nicola Maffulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi (SA), Italy; (M.C.C.); (G.D.P.); (N.M.)
- Department of Musculoskeletal Disorders, Faculty of Medicine and Surgery, University of Salerno, Via San Leonardo 1, 84131 Salerno, Italy
- Centre for Sports and Exercise Medicine, Barts and The London School of Medicine and Dentistry, Mile End Hospital, Queen Mary University of London, 275 Bancroft Road, London E1 4DG, UK
- School of Pharmacy and Bioengineering, Keele University School of Medicine, Thornburrow Drive, Stoke on Trent ST5 5BG, UK
| | - Barbara Barboni
- Unit of Basic and Applied Biosciences, Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, 64100 Teramo, Italy; (V.R.); (A.M.); (M.E.K.); (M.D.M.); (B.B.)
| |
Collapse
|
22
|
Matos AM, Gonçalves AI, El Haj AJ, Gomes ME. Magnetic biomaterials and nano-instructive tools as mediators of tendon mechanotransduction. NANOSCALE ADVANCES 2020; 2:140-148. [PMID: 36133967 PMCID: PMC9417540 DOI: 10.1039/c9na00615j] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/29/2019] [Indexed: 05/29/2023]
Abstract
Tendon tissues connect muscle to bone allowing the transmission of forces resulting in joint movement. Tendon injuries are prevalent in society and the impact on public health is of utmost concern. Thus, clinical options for tendon treatments are in demand, and tissue engineering aims to provide reliable and successful long-term regenerative solutions. Moreover, the possibility of regulating cell fate by triggering intracellular pathways is a current challenge in regenerative medicine. In the last decade, the use of magnetic nanoparticles as nano-instructive tools has led to great advances in diagnostics and therapeutics. Recent advances using magnetic nanomaterials for regenerative medicine applications include the incorporation of magnetic biomaterials within 3D scaffolds resulting in mechanoresponsive systems with unprecedented properties and the use of nanomagnetic actuators to control cell signaling. Mechano-responsive scaffolds and nanomagnetic systems can act as mechanostimulation platforms to apply forces directly to single cells and multicellular biological tissues. As transmitters of forces in a localized manner, the approaches enable the downstream activation of key tenogenic signaling pathways. In this minireview, we provide a brief outlook on the tenogenic signaling pathways which are most associated with the conversion of mechanical input into biochemical signals, the novel bio-magnetic approaches which can activate these pathways, and the efforts to translate magnetic biomaterials into regenerative platforms for tendon repair.
Collapse
Affiliation(s)
- Ana M Matos
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Ana I Gonçalves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Alicia J El Haj
- Healthcare Technologies Institute, Birmingham University B15 2TT Birmingham UK
| | - Manuela E Gomes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at the University of Minho Avepark, 4805-017 Barco Guimarães Portugal
| |
Collapse
|
23
|
Vinhas A, Rodrigues MT, Gonçalves AI, Reis RL, Gomes ME. Pulsed Electromagnetic Field Modulates Tendon Cells Response in IL-1β-Conditioned Environment. J Orthop Res 2020; 38:160-172. [PMID: 31769535 DOI: 10.1002/jor.24538] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 11/19/2019] [Indexed: 02/04/2023]
Abstract
Strategies aiming at controlling and modulating inflammatory cues may offer therapeutic solutions for improving tendon regeneration. This study aims to investigate the modulatory effect of pulsed electromagnetic field (PEMF) on the inflammatory profile of human tendon-derived cells (hTDCs) after supplementation with interleukin-1β (IL-1β). IL-1β was used to artificially induce inflammatory cues associated with injured tendon environments. The PEMF effect was investigated varying the frequency (5 or 17 Hz), intensity (1.5, 4, or 5 mT), and duty-cycle (10% or 50%) parameters to which IL-1β-treated hTDCs were exposed to. A PEMF actuation with 4 mT, 5 Hz and a 50% duty cycle decreased the production of IL-6 and tumor necrosis factor-α (TNF-α), as well as the expression of TNFα, IL-6, IL-8, COX-2, MMP-1, MMP-2, and MMP-3, while IL-4, IL-10, and TIMP-1 expression increased. These results suggest that PEMF stimulation can modulate hTDCs response in an inflammatory environment holding therapeutic potential for tendon regenerative strategies. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:160-172, 2020.
Collapse
Affiliation(s)
- Adriana Vinhas
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Márcia T Rodrigues
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Barco, Guimarães, 4805-017, Portugal
| | - Ana I Gonçalves
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Barco, Guimarães, 4805-017, Portugal
| | - Manuela E Gomes
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Barco, Guimarães, 4805-017, Portugal
| |
Collapse
|
24
|
Vadlamani RA, Nie Y, Detwiler DA, Dhanabal A, Kraft AM, Kuang S, Gavin TP, Garner AL. Nanosecond pulsed electric field induced proliferation and differentiation of osteoblasts and myoblasts. J R Soc Interface 2019; 16:20190079. [PMID: 31213169 DOI: 10.1098/rsif.2019.0079] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Low-intensity electric fields can induce changes in cell differentiation and cytoskeletal stresses that facilitate manipulation of osteoblasts and mesenchymal stem cells; however, the application times (tens of minutes) are of the order of physiological mechanisms, which can complicate treatment consistency. Intense nanosecond pulsed electric fields (nsPEFs) can overcome these challenges by inducing similar stresses on shorter timescales while additionally inducing plasma membrane nanoporation, ion transport and intracellular structure manipulation. This paper shows that treating myoblasts and osteoblasts with five 300 ns PEFs with intensities from 1.5 to 25 kV cm-1 increased proliferation and differentiation. While nsPEFs above 5 kV cm-1 decreased myoblast population growth, 10 and 20 kV cm-1 trains increased myoblast population by approximately fivefold 48 h after exposure when all cell densities were set to the same level after exposure. Three trials of the PEF-treated osteoblasts showed that PEF trains between 2.5 and 10 kV cm-1 induced the greatest population growth compared to the control 48 h after treatment. Trains of nsPEFs between 1.5 and 5 kV cm-1 induced the most nodule formation in osteoblasts, indicating bone formation. These results demonstrate the potential utility for nsPEFs to rapidly modulate stem cells for proliferation and differentiation and motivate future experiments to optimize PEF parameters for in vivo applications.
Collapse
Affiliation(s)
- Ram Anand Vadlamani
- 1 School of Nuclear Engineering, Purdue University , West Lafayette, IN 47907 , USA
| | - Yaohui Nie
- 2 Department of Health and Kinesiology, Purdue University , West Lafayette, IN 47907 , USA
| | | | - Agni Dhanabal
- 3 Department of Agricultural and Biological Engineering, Purdue University , West Lafayette, IN 47907 , USA
| | - Alan M Kraft
- 1 School of Nuclear Engineering, Purdue University , West Lafayette, IN 47907 , USA
| | - Shihuan Kuang
- 4 Department of Animal Sciences, Purdue University , West Lafayette, IN 47907 , USA
| | - Timothy P Gavin
- 2 Department of Health and Kinesiology, Purdue University , West Lafayette, IN 47907 , USA
| | - Allen L Garner
- 1 School of Nuclear Engineering, Purdue University , West Lafayette, IN 47907 , USA.,3 Department of Agricultural and Biological Engineering, Purdue University , West Lafayette, IN 47907 , USA.,5 School of Electrical and Computer Engineering, Purdue University , West Lafayette, IN 47907 , USA
| |
Collapse
|
25
|
Wang Z, Liu X, Davies MR, Horne D, Kim H, Feeley BT. A Mouse Model of Delayed Rotator Cuff Repair Results in Persistent Muscle Atrophy and Fatty Infiltration. Am J Sports Med 2018; 46:2981-2989. [PMID: 30198747 PMCID: PMC6730552 DOI: 10.1177/0363546518793403] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Rotator cuff (RC) tears are common tendon injuries seen in orthopaedic patients. Successful repair of large and massive RC tears remains a challenge due to our limited understanding of the pathophysiological features of this injury. Clinically relevant small animal models that can be used to study the pathophysiological response to repair are limited by the lack of chronic repair models. PURPOSE To develop a highly clinically relevant mouse model of delayed RC repair. STUDY DESIGN Controlled laboratory study. METHODS Three-month-old C57BL/6J mice underwent unilateral supraspinatus (SS) and infraspinatus (IS) tendon tear with immediate, 2-week delayed, or 6-week delayed tendon repair. Animals with no repair or sham surgery served as controls. Gait analysis was conducted to measure shoulder function at 2 weeks and 6 weeks after surgery. Animals were sacrificed 6 weeks after the last surgery. Shoulder joint, SS, and IS muscles were harvested and analyzed histologically. Ex vivo mechanical testing of intact and repaired SS and IS tendons was conducted. Reverse-transcriptase polymerase chain reaction was performed on SS and IS muscles to quantify atrophy, fibrosis, and fatty infiltration-related gene expression. RESULTS Histological and tendon mechanical testing showed that torn tendons could be successfully repaired as late as 6 weeks after transection. However, significant atrophy and fatty infiltration of muscle, with impaired shoulder function, were persistent in the 6-week delayed repair group. Shoulder function correlated with the severity of RC muscle weight loss and fatty infiltration. CONCLUSION We successfully developed a clinically relevant mouse model of delayed RC repair. Six-week delayed RC repair resulted in persistent muscle atrophy and fatty infiltration with inferior shoulder function compared with acute repair. CLINICAL RELEVANCE Our novel mouse model could serve as a powerful tool to understand the pathophysiological and cellular/molecular mechanisms of RC muscle and tendon degeneration, eventually improving our strategies for treating and repairing RC tears.
Collapse
Affiliation(s)
- Zili Wang
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China.,San Francisco Veterans Affairs Medical Center, Department of Veterans Affairs, San Francisco, CA. USA.,Department of Orthopedic Surgery, University of California at San Francisco, San Francisco, CA. USA
| | - Xuhui Liu
- San Francisco Veterans Affairs Medical Center, Department of Veterans Affairs, San Francisco, CA. USA.,Department of Orthopedic Surgery, University of California at San Francisco, San Francisco, CA. USA
| | - Michael R. Davies
- Department of Orthopedic Surgery, University of California at San Francisco, San Francisco, CA. USA
| | - Devante Horne
- Department of Orthopedic Surgery, University of California at San Francisco, San Francisco, CA. USA
| | - Hubert Kim
- San Francisco Veterans Affairs Medical Center, Department of Veterans Affairs, San Francisco, CA. USA.,Department of Orthopedic Surgery, University of California at San Francisco, San Francisco, CA. USA
| | - Brian T. Feeley
- San Francisco Veterans Affairs Medical Center, Department of Veterans Affairs, San Francisco, CA. USA.,Department of Orthopedic Surgery, University of California at San Francisco, San Francisco, CA. USA
| |
Collapse
|
26
|
Pulsed Electromagnetic Fields Improve Tenogenic Commitment of Umbilical Cord-Derived Mesenchymal Stem Cells: A Potential Strategy for Tendon Repair-An In Vitro Study. Stem Cells Int 2018; 2018:9048237. [PMID: 30154867 PMCID: PMC6091420 DOI: 10.1155/2018/9048237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/05/2018] [Accepted: 04/22/2018] [Indexed: 02/07/2023] Open
Abstract
Tendon repair is a challenging procedure in orthopaedics. The use of mesenchymal stem cells (MSCs) and pulsed electromagnetic fields (PEMF) in tendon regeneration is still investigational. In this perspective, MSCs isolated from the human umbilical cord (UC) may represent a possible candidate for tendon tissue engineering. The aim of the study is to evaluate the effect of low-frequency PEMF on tenogenic differentiation of MSCs isolated from the human umbilical cord (UC-MSCs) in vitro. 15 fresh UC samples from women with healthy pregnancies were retrieved at the end of caesarean deliveries. UC samples were manually minced into small fragments (less than 4 mm length) and cultured in MSC expansion medium. Part of the UC-MSCs was subsequently cultured with PEMF and tenogenic growth factors. UC-MSCs were subjected to pulsed electromagnetic fields for 2 h/day, 4 h/day, or 8 h/day. UC-MSCs cultured with FGF-2 and stimulated with PEMF showed a greater production of collagen type I and scleraxis. The prolonged exposure to PEMF was also related to the greatest expression of tenogenic markers. Thus, the exposure to PEMF provides a positive preconditioning biophysical stimulus, which may enhance UC-MSC tenogenic potential.
Collapse
|
27
|
Yang J, Sun L, Fan X, Yin B, Kang Y, An S, Tang L. Pulsed electromagnetic fields alleviate streptozotocin‑induced diabetic muscle atrophy. Mol Med Rep 2018; 18:1127-1133. [PMID: 29845230 DOI: 10.3892/mmr.2018.9067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 05/15/2018] [Indexed: 11/06/2022] Open
Abstract
Diabetic muscle atrophy causes a reduction of skeletal muscle size and strength, which affects normal daily activities. However, pulsed electromagnetic fields (PEMFs) can retard the atrophy of type II fibers (ActRIIB) in denervated muscles. Therefore, the purpose of the present study was to determine whether PEMFs can alleviate streptozotocin (STZ)‑induced diabetic muscle atrophy. To do this, 40 Sprague‑Dawley (SD) rats were randomly divided into four groups (n=10 per group): The normal control group (NC; nondiabetic rats without treatment); the diabetic mellitus group (DM; STZ‑induced rats without treatment); the diabetic insulin‑treated group (DT; diabetic rats on insulin treatment, 6‑8 U/d twice a day for 6 weeks) as a positive control; and the diabetic PEMFs therapy group (DP; diabetic rats with PEMFs exposure treatment, 15 Hz, 1.46 mT, 30 min/day for 6 weeks). Body weight, muscle strength, muscle mass and serum insulin level were significantly increased in the DP group compared with the DM group. PEMFs also decreased the blood glucose level and altered the activity of metabolic enzymes. PEMFs significantly increased the cross‑sectional area of muscle fiber. In addition, PEMFs significantly activated protein kinase B (Akt) and mammalian target of rapamycin (mTOR), and inhibited the activity of myostatin (MSTN), ActRIIB and forkhead box protein O1 (FoxO1) compared with the DM group. Thus indicating that the Akt/mTOR and Akt/FoxO1 signaling pathways may be involved in the promotion of STZ‑induced diabetic muscle atrophy by PEMFs. The results of the present study suggested that PEMFs stimulation may alleviate diabetic muscle atrophy in the STZ model, and that this is associated with alterations in multiple signaling pathways in which MSTN may be an integral factor. MSTN‑associated signaling pathways may provide therapeutic targets to attenuate severe diabetic muscle wasting.
Collapse
Affiliation(s)
- Jin Yang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710062, P.R. China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| | - Bo Yin
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| | - Yiting Kang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| | - Shucheng An
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710062, P.R. China
| | - Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an, Shaanxi 710119, P.R. China
| |
Collapse
|
28
|
Yin Y, Chen P, Yu Q, Peng Y, Zhu Z, Tian J. The Effects of a Pulsed Electromagnetic Field on the Proliferation and Osteogenic Differentiation of Human Adipose-Derived Stem Cells. Med Sci Monit 2018; 24:3274-3282. [PMID: 29775452 PMCID: PMC5987610 DOI: 10.12659/msm.907815] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background A low frequency pulsed electromagnetic field (PEMF) has been confirmed to play an important role in promoting the osteogenic differentiation of human bone marrow stem cells (BMSCs). Adipose-derived stem cells (ASCs) possess some attractive characteristics for clinical application compared to BMSCs, such as abundant stem cells from lipoaspirates, faster growth, less discomfort and morbidity during surgery. ASCs can become adipocytes, osteoblasts, chondrocytes, myocytes, neurocytes, and other cell types. Thus, ASCs might be a good alternative in clinical work involving treatment with PEMF. Material/Methods Human ASCs (hASCs)were divided into a control group (without PEMF exposure) and an experimental group (PEMF for two hours per day). We examined the effect of PEMF on promoting cell proliferation and osteogenic differentiation from several aspects: CCK-8 proliferation assay, RNA extraction, qRT-PCR detection, western blotting, and immunofluorescence staining experiments. Results PEMF could promote cell proliferation of human ASCs (hASCs) at an early stage as determined by CCK-8 assay. A specific intensity (1 mT) and frequency (50 Hz) of PEMF promoted osteogenic differentiation in hASCs in alkaline phosphatase (ALP) staining experiments. In addition, bone-related gene expression increased after two weeks of PEMF exposure, the protein expression of OPN, OCN, and RUNX-2 also increased after a longer period (three weeks) of PEMF treatment as determined by western blotting and immunofluorescence staining. Conclusions We found for the first time that PMEF has a role in stimulating cell proliferation of hASCs at an early period, subsequently promoting bone-related gene expression and inducing the expression of related proteins to stimulate osteogenic differentiation.
Collapse
Affiliation(s)
- Yukun Yin
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Ping Chen
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Qiang Yu
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Yan Peng
- Department of Human Anatomy, Basic Medical College, Southern Medical University, Baiyun, Guangzhou, China (mainland)
| | - ZeHao Zhu
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Haizhu, Guangzhou, China (mainland)
| | - Jing Tian
- Department of Orthopedics, Zhujiang Hospital,Southern Medical University, Haizhu, Guangzhou, China (mainland)
| |
Collapse
|
29
|
Tang X, Alliston T, Coughlin D, Miller S, Zhang N, Waldorff EI, Ryaby JT, Lotz JC. Dynamic imaging demonstrates that pulsed electromagnetic fields (PEMF) suppress IL-6 transcription in bovine nucleus pulposus cells. J Orthop Res 2018; 36:778-787. [PMID: 28851112 PMCID: PMC5873378 DOI: 10.1002/jor.23713] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 08/19/2017] [Indexed: 02/04/2023]
Abstract
Inflammatory cytokines play a dominant role in the pathogenesis of disc degeneration. Pulsed electromagnetic fields (PEMF) are noninvasive biophysical stimulus that has been used extensively in the orthopaedic field for many years. However, the specific cellular responses and mechanisms involved are still unclear. The objective of this study was to assess the time-dependent PEMF effects on pro-inflammatory factor IL-6 expression in disc nucleus pulposus cells using a novel green fluorescence protein (GFP) reporter system. An MS2-tagged GFP reporter system driven by IL-6 promoter was constructed to visualize PEMF treatment effect on IL-6 transcription in single living cells. IL-6-MS2 reporter-labeled cells were treated with IL-1α to mimic the in situ inflammatory environment of degenerative disc while simultaneously exposed to PEMF continuously for 4 h. Time-lapse imaging was recorded using a confocal microscope to track dynamic IL-6 transcription activity that was demonstrated by GFP. Finally, real-time RT-PCR was performed to confirm the imaging data. Live cell imaging demonstrated that pro-inflammatory factor IL-1α significantly promoted IL-6 transcription over time as compared with DMEM basal medium condition. Imaging and PCR data demonstrated that the inductive effect of IL-1α on IL-6 expression could be significantly inhibited by PEMF treatment in a time-dependent manner (early as 2 h of stimulus initiation). Our data suggest that PEMF may have a role in the clinical management of patients with chronic low back pain. Furthermore, this study shows that the MS2-tagged GFP reporter system is a useful tool for visualizing the dynamic events of mechanobiology in musculoskeletal research. © 2017 The Authors. Journal of Orthopaedic Research® Published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 35:778-787, 2018.
Collapse
Affiliation(s)
- Xinyan Tang
- Department of Orthopaedic SurgeryUniversity of California San FranciscoSan FranciscoCalifornia
| | - Tamara Alliston
- Department of Orthopaedic SurgeryUniversity of California San FranciscoSan FranciscoCalifornia
| | - Dezba Coughlin
- Department of Orthopaedic SurgeryUniversity of California San FranciscoSan FranciscoCalifornia
| | - Stephanie Miller
- Department of Orthopaedic SurgeryUniversity of California San FranciscoSan FranciscoCalifornia
| | | | | | | | - Jeffrey C. Lotz
- Department of Orthopaedic SurgeryUniversity of California San FranciscoSan FranciscoCalifornia
| |
Collapse
|
30
|
Iwasa K, Reddi AH. Pulsed Electromagnetic Fields and Tissue Engineering of the Joints. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:144-154. [PMID: 29020880 DOI: 10.1089/ten.teb.2017.0294] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Bone and joint formation, maintenance, and regeneration are regulated by both chemical and physical signals. Among the physical signals there is an increasing realization of the role of pulsed electromagnetic fields (PEMF) in the treatment of nonunions of bone fractures. The discovery of the piezoelectric properties of bone by Fukada and Yasuda in 1953 in Japan established the foundation of this field. Pioneering research by Bassett and Brighton and their teams resulted in the approval by the Food and Drug Administration (FDA) of the use of PEMF in the treatment of fracture healing. Although PEMF has potential applications in joint regeneration in osteoarthritis (OA), this evolving field is still in its infancy and offers novel opportunities. METHODS We have systematically reviewed the literature on the influence of PEMF in joints, including articular cartilage, tendons, and ligaments, of publications from 2000 to 2016. CONCLUSIONS PEMF stimulated chondrocyte proliferation, differentiation, and extracellular matrix synthesis by release of anabolic morphogens such as bone morphogenetic proteins and anti-inflammatory cytokines by adenosine receptors A2A and A3 in both in vitro and in vivo investigations. It is noteworthy that in clinical translational investigations a beneficial effect was observed on improving function in OA knees. However, additional systematic studies on the mechanisms of action of PEMF on joints and tissues therein, articular cartilage, tendons, and ligaments are required.
Collapse
Affiliation(s)
- Kenjiro Iwasa
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California , Davis, Davis, California
| | - A Hari Reddi
- Department of Orthopaedic Surgery, Lawrence Ellison Center for Tissue Regeneration and Repair, School of Medicine, University of California , Davis, Davis, California
| |
Collapse
|
31
|
Ng JL, Kersh ME, Kilbreath S, Knothe Tate M. Establishing the Basis for Mechanobiology-Based Physical Therapy Protocols to Potentiate Cellular Healing and Tissue Regeneration. Front Physiol 2017; 8:303. [PMID: 28634452 PMCID: PMC5460618 DOI: 10.3389/fphys.2017.00303] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 04/27/2017] [Indexed: 12/17/2022] Open
Abstract
Life is mechanobiological: mechanical stimuli play a pivotal role in the formation of structurally and functionally appropriate body templates through mechanobiologically-driven cellular and tissue re/modeling. The body responds to mechanical stimuli engendered through physical movement in an integrated fashion, internalizing and transferring forces from organ, through tissue and cellular length scales. In the context of rehabilitation and therapeutic outcomes, such mechanical stimuli are referred to as mechanotherapy. Physical therapists use mechanotherapy and mechanical interventions, e.g., exercise therapy and manual mobilizations, to restore function and treat disease and/or injury. While the effect of directed movement, such as in physical therapy, is well documented at the length scale of the body and its organs, a number of recent studies implicate its integral effect in modulating cellular behavior and subsequent tissue adaptation. Yet the link between movement biomechanics, physical therapy, and subsequent cellular and tissue mechanoadaptation is not well established in the literature. Here we review mechanoadaptation in the context of physical therapy, from organ to cell scale mechanotransduction and cell to organ scale extracellular matrix genesis and re/modeling. We suggest that physical therapy can be developed to harness the mechanosensitivity of cells and tissues, enabling prescriptive definition of physical and mechanical interventions to enhance tissue genesis, healing, and rehabilitation.
Collapse
Affiliation(s)
- Joanna L. Ng
- Graduate School of Biomedical Engineering, University of New South WalesSydney, NSW, Australia
| | - Mariana E. Kersh
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-ChampaignChampaign, IL, United States
| | - Sharon Kilbreath
- Faculty of Health Sciences, University of SydneySydney, NSW, Australia
| | - M. Knothe Tate
- Graduate School of Biomedical Engineering, University of New South WalesSydney, NSW, Australia
| |
Collapse
|
32
|
Waldorff EI, Zhang N, Ryaby JT. Pulsed electromagnetic field applications: A corporate perspective. J Orthop Translat 2017; 9:60-68. [PMID: 29662800 PMCID: PMC5822965 DOI: 10.1016/j.jot.2017.02.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 11/30/2022] Open
Abstract
Corporate establishment of US Food & Drug Administration approved pulsed electromagnetic fields (PEMFs) for clinical applications has been achieved. However, optimization of PEMFs for improvement in efficacy for current indications, in addition to the expansion into new indications, is not trivial. Moving directly into a clinical trial can be costly and carries little guarantee for success, necessitating the need for preclinical studies as supported by this review of the extensive corporate preclinical experience by Orthofix, Inc. The Translational Potential of this Article: This review illustrates the need to gain enough in vitro/in vivo knowledge of specific PEMF signals and its target tissue interaction to enable a high success rate in clinical trials.
Collapse
|