1
|
Liu Y, Tanaka E. Pathogenesis, Diagnosis, and Management of Trigeminal Neuralgia: A Narrative Review. J Clin Med 2025; 14:528. [PMID: 39860534 PMCID: PMC11765769 DOI: 10.3390/jcm14020528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Trigeminal neuralgia (TN) is an excruciating neurological disorder characterized by intense, stimulus-induced, and transient facial stabbing pain. The classification of TN has changed as a result of new discoveries in the last decade regarding its symptomatology, pathogenesis, and management. Because different types of facial pain have different clinical therapy and neuroimaging interpretations, a precise diagnosis is essential. Diagnosis should include magnetic resonance imaging with specific sequences to rule out secondary causes and to identify possible neurovascular contact. The purpose of demonstrating a neurovascular contact is to aid in surgical decision making, not to validate a diagnosis. Microvascular decompression is the first-line procedure for individuals who do not respond to medical management, whereas carbamazepine and oxcarbazepine are the preferred medications for long-term care. New developments in animal models and neuroimaging methods will shed more light on the biology and etiology of TN. This paper reviews the pathogenesis, the clinical features, the diagnosis, and the management of TN. Furthermore, the potential role of low-intensity pulsed ultrasound in neurological disorders is discussed.
Collapse
Affiliation(s)
- Yao Liu
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China;
| | - Eiji Tanaka
- Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China;
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8504, Japan
| |
Collapse
|
2
|
Pirri C. Exploring the Revolutionary Impact of YAP Pathways on Physical and Rehabilitation Medicine. Biomolecules 2025; 15:96. [PMID: 39858490 PMCID: PMC11764055 DOI: 10.3390/biom15010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Cellular behavior is strongly influenced by mechanical signals in the surrounding microenvironment, along with external factors such as temperature fluctuations, changes in blood flow, and muscle activity, etc. These factors are key in shaping cellular states and can contribute to the development of various diseases. In the realm of rehabilitation physical therapies, therapeutic exercise and manual treatments, etc., are frequently employed, not just for pain relief but also to support recovery from diverse health conditions. However, the detailed molecular pathways through which these therapies interact with tissues and influence gene expression are not yet fully understood. The identification of YAP has been instrumental in closing this knowledge gap. YAP is known for its capacity to perceive and translate mechanical signals into specific transcriptional programs within cells. This insight has opened up new perspectives on how physical and rehabilitation medicine may exert its beneficial effects. The review investigates the involvement of the Hippo/YAP signaling pathway in various diseases and considers how different rehabilitation techniques leverage this pathway to aid in healing. Additionally, it examines the therapeutic potential of modulating the Hippo/YAP pathway within the context of rehabilitation, while also addressing the challenges and controversies that surround its use in physical and rehabilitation medicine.
Collapse
Affiliation(s)
- Carmelo Pirri
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, 35121 Padova, Italy
| |
Collapse
|
3
|
Marcotulli M, Barbetta A, Scarpa E, Bini F, Marinozzi F, Ruocco G, Casciola CM, Scognamiglio C, Carugo D, Cidonio G. Jingle Cell Rock: Steering Cellular Activity With Low-Intensity Pulsed Ultrasound (LIPUS) to Engineer Functional Tissues in Regenerative Medicine. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1973-1986. [PMID: 39289118 DOI: 10.1016/j.ultrasmedbio.2024.08.016] [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: 03/15/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024]
Abstract
Acoustic manipulation or perturbation of biological soft matter has emerged as a promising clinical treatment for a number of applications within regenerative medicine, ranging from bone fracture repair to neuromodulation. The potential of ultrasound (US) endures in imparting mechanical stimuli that are able to trigger a cascade of molecular signals within unscathed cells. Particularly, low-intensity pulsed ultrasound (LIPUS) has been associated with bio-effects such as activation of specific cellular pathways and alteration of cell morphology and gene expression, the extent of which can be modulated by fine tuning of LIPUS parameters including intensity, frequency and exposure time. Although the molecular mechanisms underlying LIPUS are not yet fully elucidated, a number of studies clearly define the modulation of specific ultrasonic parameters as a means to guide the differentiation of a specific set of stem cells towards adult and fully differentiated cell types. Herein, we outline the applications of LIPUS in regenerative medicine and the in vivo and in vitro studies that have confirmed the unbounded clinical potential of this platform. We highlight the latest developments aimed at investigating the physical and biological mechanisms of action of LIPUS, outlining the most recent efforts in using this technology to aid tissue engineering strategies for repairing tissue or modelling specific diseases. Ultimately, we detail tissue-specific applications harnessing LIPUS stimuli, offering insights over the engineering of new constructs and therapeutic modalities. Overall, we aim to lay the foundation for a deeper understanding of the mechanisms governing LIPUS-based therapy, to inform the development of safer and more effective tissue regeneration strategies in the field of regenerative medicine.
Collapse
Affiliation(s)
- Martina Marcotulli
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy; Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy; Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Andrea Barbetta
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Edoardo Scarpa
- Infection Dynamics Laboratory, Department of Pharmaceutical Sciences, University of Milan, Milan, Italy; National Institute of Molecular Gentics (INGM), Milan, Italy; Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - Fabiano Bini
- Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy
| | - Franco Marinozzi
- Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ruocco
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy
| | - Carlo Massimo Casciola
- Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy
| | - Chiara Scognamiglio
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy
| | - Dario Carugo
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Gianluca Cidonio
- 3D Microfluidic Bioprinting Lab, Center for Life Nano- & Neuro-Science (CLN2S), Italian Institute of Technology (IIT), Rome, Italy; Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, UK; Department of Mechanical and Aerospace Engineering (DIMA), Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
4
|
Qi L, Wang C, Deng L, Pan JJ, Suo Q, Wu S, Cai L, Shi X, Sun J, Wang Y, Tang Y, Qiu W, Yang GY, Wang J, Zhang Z. Low-intensity focused ultrasound stimulation promotes stroke recovery via astrocytic HMGB1 and CAMK2N1 in mice. Stroke Vasc Neurol 2024; 9:505-518. [PMID: 38191183 PMCID: PMC11732924 DOI: 10.1136/svn-2023-002614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 11/28/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Low-intensity focused ultrasound stimulation (LIFUS) has been developed to enhance neurological repair and remodelling during the late acute stage of ischaemic stroke in rodents. However, the cellular and molecular mechanisms of neurological repair and remodelling after LIFUS in ischaemic stroke are unclear. METHODS Ultrasound stimulation was treated in adult male mice 7 days after transient middle cerebral artery occlusion. Angiogenesis was measured by laser speckle imaging and histological analyses. Electromyography and fibre photometry records were used for synaptogenesis. Brain atrophy volume and neurobehaviour were assessed 0-14 days after ischaemia. iTRAQ proteomic analysis was performed to explore the differentially expressed protein. scRNA-seq was used for subcluster analysis of astrocytes. Fluorescence in situ hybridisation and Western blot detected the expression of HMGB1 and CAMK2N1. RESULTS Optimal ultrasound stimulation increased cerebral blood flow, and improved neurobehavioural outcomes in ischaemic mice (p<0.05). iTRAQ proteomic analysis revealed that the expression of HMGB1 increased and CAMK2N1 decreased in the ipsilateral hemisphere of the brain at 14 days after focal cerebral ischaemia with ultrasound treatment (p<0.05). scRNA-seq revealed that this expression pattern belonged to a subcluster of astrocytes after LIFUS in the ischaemic brain. LIFUS upregulated HMGB1 expression, accompanied by VEGFA elevation compared with the control group (p<0.05). Inhibition of HMGB1 expression in astrocytes decreased microvessels counts and cerebral blood flow (p<0.05). LIFUS reduced CAMK2N1 expression level, accompanied by increased extracellular calcium ions and glutamatergic synapses (p<0.05). CAMK2N1 overexpression in astrocytes decreased dendritic spines, and aggravated neurobehavioural outcomes (p<0.05). CONCLUSION Our results demonstrated that LIFUS promoted angiogenesis and synaptogenesis after focal cerebral ischaemia by upregulating HMGB1 and downregulating CAMK2N1 in a subcluster of astrocytes, suggesting that LIFUS activated specific astrocyte subcluster could be a key target for ischaemic brain therapy.
Collapse
MESH Headings
- Animals
- Astrocytes/metabolism
- Astrocytes/pathology
- Astrocytes/enzymology
- Male
- Recovery of Function
- Disease Models, Animal
- Infarction, Middle Cerebral Artery/metabolism
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/therapy
- Infarction, Middle Cerebral Artery/physiopathology
- Infarction, Middle Cerebral Artery/genetics
- Infarction, Middle Cerebral Artery/enzymology
- Mice, Inbred C57BL
- HMGB1 Protein/metabolism
- HMGB1 Protein/genetics
- Neovascularization, Physiologic
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Cerebrovascular Circulation
- Ultrasonic Therapy
- Signal Transduction
- Ischemic Stroke/metabolism
- Ischemic Stroke/physiopathology
- Ischemic Stroke/therapy
- Ischemic Stroke/pathology
- Behavior, Animal
- Time Factors
- Mice
- Proteomics
- Neurogenesis
Collapse
Affiliation(s)
- Lin Qi
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Cheng Wang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Lidong Deng
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Jia-Ji Pan
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical Collage, Fudan University, Shanghai, China
| | - Qian Suo
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Shengju Wu
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Lin Cai
- Department of Neurosurgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Xudong Shi
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Beijing, China
| | - Junfeng Sun
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Yongting Wang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Yaohui Tang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Weibao Qiu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Beijing, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, China
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Jixian Wang
- Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Zhijun Zhang
- Shanghai Jiao Tong Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| |
Collapse
|
5
|
Yang F, Lv J, Ma W, Yang Y, Hu X, Yang Z. Engineering Sonosensitizer-Derived Nanotheranostics for Augmented Sonodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402669. [PMID: 38970544 DOI: 10.1002/smll.202402669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/13/2024] [Indexed: 07/08/2024]
Abstract
Sonodynamic therapy (SDT), featuring noninvasive, deeper penetration, low cost, and repeatability, is a promising therapy approach for deep-seated tumors. However, the general or only utilization of SDT shows low efficiency and unsatisfactory treatment outcomes due to the complicated tumor microenvironment (TME) and SDT process. To circumvent the issues, three feasible approaches for enhancing SDT-based therapeutic effects, including sonosensitizer optimization, strategies for conquering hypoxia TME, and combinational therapy are summarized, with a particular focus on the combination therapy of SDT with other therapy modalities, including chemodynamic therapy, photodynamic therapy, photothermal therapy, chemotherapy, starvation therapy, gas therapy, and immunotherapy. In the end, the current challenges in SDT-based therapy on tumors are discussed and feasible approaches for enhanced therapeutic effects are provided. It is envisioned that this review will provide new insight into the strategic design of high-efficiency sonosensitizer-derived nanotheranostics, thereby augmenting SDT and accelerating the potential clinical transformation.
Collapse
Affiliation(s)
- Fuhong Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Jingqi Lv
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Wen Ma
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Yanling Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| | - Xiaoming Hu
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
- Jiangxi Key Laboratory of Nanobiomaterials, School of Materials Science and Engineering, East China Jiaotong University, Nanchang, 330013, China
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, 350117, China
| |
Collapse
|
6
|
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
|
7
|
Hu X, Duan H, Zou D, Dong C, Wang Y, Wang Y, Li Z, Li Z. Acoustic vibration promotes in vitro expansion of human embryonic stem cells. AMERICAN JOURNAL OF STEM CELLS 2024; 13:143-151. [PMID: 39021373 PMCID: PMC11249672 DOI: 10.62347/pjfc2708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/06/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVES This study aimed to investigate the effect of acoustic vibration on the pluripotency of human embryonic stem cells (hESCs) and evaluate cell proliferation and self-renewal ability post-treatment. METHODS The human ES cell line H1 was used for the experiments. hESCs were treated with an acoustic vibration device. Their proliferative ability was subsequently detected using a colony formation assay, while the expression of pluripotency-related markers was detected via immunofluorescence staining. Finally, changes in gene expression levels were examined using quantitative polymerase chain reaction (qPCR) in the presence of appropriate primers. RESULTS Compared with normal cells in the control group, the morphology of experimental cells subjected to acoustic vibration did not significantly change. Contrastingly, the colony-forming efficiency of the experimental cells significantly increased. Immunofluorescence staining results showed the cells in experimental group were positive for the pluripotency markers NANOG, octamer-binding transcription factor 4 gene (OCT4), and SRY (sex determining region Y)-box 2 (SOX2). In addition, the expression levels of pluripotency genes NANOG, OCT4, SOX2, and Yes-associated protein (YAP)-related genes were up-regulated following acoustic vibration. CONCLUSIONS Our results revealed that acoustic vibration enhanced the proliferative ability of hESCs and increased the expression levels of NANOG, OCT4, SOX2, and YAP-related genes, indicating that acoustic vibration can optimize the self-renewal ability of hESCs and that the YAP signaling pathway may play a critical role in the functional process of acoustic vibration.
Collapse
Affiliation(s)
- Xiangyue Hu
- Shandong First Medical University (Shandong Academy of Medical Sciences)Jinan 250000, Shandong, China
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
| | - Haoyun Duan
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
| | - Dulei Zou
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
- Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital)Jinan 250021, Shandong, China
| | - Chunxiao Dong
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
- Eye Institute of Shandong First Medical University, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital)Jinan 250021, Shandong, China
| | - Yani Wang
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
| | - Yao Wang
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
| | - Zongren Li
- 970 Hospital of Chinese PLA Joint Logistic Support ForceWeihai 264200, Shandong, China
| | - Zongyi Li
- Shandong First Medical University (Shandong Academy of Medical Sciences)Jinan 250000, Shandong, China
- Eye Institute of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of OphthalmologyQingdao 266071, Shandong, China
| |
Collapse
|
8
|
Pan C, Hao X, Deng X, Lu F, Liu J, Hou W, Xu T. The roles of Hippo/YAP signaling pathway in physical therapy. Cell Death Discov 2024; 10:197. [PMID: 38670949 PMCID: PMC11053014 DOI: 10.1038/s41420-024-01972-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Cellular behavior is regulated by mechanical signals within the cellular microenvironment. Additionally, changes of temperature, blood flow, and muscle contraction also affect cellular state and the development of diseases. In clinical practice, physical therapy techniques such as ultrasound, vibration, exercise, cold therapy, and hyperthermia are commonly employed to alleviate pain and treat diseases. However, the molecular mechanism about how these physiotherapy methods stimulate local tissues and control gene expression remains unknow. Fortunately, the discovery of YAP filled this gap, which has been reported has the ability to sense and convert a wide variety of mechanical signals into cell-specific programs for transcription, thereby offering a fresh perspective on the mechanisms by which physiotherapy treat different diseases. This review examines the involvement of Hippo/YAP signaling pathway in various diseases and its role in different physical therapy approaches on diseases. Furthermore, we explore the potential therapeutic implications of the Hippo/YAP signaling pathway and address the limitations and controversies surrounding its application in physiotherapy.
Collapse
Affiliation(s)
- Chunran Pan
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Hao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofeng Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Lu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Liu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjie Hou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
9
|
Ortiz R, Motyka T, Petterson S, Krystofiak J. Sustained acoustic medicine treatment of discogenic chronic low back pain: A randomized, multisite, double-blind, placebo-controlled trial. J Back Musculoskelet Rehabil 2024; 37:1321-1332. [PMID: 38905030 PMCID: PMC11492000 DOI: 10.3233/bmr-230402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/28/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Sustained acoustic medicine (SAM) is a noninvasive long-term treatment that provides essential mechanical and thermal stimulus to accelerate soft tissue healing, alleviate pain, and improve physical activity. SAM increases localized deep tissue temperature, blood flow, cellular proliferation, migration, and nutrition exchange, resulting in reduced inflammation and an increased rate of tissue regeneration. OBJECTIVE To assess the efficacy of SAM treatment of discogenic back pain in the lower spinal column to reduce pain, improve quality of life, and lower pharmacotherapy use. METHODS Sixty-five subjects with chronic low back pain were randomly assigned to SAM (N= 33) or placebo (N= 32) groups. Subjects self-applied SAM device bilaterality on the lower lumbar region for 4 hours daily for 8 weeks and completed daily pain diaries before, during, and after treatment. Subjects recorded pain reduction using a numeric rating scale (NRS), medication use, and physical activity using the Global Rating of Change (GROC) and Oswestry Disability Index (ODI). RESULTS SAM treatment significantly reduced chronic lower back pain from baseline relative to placebo treatment (p< 0.0001). SAM treated subjects reported significantly lower back pain at 4 weeks, with the highest pain reduction (-2.58 points NRS, p< 0.0001) reported at 8 weeks. Similar trends were observed in improved physical activity (3.48 GROC, p< 0.0001, 69-88% ODI, p< 0.0001) and 22.5% (15.2 morphine milligram equivalent) reduction in the use of opioid medication from baseline to 8 weeks. CONCLUSION Daily, home-use SAM treatment significantly improves the clinical symptoms of chronic lower back pain, improves physical mobility, and reduces daily medication use. SAM treatment is well-tolerated by patients and may be considered a safe, non-invasive treatment option for chronic discogenic, lower back pain.
Collapse
Affiliation(s)
- Ralph Ortiz
- Department of Pain Management, Cayuga Medical Center, Ithaca, NY, USA
| | - Thomas Motyka
- Department of Osteopathic Medicine, Campbell University, Buies Creek, NC, USA
| | | | - Jason Krystofiak
- Department of Orthopedics, Rutgers University, Barnabas Health, New Brunswick, NJ, USA
| |
Collapse
|
10
|
Sasaki R, Sakamoto J, Honda Y, Motokawa S, Kataoka H, Origuchi T, Okita M. Low-intensity pulsed ultrasound phonophoresis with diclofenac alleviated inflammation and pain via downregulation of M1 macrophages in rats with carrageenan-induced knee joint arthritis. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 15:100148. [PMID: 38174056 PMCID: PMC10758990 DOI: 10.1016/j.ynpai.2023.100148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 01/05/2024]
Abstract
Objective This study aimed to investigate the effects of low-intensity pulsed ultrasound (LIPUS) phonophoresis with diclofenac on inflammation and pain in the acute phase of carrageenan-induced arthritis in rats. Design 60 male Wistar rats were randomly divided into the arthritis, diclofenac, LIPUS, phonophoresis, and sham-arthritis control groups. LIPUS and transdermal diclofenac gel were applied to the lateral side of the inflamed knee for 7 days, initiated postinjection day 1. In the phonophoresis group, diclofenac gel was rubbed onto the skin, followed by LIPUS application over the medication. Knee joint transverse diameters, pressure pain thresholds (PPTs), and paw withdrawal thresholds (PWT) were evaluated. The number of CD68-, CD11c-, and CD206-positive cells, and IL-1β and COX-2 mRNA expression were analyzed 8 days after injection. Results In the phonophoresis group, the transverse diameter, PPT, PWT significantly recovered at the day 8 compared to those in the LIPUS and diclofenac groups. The number of CD68- and CD11c-positive cells in the phonophoresis group was significantly lower than that in the LIPUS and diclofenac groups, but no significant differences were observed among three groups in CD206-positive cells. IL-1β and COX-2 mRNA levels were lower in the phonophoresis group than in the arthritis group, although there were no differences among the LIPUS, diclofenac, and phonophoresis groups. Conclusion LIPUS phonophoresis with diclofenac is more effective to ameliorate inflammation and pain compared to diclofenac or LIPUS alone, and the mechanism involves the decrease of M1 macrophages.
Collapse
Affiliation(s)
- Ryo Sasaki
- Department of Rehabilitation, Juzenkai Hospital, Nagasaki, Japan
- Department of Physical Therapy Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Junya Sakamoto
- Department of Physical Therapy Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Institute of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yuichiro Honda
- Department of Physical Therapy Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Institute of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Satoko Motokawa
- Department of Physical Therapy Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Rehabilitation, Nagasaki Rehabilitation Hospital, Nagasaki, Japan
| | - Hideki Kataoka
- Department of Physical Therapy Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Rehabilitation, Nagasaki Memorial Hospital, Nagasaki, Japan
| | - Tomoki Origuchi
- Department of Physical Therapy Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Institute of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Minoru Okita
- Department of Physical Therapy Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Institute of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
11
|
Wang Y, Liu Z, Pan C, Zheng Y, Chen Y, Lian X, Jiang Y, Chen C, Xue K, Zhang Y, Xu P, Liu K. Ultrasound-Driven Healing: Unleashing the Potential of Chondrocyte-Derived Extracellular Vesicles for Chondrogenesis in Adipose-Derived Stem Cells. Biomedicines 2023; 11:2836. [PMID: 37893208 PMCID: PMC10604747 DOI: 10.3390/biomedicines11102836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Repairing cartilage defects represents a significant clinical challenge. While adipose-derived stem cell (ADSC)-based strategies hold promise for cartilage regeneration, their inherent chondrogenic potential is limited. Extracellular vesicles (EVs) derived from chondrocytes (CC-EVs) have shown potential in enhancing chondrogenesis, but their role in promoting chondrogenic differentiation of ADSCs remains poorly understood. Moreover, the clinical application of EVs faces limitations due to insufficient quantities for in vivo use, necessitating the development of effective methods for extracting significant amounts of CC-EVs. Our previous study demonstrated that low-intensity ultrasound (LIUS) stimulation enhances EV secretion from mesenchymal stem cells. Here, we identified a specific LIUS parameter for chondrocytes that increased EV secretion by 16-fold. CC-EVs were found to enhance cell activity, proliferation, migration, and 21-day chondrogenic differentiation of ADSCs in vitro, while EVs secreted by chondrocytes following LIUS stimulation (US-CC-EVs) exhibited superior efficacy. miRNA-seq revealed that US-CC-EVs were enriched in cartilage-regeneration-related miRNAs, contributing to chondrogenesis in various biological processes. In conclusion, we found that CC-EVs can enhance the chondrogenesis of ADSCs in vitro. In addition, our study introduces ultrasound-driven healing as an innovative method to enhance the quantity and quality of CC-EVs, meeting clinical demand and addressing the limited chondrogenic potential of ADSCs. The ultrasound-driven healing unleashes the potential of CC-EVs for chondrogenesis possibly through the enrichment of cartilage-regeneration-associated miRNAs in EVs, suggesting their potential role in cartilage reconstruction. These findings hold promise for advancing cartilage regeneration strategies and may pave the way for novel therapeutic interventions in regenerative medicine.
Collapse
Affiliation(s)
- Yikai Wang
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Zibo Liu
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Chuqiao Pan
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Yi Zheng
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Yahong Chen
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Xiang Lian
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Yu Jiang
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Chuhsin Chen
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Ke Xue
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC 27101, USA;
| | - Peng Xu
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| | - Kai Liu
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; (Y.W.); (Z.L.); (C.P.); (Y.Z.); (Y.C.); (X.L.); (Y.J.); (C.C.); (K.X.)
| |
Collapse
|
12
|
Vinikoor T, Dzidotor GK, Le TT, Liu Y, Kan HM, Barui S, Chorsi MT, Curry EJ, Reinhardt E, Wang H, Singh P, Merriman MA, D'Orio E, Park J, Xiao S, Chapman JH, Lin F, Truong CS, Prasadh S, Chuba L, Killoh S, Lee SW, Wu Q, Chidambaram RM, Lo KWH, Laurencin CT, Nguyen TD. Injectable and biodegradable piezoelectric hydrogel for osteoarthritis treatment. Nat Commun 2023; 14:6257. [PMID: 37802985 PMCID: PMC10558537 DOI: 10.1038/s41467-023-41594-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/11/2023] [Indexed: 10/08/2023] Open
Abstract
Osteoarthritis affects millions of people worldwide but current treatments using analgesics or anti-inflammatory drugs only alleviate symptoms of this disease. Here, we present an injectable, biodegradable piezoelectric hydrogel, made of short electrospun poly-L-lactic acid nanofibers embedded inside a collagen matrix, which can be injected into the joints and self-produce localized electrical cues under ultrasound activation to drive cartilage healing. In vitro, data shows that the piezoelectric hydrogel with ultrasound can enhance cell migration and induce stem cells to secrete TGF-β1, which promotes chondrogenesis. In vivo, the rabbits with osteochondral critical-size defects receiving the ultrasound-activated piezoelectric hydrogel show increased subchondral bone formation, improved hyaline-cartilage structure, and good mechanical properties, close to healthy native cartilage. This piezoelectric hydrogel is not only useful for cartilage healing but also potentially applicable to other tissue regeneration, offering a significant impact on the field of regenerative tissue engineering.
Collapse
Affiliation(s)
- Tra Vinikoor
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Godwin K Dzidotor
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Thinh T Le
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Yang Liu
- Center of Digital Dentistry/Department of Prosthodontics/Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Research Center of Engineering and Technology for Computerized Dentistry & NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Ho-Man Kan
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Srimanta Barui
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Meysam T Chorsi
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Eli J Curry
- Eli Lilly and Company, 450 Kendall Street, Cambridge, MA, 02142, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Emily Reinhardt
- Department of Pathobiology and Veterinary Science, University of Connecticut, 61 North Eagleville Road, Unit 3089, Storrs, CT, 06269, USA
| | - Hanzhang Wang
- Pathology and Laboratory Medicine, University of Connecticut Health Center, 63 Farmington Avenue, Farmington, CT, 06030, USA
| | - Parbeen Singh
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Marc A Merriman
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Ethan D'Orio
- Department of Advanced Manufacturing for Energy Systems Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Jinyoung Park
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Shuyang Xiao
- Department of Materials Science and Engineering & Institute of Materials Science, University of Connecticut, 25 King Hill Road, Unit 3136, Storrs, CT, 06269-3136, USA
| | - James H Chapman
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
| | - Feng Lin
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Cao-Sang Truong
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Somasundaram Prasadh
- Center for Clean Energy Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Lisa Chuba
- Center for Comparative Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Shaelyn Killoh
- Center for Comparative Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Seok-Woo Lee
- Department of Materials Science and Engineering & Institute of Materials Science, University of Connecticut, 25 King Hill Road, Unit 3136, Storrs, CT, 06269-3136, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
| | - Qian Wu
- Pathology and Laboratory Medicine, University of Connecticut Health Center, 63 Farmington Avenue, Farmington, CT, 06030, USA
| | - Ramaswamy M Chidambaram
- Center for Comparative Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Kevin W H Lo
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Cato T Laurencin
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, 06030, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science and Engineering & Institute of Materials Science, University of Connecticut, 25 King Hill Road, Unit 3136, Storrs, CT, 06269-3136, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery University of Connecticut Health, Farmington, CT, 06030, USA
| | - Thanh D Nguyen
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, USA.
| |
Collapse
|
13
|
Sharif NA. Electrical, Electromagnetic, Ultrasound Wave Therapies, and Electronic Implants for Neuronal Rejuvenation, Neuroprotection, Axonal Regeneration, and IOP Reduction. J Ocul Pharmacol Ther 2023; 39:477-498. [PMID: 36126293 DOI: 10.1089/jop.2022.0046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The peripheral nervous system (PNS) of mammals and nervous systems of lower organisms possess significant regenerative potential. In contrast, although neural plasticity can provide some compensation, the central nervous system (CNS) neurons and nerves of adult mammals generally fail to regenerate after an injury or damage. However, use of diverse electrical, electromagnetic and sonographic energy waves are illuminating novel ways to stimulate neuronal differentiation, proliferation, neurite growth, and axonal elongation/regeneration leading to various levels of functional recovery in animals and humans afflicted with disorders of the CNS, PNS, retina, and optic nerve. Tools such as acupuncture, electroacupuncture, electroshock therapy, electrical stimulation, transcranial magnetic stimulation, red light therapy, and low-intensity pulsed ultrasound therapy are demonstrating efficacy in treating many different maladies. These include wound healing, partial recovery from motor dysfunctions, recovery from ischemic/reperfusion insults and CNS and ocular remyelination, retinal ganglion cell (RGC) rejuvenation, and RGC axonal regeneration. Neural rejuvenation and axonal growth/regeneration processes involve activation or intensifying of the intrinsic bioelectric waves (action potentials) that exist in every neuronal circuit of the body. In addition, reparative factors released at the nerve terminals and via neuronal dendrites (transmitter substances), extracellular vesicles containing microRNAs and neurotrophins, and intercellular communication occurring via nanotubes aid in reestablishing lost or damaged connections between the traumatized tissues and the PNS and CNS. Many other beneficial effects of the aforementioned treatment paradigms are mediated via gene expression alterations such as downregulation of inflammatory and death-signal genes and upregulation of neuroprotective and cytoprotective genes. These varied techniques and technologies will be described and discussed covering cell-based and animal model-based studies. Data from clinical applications and linkage to human ocular diseases will also be discussed where relevant translational research has been reported.
Collapse
Affiliation(s)
- Najam A Sharif
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, California, USA
- Singapore Eye Research Institute (SERI), Singapore
- SingHealth Duke-NUS Ophthalmology and Visual Sciences Academic Clinical Programme, Duke-National University of Singapore Medical School, Singapore
- Department of Surgery and Cancer, Imperial College of Science and Technology, London, United Kingdom
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center, Fort Worth, Texas, USA
- Department of Pharmacy Sciences, Creighton University, Omaha, Nebraska, USA
- Insitute of Ophthalmology, University College London (UCL), London, United Kingdom
| |
Collapse
|
14
|
Morales-Hernandez AG, Martinez-Aguilar V, Chavez-Gonzalez TM, Mendez-Avila JC, Frias-Becerril JV, Morales-Hernandez LA, Cruz-Albarran IA. Short-Term Thermal Effect of Continuous Ultrasound from 3 MHz to 1 and 0.5 W/cm 2 Applied to Gastrocnemius Muscle. Diagnostics (Basel) 2023; 13:2644. [PMID: 37627903 PMCID: PMC10453025 DOI: 10.3390/diagnostics13162644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Continuous ultrasound is recognized for its thermal effect and use in the tissue repair process. However, there is controversy about its dosage and efficacy. This study used infrared thermography, a non-invasive technique, to measure the short-term thermal effect of 3 MHz continuous ultrasound vs. a placebo, referencing the intensity applied. It was a single-blind, randomized clinical trial of 60 healthy volunteers (19-24 years old) divided into three equal groups. Group 1:1 W/cm2 for 5 min; Group 2: 0.5 W/cm2 for 10 min; and Group 3: the placebo for 5 min. The temperature was recorded through five thermographic images per patient: pre- and post-application, 5, 10, and 15 min later. After statistical analysis, a more significant decrease in temperature (p<0.05 ) was observed in the placebo group compared with the remaining groups after the application of continuous ultrasound. Group 1 was the one that generated the highest significant thermal effect (p<0.001), with an increase of 3.05 °C at 15 min, compared with the other two groups. It is concluded that to generate a thermal effect in the muscle, intensities of ≥1 W/cm2 are required, since the dosage maintained a temperature increase for more than 5 min.
Collapse
Affiliation(s)
- Arely G. Morales-Hernandez
- Faculty of Nursing, Autonomous University of Queretaro, Queretaro 76010, Mexico
- Education, Movement and Health, Faculty of Nursing, Autonomous University of Queretaro, Queretaro 76010, Mexico
| | - Violeta Martinez-Aguilar
- Faculty of Nursing, Autonomous University of Queretaro, Campus Corregidora, Queretaro 76912, Mexico
| | | | - Julio C. Mendez-Avila
- Faculty of Nursing, Autonomous University of Queretaro, Queretaro 76010, Mexico
- Education, Movement and Health, Faculty of Nursing, Autonomous University of Queretaro, Queretaro 76010, Mexico
| | | | - Luis A. Morales-Hernandez
- Laboratory of Artificial Vision and Thermography/Mechatronics, Faculty of Engineering, Autonomous University of Queretaro, Campus San Juan del Rio, San Juan del Río 76807, Mexico
| | - Irving A. Cruz-Albarran
- Faculty of Nursing, Autonomous University of Queretaro, Queretaro 76010, Mexico
- Laboratory of Artificial Vision and Thermography/Mechatronics, Faculty of Engineering, Autonomous University of Queretaro, Campus San Juan del Rio, San Juan del Río 76807, Mexico
- Artificial Intelligence Systems Applied to Biomedical and Mechanical Models, Faculty of Engineering, Autonomus University of Queretaro, Campus San Juan del Rio, San Juan del Rio 76807, Mexico
| |
Collapse
|
15
|
Riis Porsborg S, Krzyslak H, Pierchala MK, Trolé V, Astafiev K, Lou-Moeller R, Pennisi CP. Exploring the Potential of Ultrasound Therapy to Reduce Skin Scars: An In Vitro Study Using a Multi-Well Device Based on Printable Piezoelectric Transducers. Bioengineering (Basel) 2023; 10:bioengineering10050566. [PMID: 37237636 DOI: 10.3390/bioengineering10050566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/24/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Excessive skin scarring affects over 100 million patients worldwide, with effects ranging from cosmetic to systemic problems, and an effective treatment is yet to be found. Ultrasound-based therapies have been used to treat a variety of skin disorders, but the exact mechanisms behind the observed effects are still unclear. The aim of this work was to demonstrate the potential of ultrasound for the treatment of abnormal scarring by developing a multi-well device based on printable piezoelectric material (PiezoPaint™). First, compatibility with cell cultures was evaluated using measurements of heat shock response and cell viability. Second, the multi-well device was used to treat human fibroblasts with ultrasound and quantify their proliferation, focal adhesions, and extracellular matrix (ECM) production. Ultrasound caused a significant reduction in fibroblast growth and ECM deposition without changes in cell viability or adhesion. The data suggest that these effects were mediated by nonthermal mechanisms. Interestingly, the overall results suggest that ultrasound treatment would a be beneficial therapy for scar reduction. In addition, it is expected that this device will be a useful tool for mapping the effects of ultrasound treatment on cultured cells.
Collapse
Affiliation(s)
- Simone Riis Porsborg
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, DK-9260 Gistrup, Denmark
| | - Hubert Krzyslak
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, DK-9260 Gistrup, Denmark
| | | | - Vincent Trolé
- CTS Ferroperm Piezoceramics, DK-3490 Kvistgaard, Denmark
| | | | | | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, DK-9260 Gistrup, Denmark
| |
Collapse
|
16
|
Chen Y, Yang H, Wang Z, Zhu R, Cheng L, Cheng Q. Low-intensity pulsed ultrasound promotes mesenchymal stem cell transplantation-based articular cartilage regeneration via inhibiting the TNF signaling pathway. Stem Cell Res Ther 2023; 14:93. [PMID: 37069673 PMCID: PMC10111837 DOI: 10.1186/s13287-023-03296-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 03/22/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) transplantation therapy is highly investigated for the regenerative repair of cartilage defects. Low-intensity pulsed ultrasound (LIPUS) has the potential to promote chondrogenic differentiation of MSCs. However, its underlying mechanism remains unclear. Here, we investigated the promoting effects and mechanisms underlying LIPUS stimulation on the chondrogenic differentiation of human umbilical cord mesenchymal stem cells (hUC-MSCs) and further evaluated its regenerative application value in articular cartilage defects in rats. METHODS LIPUS was applied to stimulate cultured hUC-MSCs and C28/I2 cells in vitro. Immunofluorescence staining, qPCR analysis, and transcriptome sequencing were used to detect mature cartilage-related markers of gene and protein expression for a comprehensive evaluation of differentiation. Injured articular cartilage rat models were established for further hUC-MSC transplantation and LIPUS stimulation in vivo. Histopathology and H&E staining were used to evaluate the repair effects of the injured articular cartilage with LIPUS stimulation. RESULTS The results showed that LIPUS stimulation with specific parameters effectively promoted the expression of mature cartilage-related genes and proteins, inhibited TNF-α gene expression in hUC-MSCs, and exhibited anti-inflammation in C28/I2 cells. In addition, the articular cartilage defects of rats were significantly repaired after hUC-MSC transplantation and LIPUS stimulation. CONCLUSIONS Taken together, LIPUS stimulation could realize articular cartilage regeneration based on hUC-MSC transplantation due to the inhibition of the TNF signaling pathway, which is of clinical value for the relief of osteoarthritis.
Collapse
Affiliation(s)
- Yiming Chen
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Huiyi Yang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zhaojie Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
- School of Life Science and Technology, Tongji University, Shanghai, 200065, China
| | - Rongrong Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
- School of Life Science and Technology, Tongji University, Shanghai, 200065, China
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Qian Cheng
- Institute of Acoustics, School of Physics Science and Engineering, Tongji University, Shanghai, 200092, China.
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University School of Medicine, Tongji University, Shanghai, 200065, China.
- Frontiers Science Center for Intelligent Autonomous Systems, Shanghai, 201210, China.
| |
Collapse
|
17
|
Carr BJ. Regenerative Medicine and Rehabilitation Therapy in the Canine. Vet Clin North Am Small Anim Pract 2023; 53:801-827. [PMID: 36997410 DOI: 10.1016/j.cvsm.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Regenerative medicine is used in the canine to optimize tissue healing and treat osteoarthritis and soft tissue injuries. Rehabilitation therapy is also often implemented in the treatment and management of musculoskeletal conditions in the canine. Initial experimental studies have shown that regenerative medicine and rehabilitation therapy may work safely and synergistically to enhance tissue healing. Although additional study is required to define optional rehabilitation therapy protocols after regenerative medicine therapy in the canine, certain fundamental principles of rehabilitation therapy still apply to patients treated with regenerative medicine.
Collapse
|
18
|
Priming Effects of Anodal Transcranial Direct Current Stimulation on the Effects of Conventional Physiotherapy on Balance and Muscle Performance in Athletes With Anterior Cruciate Ligament Injury. J Sport Rehabil 2023; 32:315-324. [PMID: 36623509 DOI: 10.1123/jsr.2022-0188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 10/02/2022] [Accepted: 11/07/2022] [Indexed: 01/11/2023]
Abstract
CONTEXT In athletes, postural control impairment and knee muscle dysfunction are the most common disorders following anterior cruciate ligament (ACL) injury. Because of functional changes in the motor cortex following ACL injury, physiotherapy (PT) is not enough for treatment and using neuromodulators, such as trans-cranial direct current stimulation (tDCS) may be necessary. The present study focused on the effects of anodal tDCS (a-tDCS) over the primary motor cortex (M1) concurrent with PT on postural control and muscular performance in the athletes with ACL injury. DESIGN In this study, 34 athletes with ACL injury were randomly assigned in 2 groups of intervention group (active M1 a-tDCS concurrent with PT, n = 16) and control group (sham M1 a-tDCS concurrent with PT, n = 16). METHODS The participants of all groups received 20-minute 2 mA M1 a-tDCS with PT during 10 sessions, while tDCS was turned off after 30 seconds in the sham group. Before, immediately following, and 1 month after the interventions, the center of pressure and the average of power of flexor and extensor muscles at 2 velocities of 30°/s and 60°/s were measured by force plate and isokinetic devices, respectively. RESULTS One month after treatment, the displacement of center of pressure was decreased in the intervention group (P < .05), while there were no changes in the control group. Y-axis of center of pressure decreased in the intervention group relative to the control group, although average of power of flexor and extensor muscles increased immediately in both groups, but the rise in the intervention group was larger than that in the control group (P < .05). CONCLUSION The findings indicated that M1 a-tDCS can induce the efficacy of PT, which has a lasting effect on the improvement of the postural control in athletes with ACL injury.
Collapse
|
19
|
Bakhshandeh B, Ranjbar N, Abbasi A, Amiri E, Abedi A, Mehrabi M, Dehghani Z, Pennisi CP. Recent progress in the manipulation of biochemical and biophysical cues for engineering functional tissues. Bioeng Transl Med 2023; 8:e10383. [PMID: 36925674 PMCID: PMC10013802 DOI: 10.1002/btm2.10383] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/28/2022] [Accepted: 07/16/2022] [Indexed: 11/11/2022] Open
Abstract
Tissue engineering (TE) is currently considered a cutting-edge discipline that offers the potential for developing treatments for health conditions that negatively affect the quality of life. This interdisciplinary field typically involves the combination of cells, scaffolds, and appropriate induction factors for the regeneration and repair of damaged tissue. Cell fate decisions, such as survival, proliferation, or differentiation, critically depend on various biochemical and biophysical factors provided by the extracellular environment during developmental, physiological, and pathological processes. Therefore, understanding the mechanisms of action of these factors is critical to accurately mimic the complex architecture of the extracellular environment of living tissues and improve the efficiency of TE approaches. In this review, we recapitulate the effects that biochemical and biophysical induction factors have on various aspects of cell fate. While the role of biochemical factors, such as growth factors, small molecules, extracellular matrix (ECM) components, and cytokines, has been extensively studied in the context of TE applications, it is only recently that we have begun to understand the effects of biophysical signals such as surface topography, mechanical, and electrical signals. These biophysical cues could provide a more robust set of stimuli to manipulate cell signaling pathways during the formation of the engineered tissue. Furthermore, the simultaneous application of different types of signals appears to elicit synergistic responses that are likely to improve functional outcomes, which could help translate results into successful clinical therapies in the future.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Nika Ranjbar
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Ardeshir Abbasi
- Department of Immunology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Elahe Amiri
- Department of Life Science Engineering, Faculty of New Sciences and TechnologyUniversity of TehranTehranIran
| | - Ali Abedi
- Department of Life Science Engineering, Faculty of New Sciences and TechnologyUniversity of TehranTehranIran
| | - Mohammad‐Reza Mehrabi
- Department of Microbial Biotechnology, School of Biology, College of ScienceUniversity of TehranTehranIran
| | - Zahra Dehghani
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and TechnologyAalborg UniversityAalborgDenmark
| |
Collapse
|
20
|
Aimaijiang M, Liu Y, Zhang Z, Qin Q, Liu M, Abulikemu P, Liu L, Zhou Y. LIPUS as a potential strategy for periodontitis treatment: A review of the mechanisms. Front Bioeng Biotechnol 2023; 11:1018012. [PMID: 36911184 PMCID: PMC9992218 DOI: 10.3389/fbioe.2023.1018012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Periodontitis is a chronic inflammatory condition triggered by oral bacteria. A sustained inflammatory state in periodontitis could eventually destroy the alveolar bone. The key objective of periodontal therapy is to terminate the inflammatory process and reconstruct the periodontal tissues. The traditional Guided tissue regeneration (GTR) procedure has unstable results due to multiple factors such as the inflammatory environment, the immune response caused by the implant, and the operator's technique. Low-intensity pulsed ultrasound (LIPUS), as acoustic energy, transmits the mechanical signals to the target tissue to provide non-invasive physical stimulation. LIPUS has positive effects in promoting bone regeneration, soft-tissue regeneration, inflammation inhibition, and neuromodulation. LIPUS can maintain and regenerate alveolar bone during an inflammatory state by suppressing the expression of inflammatory factors. LIPUS also affects the cellular behavior of periodontal ligament cells (PDLCs), thereby protecting the regenerative potential of bone tissue in an inflammatory state. However, the underlying mechanisms of the LIPUS therapy are still yet to be summarized. The goal of this review is to outline the potential cellular and molecular mechanisms of periodontitis-related LIPUS therapy, as well as to explain how LIPUS manages to transmit mechanical stimulation into the signaling pathway to achieve inflammatory control and periodontal bone regeneration.
Collapse
Affiliation(s)
- Maierhaba Aimaijiang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yiping Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhiying Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Qiuyue Qin
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Manxuan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Palizi Abulikemu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Lijun Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
21
|
Effect of Therapeutic Ultrasound on the Mechanical and Biological Properties of Fibroblasts. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00281-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose
This paper explores the effect of therapeutic ultrasound on the mechanical and biological properties of ligament fibroblasts.
Methods and Results
We assessed pulsed ultrasound doses of 1.0 and 2.0 W/cm2 at 1 MHz frequency for five days on ligament fibroblasts using a multidisciplinary approach. Atomic force microscopy showed a decrease in cell elastic modulus for both doses, but the treated cells were still viable based on flow cytometry. Finite element method analysis exhibited visible cytoskeleton displacements and decreased harmonics in treated cells. Colorimetric assay revealed increased cell proliferation, while scratch assay showed increased migration at a low dose. Enzyme-linked immunoassay detected increased collagen and fibronectin at a high dose, and immunofluorescence imaging technique visualized β-actin expression for both treatments.
Conclusion
Both doses of ultrasound altered the fibroblast mechanical properties due to cytoskeletal reorganization and enhanced the regenerative and remodeling stages of cell repair.
Lay Summary
Knee ligament injuries are a lesion of the musculoskeletal system frequently diagnosed in active and sedentary lifestyles in young and older populations. Therapeutic ultrasound is a rehabilitation strategy that may lead to the regenerative and remodeling of ligament wound healing. This research demonstrated that pulsed therapeutic ultrasound applied for 5 days reorganized the ligament fibroblasts structure to increase the cell proliferation and migration at a low dose and to increase the releasing proteins that give the stiffness of the healed ligament at a high dose.
Future Works
Future research should further develop and confirm that therapeutic ultrasound may improve the regenerative and remodeling stages of the ligament healing process applied in clinical trials in active and sedentary lifestyles in young and older populations.
Graphical abstract
Collapse
|
22
|
Chen H, Li Z, Li X, Yang Y, Dai Y, Xie Z, Xiao J, Liu X, Yang L, Shi C, Zhi E, Tian R, Li P, Chen H, Zhao F, Hu J, Yao C, Lin G, Lue TF, Xia S. The Efficacy and Safety of Thrice vs Twice per Week Low-Intensity Pulsed Ultrasound Therapy for Erectile Dysfunction: A Randomized Clinical Trial. J Sex Med 2022; 19:1536-1545. [PMID: 35999130 DOI: 10.1016/j.jsxm.2022.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/28/2022] [Accepted: 06/13/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND A recent sham-controlled clinical study has shown that low-intensity pulsed ultrasound twice per week can safely and effectively treat patients with mild-to-moderate erectile dysfunction (ED). However, large-scale clinical trials are needed to verify its efficacy and safety and determine a reasonable treatment interval. AIM To study whether low-intensity pulsed ultrasound therapy thrice per week is non-inferior to twice per week in patients with mild-to-moderate ED. METHODS A randomized, open-label, parallel-group, non-inferiority clinical trial was conducted in 7 hospitals in China. A total of 323 patients with mild-to-moderate ED were randomized (1:1) into thrice per week (3/W) and twice per week (2/W) groups. Low-intensity pulsed ultrasound was applied on each side of the penis for 16 sessions. OUTCOMES The primary outcome was response rate using the minimal clinically important difference in the International Index of Erectile Function (IIEF-EF) score at week 12. Secondary outcomes included Erection Hardness Score (EHS), Sexual Encounter Profile, Global Assessment Question, and Self Esteem and Relationship Questionnaire. RESULTS Response rates in 3/W and 2/W groups were 62.0% and 62.5%, respectively. Treatment effect in the 3/W group was noninferior to that of the 2/W group, with rate difference lower bound of -0.01% [95% confidence interval -0.11 to 0.10%] within the acceptable margin (-14.0%). No significant difference was found among secondary outcomes. IIEF-EF score showed a significant increase from baseline in the 3/W group (16.8 to 20.7) and 2/W group (17.8 to 21.7), and the percentage of patients with EHS ≥3 increased in the 3/W (54.9% to 84.0%) and 2/W (59.5% to 83.5%) groups. There was no significant difference in response rate between the 2 groups after controlling for strata factors and homogeneous tests. No treatment-related adverse events were reported. CLINICAL IMPLICATIONS Low-intensity pulsed ultrasound therapy displays similar efficacy and safety for mild-to-moderate ED when administered thrice or twice per week for 16 sessions. This study provides two options to suit patients' needs. STRENGTHS & LIMITATIONS This is a large-sample, randomized, controlled, noninferiority trial study. Short-term follow-up and mostly younger patients are the main limitations. CONCLUSION Low-intensity pulsed ultrasound therapy thrice and twice per week showed equivalent therapeutic effects and safety for mild-to-moderate ED in a young and generally healthy population. This therapy warrants further investigation of its potential value in rehabilitation of ED.
Collapse
|
23
|
Furusawa Y, Kondo T, Tachibana K, Feril LB. Ultrasound-Induced DNA Damage and Cellular Response: Historical Review, Mechanisms Analysis, and Therapeutic Implications. Radiat Res 2022; 197:662-672. [PMID: 35275998 DOI: 10.1667/rade-21-00140.1.s1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/22/2022] [Indexed: 11/03/2022]
Abstract
The biological effects of ultrasound may be classified into thermal and nonthermal mechanisms. The nonthermal effects may be further classified into cavitational and noncavitational mechanisms. DNA damage induced by ultrasound is considered to be related to nonthermal cavitations. For this aspect, many in vitro studies on DNA have been conducted for evaluating the safety of diagnostic ultrasound, particularly in fetal imaging. Technological advancement in detecting DNA damage both in vitro and in vivo have elucidated the mechanism of DNA damage formation and their cellular response. Damage to DNA, and the residual damages after DNA repair are implicated in the biological effects. Here, we discuss the historical evidence of ultrasound on DNA damage and the mechanism of DNA damage formation both in vitro and in vivo, compared with those induced by ionizing radiation. We also offer a commentary on the safety of ultrasound over X-ray-based imaging. Also, understanding the various mechanisms involved in the bioeffects of ultrasound will lead us to alternative strategies for use of ultrasound for therapy.
Collapse
Affiliation(s)
- Yukihiro Furusawa
- Department of Liberal Arts and Sciences, Toyama Prefecture University, Toyama 939-0398, Japan
| | - Takashi Kondo
- Department of Radiological Sciences Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Katsuro Tachibana
- Department of Anatomy. Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Loreto B Feril
- Department of Anatomy. Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| |
Collapse
|
24
|
Acheta J, Stephens SBZ, Belin S, Poitelon Y. Therapeutic Low-Intensity Ultrasound for Peripheral Nerve Regeneration – A Schwann Cell Perspective. Front Cell Neurosci 2022; 15:812588. [PMID: 35069118 PMCID: PMC8766802 DOI: 10.3389/fncel.2021.812588] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/09/2021] [Indexed: 01/22/2023] Open
Abstract
Peripheral nerve injuries are common conditions that can arise from trauma (e.g., compression, severance) and can lead to neuropathic pain as well as motor and sensory deficits. Although much knowledge exists on the mechanisms of injury and nerve regeneration, treatments that ensure functional recovery following peripheral nerve injury are limited. Schwann cells, the supporting glial cells in peripheral nerves, orchestrate the response to nerve injury, by converting to a “repair” phenotype. However, nerve regeneration is often suboptimal in humans as the repair Schwann cells do not sustain their repair phenotype long enough to support the prolonged regeneration times required for successful nerve regrowth. Thus, numerous strategies are currently focused on promoting and extending the Schwann cells repair phenotype. Low-intensity ultrasound (LIU) is a non-destructive therapeutic approach which has been shown to facilitate peripheral nerve regeneration following nerve injury in rodents. Still, clinical trials in humans are scarce and limited to small population sizes. The benefit of LIU on nerve regeneration could possibly be mediated through the repair Schwann cells. In this review, we discuss the known and possible molecular mechanisms activated in response to LIU in repair Schwann cells to draw support and attention to LIU as a compelling regenerative treatment for peripheral nerve injury.
Collapse
|
25
|
Luo L, Wu Z, Wang Y, Li H. Regulating the production and biological function of small extracellular vesicles: current strategies, applications and prospects. J Nanobiotechnology 2021; 19:422. [PMID: 34906146 PMCID: PMC8670141 DOI: 10.1186/s12951-021-01171-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 11/30/2021] [Indexed: 02/08/2023] Open
Abstract
Numerous studies have confirmed the great application potentials of small extracellular vesicles (sEVs) in biological medical field, especially in tissue repair and regeneration. However, the production capability of sEVs by noncancerous cells is very limited, while their dosage requirements in disease treatments are usually very high. Meanwhile, as cell aging, the sEV production capability of cells decreases and the biological function of sEVs changes accordingly. In addition, for special applications, sEVs carrying desired bioactive substances should be designed to perform their expected biological function. Therefore, improving the production of sEVs and precisely regulating their biological function are of great significance for promoting the clinical applications of sEVs. In this review, some of the current classic strategies in affecting the cellular behaviors of donor cells and subsequently regulating the production and biological function of their sEVs are summarized, including gene engineering methods, stress-inducing conditions, chemical regulators, physical methods, and biomaterial stimulations. Through applying these strategies, increased yield of sEVs with required biological function can be obtained for disease treatment and tissue repair, such as bone regeneration, wound healing, nerve function recovery and cancer treatment, which could not only reduce the harvest cost of sEV but promote the practical applications of sEVs in clinic.
Collapse
Affiliation(s)
- Lei Luo
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, 200030, China
| | - Zhi Wu
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China
| | - Yang Wang
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai, 200233, China.
| | - Haiyan Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, 200030, China.
- Chemical and Environmental Engineering Department, School of Engineering, RMIT University, 124 La Trobe St, Melbourne, VIC, 3001, Australia.
| |
Collapse
|
26
|
Hao Z, Xu Z, Wang X, Wang Y, Li H, Chen T, Hu Y, Chen R, Huang K, Chen C, Li J. Biophysical Stimuli as the Fourth Pillar of Bone Tissue Engineering. Front Cell Dev Biol 2021; 9:790050. [PMID: 34858997 PMCID: PMC8630705 DOI: 10.3389/fcell.2021.790050] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/26/2021] [Indexed: 01/12/2023] Open
Abstract
The repair of critical bone defects remains challenging worldwide. Three canonical pillars (biomaterial scaffolds, bioactive molecules, and stem cells) of bone tissue engineering have been widely used for bone regeneration in separate or combined strategies, but the delivery of bioactive molecules has several obvious drawbacks. Biophysical stimuli have great potential to become the fourth pillar of bone tissue engineering, which can be categorized into three groups depending on their physical properties: internal structural stimuli, external mechanical stimuli, and electromagnetic stimuli. In this review, distinctive biophysical stimuli coupled with their osteoinductive windows or parameters are initially presented to induce the osteogenesis of mesenchymal stem cells (MSCs). Then, osteoinductive mechanisms of biophysical transduction (a combination of mechanotransduction and electrocoupling) are reviewed to direct the osteogenic differentiation of MSCs. These mechanisms include biophysical sensing, transmission, and regulation. Furthermore, distinctive application strategies of biophysical stimuli are presented for bone tissue engineering, including predesigned biomaterials, tissue-engineered bone grafts, and postoperative biophysical stimuli loading strategies. Finally, ongoing challenges and future perspectives are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhenhua Xu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuan Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yi Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hanke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yingkun Hu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Renxin Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kegang Huang
- Wuhan Institute of Proactive Health Management Science, Wuhan, China
| | - Chao Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Orthopedics, Hefeng Central Hospital, Enshi, China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
27
|
Ultrasound-Enabled Therapeutic Delivery and Regenerative Medicine: Physical and Biological Perspectives. ACS Biomater Sci Eng 2021; 7:4371-4387. [PMID: 34460238 DOI: 10.1021/acsbiomaterials.1c00276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The role of ultrasound in medicine and biological sciences is expanding rapidly beyond its use in conventional diagnostic imaging. Numerous studies have reported the effects of ultrasound on cellular and tissue physiology. Advances in instrumentation and electronics have enabled successful in vivo applications of therapeutic ultrasound. Despite path breaking advances in understanding the biophysical and biological mechanisms at both microscopic and macroscopic scales, there remain substantial gaps. With the progression of research in this area, it is important to take stock of the current understanding of the field and to highlight important areas for future work. We present herein key developments in the biological applications of ultrasound especially in the context of nanoparticle delivery, drug delivery, and regenerative medicine. We conclude with a brief perspective on the current promise, limitations, and future directions for interfacing ultrasound technology with biological systems, which could provide guidance for future investigations in this interdisciplinary area.
Collapse
|
28
|
Cafarelli A, Marino A, Vannozzi L, Puigmartí-Luis J, Pané S, Ciofani G, Ricotti L. Piezoelectric Nanomaterials Activated by Ultrasound: The Pathway from Discovery to Future Clinical Adoption. ACS NANO 2021; 15:11066-11086. [PMID: 34251189 PMCID: PMC8397402 DOI: 10.1021/acsnano.1c03087] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/06/2021] [Indexed: 05/19/2023]
Abstract
Electrical stimulation has shown great promise in biomedical applications, such as regenerative medicine, neuromodulation, and cancer treatment. Yet, the use of electrical end effectors such as electrodes requires connectors and batteries, which dramatically hamper the translation of electrical stimulation technologies in several scenarios. Piezoelectric nanomaterials can overcome the limitations of current electrical stimulation procedures as they can be wirelessly activated by external energy sources such as ultrasound. Wireless electrical stimulation mediated by piezoelectric nanoarchitectures constitutes an innovative paradigm enabling the induction of electrical cues within the body in a localized, wireless, and minimally invasive fashion. In this review, we highlight the fundamental mechanisms of acoustically mediated piezoelectric stimulation and its applications in the biomedical area. Yet, the adoption of this technology in a clinical practice is in its infancy, as several open issues, such as piezoelectric properties measurement, control of the ultrasound dose in vitro, modeling and measurement of the piezo effects, knowledge on the triggered bioeffects, therapy targeting, biocompatibility studies, and control of the ultrasound dose delivered in vivo, must be addressed. This article explores the current open challenges in piezoelectric stimulation and proposes strategies that may guide future research efforts in this field toward the translation of this technology to the clinical scene.
Collapse
Affiliation(s)
- Andrea Cafarelli
- The
BioRobotics Institute, Scuola Superiore
Sant’Anna, 56127 Pisa, Italy
- Department
of Excellence in Robotics & AI, Scuola
Superiore Sant’Anna, 56127 Pisa, Italy
| | - Attilio Marino
- Smart
Bio-Interfaces, Istituto Italiano di Tecnologia, 56025 Pontedera, Italy
| | - Lorenzo Vannozzi
- The
BioRobotics Institute, Scuola Superiore
Sant’Anna, 56127 Pisa, Italy
- Department
of Excellence in Robotics & AI, Scuola
Superiore Sant’Anna, 56127 Pisa, Italy
| | - Josep Puigmartí-Luis
- Departament
de Ciència dels Materials i Química Física, Institut de Química Teòrica i Computacional, 08028 Barcelona, Spain
- Institució
Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Salvador Pané
- Multi-Scale
Robotics Lab (MSRL), Institute of Robotics and Intelligent Systems
(IRIS), ETH Zurich, 8092 Zurich, Switzerland
| | - Gianni Ciofani
- Smart
Bio-Interfaces, Istituto Italiano di Tecnologia, 56025 Pontedera, Italy
| | - Leonardo Ricotti
- The
BioRobotics Institute, Scuola Superiore
Sant’Anna, 56127 Pisa, Italy
- Department
of Excellence in Robotics & AI, Scuola
Superiore Sant’Anna, 56127 Pisa, Italy
- Tel: +39 050 883074. Mobile: +39 366 6868242.
| |
Collapse
|
29
|
Uddin SMZ, Komatsu DE, Motyka T, Petterson S. Low-Intensity Continuous Ultrasound Therapies—A Systematic Review of Current State-of-the-Art and Future Perspectives. J Clin Med 2021; 10:2698. [PMID: 34207333 PMCID: PMC8235587 DOI: 10.3390/jcm10122698] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 01/02/2023] Open
Abstract
Therapeutic ultrasound has been studied for over seven decades for different medical applications. The versatility of ultrasound applications are highly dependent on the frequency, intensity, duration, duty cycle, power, wavelength, and form. In this review article, we will focus on low-intensity continuous ultrasound (LICUS). LICUS has been well-studied for numerous clinical disorders, including tissue regeneration, pain management, neuromodulation, thrombosis, and cancer treatment. PubMed and Google Scholar databases were used to conduct a comprehensive review of all research studying the application of LICUS in pre-clinical and clinical studies. The review includes articles that specify intensity and duty cycle (continuous). Any studies that did not identify these parameters or used high-intensity and pulsed ultrasound were not included in the review. The literature review shows the vast implication of LICUS in many medical fields at the pre-clinical and clinical levels. Its applications depend on variables such as frequency, intensity, duration, and type of medical disorder. Overall, these studies show that LICUS has significant promise, but conflicting data remain regarding the parameters used, and further studies are required to fully realize the potential benefits of LICUS.
Collapse
Affiliation(s)
- Sardar M. Z. Uddin
- Department of Orthopaedics and Rehabilitation, Stony Brook University, Stony Brook, NY 11794, USA;
| | - David E. Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Thomas Motyka
- Department of Osteopathic Manipulative Medicine, Campbell University, Buies Creek, NC 27506, USA;
| | | |
Collapse
|
30
|
de Lucas B, Pérez LM, Bernal A, Gálvez BG. Application of low-intensity pulsed therapeutic ultrasound on mesenchymal precursors does not affect their cell properties. PLoS One 2021; 16:e0246261. [PMID: 33571276 PMCID: PMC7877602 DOI: 10.1371/journal.pone.0246261] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Ultrasound is considered a safe and non-invasive tool in regenerative medicine and has been used in the clinic for more than twenty years for applications in bone healing after the approval of the Exogen device, also known as low-intensity pulsed ultrasound (LIPUS). Beyond its effects on bone health, LIPUS has also been investigated for wound healing of soft tissues, with positive results for various cell processes including cell proliferation, migration and angiogenesis. As LIPUS has the potential to treat chronic skin wounds, we sought to evaluate the effects produced by a conventional therapeutic ultrasound device at low intensities (also considered LIPUS) on the migration capacity of mouse and human skin mesenchymal precursors (s-MPs). Cells were stimulated for 3 days (20 minutes per day) using a traditional ultrasound device with the following parameters: 100 mW/cm2 with 20% duty cycle and frequency of 3 MHz. At the parameters used, ultrasound failed to affect s-MP proliferation, with no evident changes in morphology or cell groupings, and no changes at the cytoskeletal level. Further, the migration and invasion ability of s-MPs were unaffected by the ultrasound protocol, and no major changes were detected in the gene/protein expression of ROCK1, integrin β1, laminin β1, type I collagen and transforming growth factor β1. Finally, RNA-seq analysis revealed that only 10 genes were differentially expressed after ultrasound stimulation. Among them, 5 encode for small nuclear RNAs and 2 encode for proteins belonging to the nuclear pore complex. Considering the results overall, while the viability of s-MPs was not affected by ultrasound stimulation and no changes were detected in proliferation/migration, RNA-seq analysis would suggest that s-MPs do respond to ultrasound. The use of 100 mW/cm2 intensity or conventional therapeutic ultrasound devices might not be optimal for the stimulation the properties of cell populations. Future studies should investigate the potential application of ultrasound using variations of the tested parameters.
Collapse
Affiliation(s)
- Beatriz de Lucas
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Laura M. Pérez
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Aurora Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz G. Gálvez
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| |
Collapse
|
31
|
Tabuchi Y, Hasegawa H, Suzuki N, Furusawa Y, Hirano T, Nagaoka R, Hirayama J, Hoshi N, Mochizuki T. Genetic response to low‑intensity ultrasound on mouse ST2 bone marrow stromal cells. Mol Med Rep 2021; 23:173. [PMID: 33398373 PMCID: PMC7821223 DOI: 10.3892/mmr.2020.11812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 10/27/2020] [Indexed: 11/05/2022] Open
Abstract
Although low‑intensity ultrasound (LIUS) is a clinically established procedure, the early cellular effect of LIUS on a genetic level has not yet been studied. The current study investigated the early response genes elicited by LIUS in bone marrow stromal cells (BMSCs) using global‑scale microarrays and computational gene expression analysis tools. Mouse ST2 BMSCs were treated with LIUS [ISATA, 25 mW/cm2 for 20 min with a frequency of 1.11 MHz in a pulsed‑wave mode (0.2‑s burst sine waves repeated at 1 kHz)], then cultured for 0.5, 1 and 3 h at 37˚C. The time course of changes in gene expression was evaluated using GeneChip® high‑density oligonucleotide microarrays and Ingenuity® Pathway Analysis tools. The results were verified by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). A single exposure of LIUS did not affect cell morphology, cell growth or alkaline phosphatase activity. However, 61 upregulated and 103 downregulated genes were identified from 0.5 to 3 h after LIUS treatment. Two significant gene networks, labeled E and H, were identified from the upregulated genes, while a third network, labeled T, was identified from the downregulated genes. Gene network E or H containing the immediate‑early genes FBJ osteosarcoma oncogene and early growth response 1 or the heat shock proteins heat shock protein 1a/b was associated mainly with the biological functions of bone physiology and protein folding or apoptosis, respectively. Gene network T containing transcription factors fos‑like antigen 1 and serum response factor was also associated with the biological functions of the gene expression. RT‑qPCR indicated that the expression of several genes in the gene networks E and H were elevated in LIUS‑treated cells. LIUS was demonstrated to induce gene expression after short application in mouse ST2 BMSCs. The results of the present study provide a basis for the elucidation of the detailed molecular mechanisms underlying the cellular effects of LIUS.
Collapse
Affiliation(s)
- Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Toyama 930-0194, Japan
| | - Hideyuki Hasegawa
- Graduate School of Science and Engineering, University of Toyama, Toyama 930‑8555, Japan
| | - Nobuo Suzuki
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Ishikawa 927‑0553, Japan
| | - Yukihiro Furusawa
- Department of Liberal Arts and Sciences, Toyama Prefectural University, Toyama 939-0398, Japan
| | - Tetsushi Hirano
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Toyama 930-0194, Japan
| | - Ryo Nagaoka
- Graduate School of Science and Engineering, University of Toyama, Toyama 930‑8555, Japan
| | - Jun Hirayama
- Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, Komatsu 923‑0961, Japan
| | - Nobuhiko Hoshi
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe 657‑8501, Japan
| | | |
Collapse
|