1
|
Liang W, Chi Y, Liang B, Fu G, Yan K, Zhang G, Cai Y. Low-frequency ultrasound alleviates pulmonary inflammation induced by Klebsiella pneumoniae in mice by inhibiting the TNFR1/NF-κB pathway. Int Immunopharmacol 2025; 154:114574. [PMID: 40188523 DOI: 10.1016/j.intimp.2025.114574] [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: 01/14/2025] [Revised: 03/18/2025] [Accepted: 03/27/2025] [Indexed: 04/08/2025]
Abstract
BACKGROUND Therapeutic ultrasound has been found to promote tissue healing and reduce inflammation in non-infectious diseases, but its efficacy in infectious inflammation remains unclear. Here, we employ the mice pneumonia model to explore the anti-inflammatory effects of low-frequency ultrasound (LFU) and elucidate its potential molecular mechanisms. METHODS Pneumonia in mice was induced by intratracheal instillation of 100 μL of a 4.5 × 108 CFU/mL Klebsiella pneumoniae (Kp) bacterial suspension. A single LFU treatment (29.36 kHz, 270 mW/cm2, 10 min) was applied to the chest of mice at 6 or 48 h after infection. Biological samples were collected for gene, protein, and cellular experiments. RESULTS LFU demonstrated good anti-inflammatory effects in mice during the recovery phase of Kp infection (48 h after infection). Although LFU alone had no bactericidal effects, it slightly reduced the pathological score of lung injury and significantly decreased the infiltration of CD45+ leukocytes. Additionally, the protein levels of TNF-α, GM-CSF and COX-2 in the bronchoalveolar lavage fluid were significantly reduced. Bulk RNA-sequencing results showed that the TNF receptor (TNFR)/NF-κB pathway was up-regulated after Kp infection, which was suppressed after LFU treatment. Western blot and immunofluorescence revealed LFU significantly reduced the protein levels of TNFR1, p-p65, and nuclear p65. The anti-inflammatory effect of LFU was comparable to a 20 mg/kg NF-κB inhibitor and superior to a 15 mg/kg TNFR antagonist. CONCLUSION LFU exerts anti-inflammatory effects by inhibiting the TNFR1/NF-κB pathway during the recovery period of Kp infection, reducing inflammatory transcription and thereby decreasing the release of inflammatory factors.
Collapse
Affiliation(s)
- Wenxin Liang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Yulong Chi
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Beibei Liang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Guanshuang Fu
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Kaicheng Yan
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Guanxuanzi Zhang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Yun Cai
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
2
|
Teng H, Liu Y, Hao R, Zhang L, Zhang X, Li S, Li S, Tong H. The mechanism of EGF in promoting skeletal muscle post-injury regeneration. Differentiation 2025; 143:100862. [PMID: 40245761 DOI: 10.1016/j.diff.2025.100862] [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: 02/16/2025] [Revised: 04/02/2025] [Accepted: 04/06/2025] [Indexed: 04/19/2025]
Abstract
Epidermal Growth Factor (EGF) is a multifunctional cytokine that plays an important role in the growth and development of skeletal muscle. In this study, a mouse skeletal muscle post-injury regeneration model and the C2C12 myoblasts cell line were used to elucidate the molecular mechanism by which EGF promotes myoblast proliferation and differentiation and then improves skeletal muscle post-injury regeneration. EGF regulates the activities of p38-MAPK and PI3K/AKT/mTOR signaling pathways through the Epidermal Growth Factor Receptor (EGFR), thereby promoting the proliferation and differentiation of myoblasts. This finding will support the treatment of skeletal muscle injury, which is of great value in resolving muscle health problems such as muscular atrophy and sarcopenia.
Collapse
Affiliation(s)
- Huaixin Teng
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China; Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Yongze Liu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China; Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Ruotong Hao
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China; Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Lu Zhang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China; Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Xiaoyu Zhang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China; Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Shufeng Li
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China; Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Shuang Li
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China; Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Huili Tong
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China; Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
3
|
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
|
4
|
Feng F, Luo K, Yuan X, Lan T, Wang S, Xu X, Lu Z. Aerobic Exercise Protects against Cardiotoxin-Induced Skeletal Muscle Injury in a DDAH1-Dependent Manner. Antioxidants (Basel) 2024; 13:1069. [PMID: 39334728 PMCID: PMC11428882 DOI: 10.3390/antiox13091069] [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: 07/31/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Dimethylarginine dimethylaminohydrolase 1 (DDAH1) is a critical enzyme that regulates nitric oxide (NO) signaling through the degradation of asymmetric dimethylarginine (ADMA). Previous studies have revealed a link between the beneficial effects of aerobic exercise and the upregulation of DDAH1 in bones and hearts. We previously reported that skeletal muscle DDAH1 plays a protective role in cardiotoxin (CTX)-induced skeletal muscle injury and regeneration. To determine the effects of aerobic exercise on CTX-induced skeletal muscle injury and the role of DDAH1 in this process, wild-type (WT) mice and skeletal muscle-specific Ddah1-knockout (Ddah1MKO) mice were subjected to swimming training for 8 weeks and then injected with CTX. In WT mice, swimming training for 8 weeks significantly promoted skeletal muscle regeneration and attenuated inflammation, oxidative stress, and apoptosis in the gastrocnemius (GA) muscle after CTX injection. These phenomena were associated with increases in the protein expression of PAX7, myogenin, MEF2A, eNOS, SOD2, and peroxiredoxin 5 and decreases in iNOS expression in GA muscles. Swimming training also decreased serum ADMA levels and increased serum nitrate/nitrite (NOx) levels and skeletal muscle DDAH1 expression. Interestingly, swimming training in Ddah1MKO mice had no obvious effect on CTX-induced skeletal muscle injury or regeneration and did not repress the CTX-induced inflammatory response, superoxide generation, or apoptosis. In summary, our data suggest that DDAH1 is important for the protective effect of aerobic exercise on skeletal muscle injury and regeneration.
Collapse
Affiliation(s)
- Fei Feng
- Sport and Health Science Department, Nanjing Sport Institute, Nanjing 210000, China
| | - Kai Luo
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyi Yuan
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ting Lan
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyu Wang
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
Liang W, Liang B, Yan K, Zhang G, Zhuo J, Cai Y. Low-Intensity Pulsed Ultrasound: A Physical Stimulus with Immunomodulatory and Anti-inflammatory Potential. Ann Biomed Eng 2024; 52:1955-1981. [PMID: 38683473 DOI: 10.1007/s10439-024-03523-y] [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: 01/28/2024] [Accepted: 04/20/2024] [Indexed: 05/01/2024]
Abstract
Ultrasound has expanded into the therapeutic field as a medical imaging and diagnostic technique. Low-intensity pulsed ultrasound (LIPUS) is a kind of therapeutic ultrasound that plays a vital role in promoting fracture healing, wound repair, immunomodulation, and reducing inflammation. Its anti-inflammatory effects are manifested by decreased pro-inflammatory cytokines and chemokines, accelerated regression of immune cell invasion, and accelerated damage repair. Although the anti-inflammatory mechanism of LIPUS is not very clear, many in vitro and in vivo studies have shown that LIPUS may play its anti-inflammatory role by activating signaling pathways such as integrin/Focal adhesion kinase (FAK)/Phosphatidylinositol 3-kinase (PI3K)/Serine threonine kinase (Akt), Vascular endothelial growth factor (VEGF)/endothelial nitric oxide synthase (eNOS), or inhibiting signaling pathways such as Toll-like receptors (TLRs)/Nuclear factor kappa-B (NF-κB) and p38-Mitogen-activated protein kinase (MAPK). As a non-invasive physical therapy, the anti-inflammatory and immunomodulatory effects of LIPUS deserve further exploration.
Collapse
Affiliation(s)
- Wenxin Liang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Beibei Liang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China
| | - Kaicheng Yan
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China
| | - Guanxuanzi Zhang
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China
| | - Jiaju Zhuo
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Yun Cai
- Center of Medicine Clinical Research, Department of Pharmacy, Medical Supplies Center of Chinese PLA General Hospital, 28 Fu Xing Road, Beijing, 100853, People's Republic of China.
| |
Collapse
|
6
|
Yang X, Liang J, Shu Y, Wei L, Wen C, Luo H, Ma L, Qin T, Wang B, Zeng S, Liu Y, Zhou C. Asperosaponin VI facilitates the regeneration of skeletal muscle injury by suppressing GSK-3β-mediated cell apoptosis. J Cell Biochem 2024; 125:115-126. [PMID: 38079224 DOI: 10.1002/jcb.30510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/26/2023] [Accepted: 11/26/2023] [Indexed: 01/16/2024]
Abstract
Asperosaponin VI (ASA VI) is a bioactive triterpenoid saponin extracted from Diptychus roots, of Diptyl, and has previously shown protective functions in rheumatoid arthritis and sepsis. This study investigates the effects and molecular mechanisms of ASA VI on skeletal muscle regeneration in a cardiotoxin (CTX)-induced skeletal muscle injury mouse model. Mice were subjected to CTX-induced injury in the tibialis anterior and C2C12 myotubes were treated with CTX. Muscle fiber histology was analyzed at 7 and 14 days postinjury. Apoptosis and autophagy-related protein expression were evaluated t s by Western blot, and muscle regeneration markers were quantified by quantitative polymerase chain reaction. Docking studies, cell viability assessments, and glycogen synthase kinase-3β (GSK-3β) activation analyses were performed to elucidate the mechanism. ASA VI was observed to improve muscle interstitial fibrosis, remodeling, and performance in CTX-treated mice, thereby increased skeletal muscle size, weight, and locomotion. Furthermore, ASA VI modulated the expression of apoptosis and autophagy-related proteins through GSK-3β inhibition and activated the transcription of regeneration genes. Our results suggest that ASA VI mitigates skeletal muscle injury by modulating apoptosis and autophagy via GSK-3β signaling and promotes regeneration, thus presenting a probable therapeutic agent for skeletal muscle injury.
Collapse
Affiliation(s)
- Xinru Yang
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jian Liang
- Department of Pediatrics, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yue Shu
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Linlin Wei
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Cailing Wen
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Hui Luo
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Liqing Ma
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Tian Qin
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Bin Wang
- Department of Cardiovascular Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Siyu Zeng
- Department of Pharmacy, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Ying Liu
- Department of Pharmacology, School of Pharmacy, Macau University of Science and Technology, Taipa, Macao, China
- Department of Pharmacology, School of Pharmacy, Guangzhou Xinhua University, Guangzhou, China
| | - Chun Zhou
- Department of Pharmacology, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Shi Y, Wu W. Multimodal non-invasive non-pharmacological therapies for chronic pain: mechanisms and progress. BMC Med 2023; 21:372. [PMID: 37775758 PMCID: PMC10542257 DOI: 10.1186/s12916-023-03076-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Chronic pain conditions impose significant burdens worldwide. Pharmacological treatments like opioids have limitations. Non-invasive non-pharmacological therapies (NINPT) encompass diverse interventions including physical, psychological, complementary and alternative approaches, and other innovative techniques that provide analgesic options for chronic pain without medications. MAIN BODY This review elucidates the mechanisms of major NINPT modalities and synthesizes evidence for their clinical potential across chronic pain populations. NINPT leverages peripheral, spinal, and supraspinal mechanisms to restore normal pain processing and limit central sensitization. However, heterogeneity in treatment protocols and individual responses warrants optimization through precision medicine approaches. CONCLUSION Future adoption of NINPT requires addressing limitations in standardization and accessibility as well as synergistic combination with emerging therapies. Overall, this review highlights the promise of NINPT as a valuable complementary option ready for integration into contemporary pain medicine paradigms to improve patient care and outcomes.
Collapse
Affiliation(s)
- Yu Shi
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Wen Wu
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
8
|
Wang J, Yuan B, Yin R, Zhang H. Inflammation Responses to Bone Scaffolds under Mechanical Stimuli in Bone Regeneration. J Funct Biomater 2023; 14:jfb14030169. [PMID: 36976093 PMCID: PMC10059255 DOI: 10.3390/jfb14030169] [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: 02/05/2023] [Revised: 03/05/2023] [Accepted: 03/18/2023] [Indexed: 03/29/2023] Open
Abstract
Physical stimuli play an important role in one tissue engineering. Mechanical stimuli, such as ultrasound with cyclic loading, are widely used to promote bone osteogenesis; however, the inflammatory response under physical stimuli has not been well studied. In this paper, the signaling pathways related to inflammatory responses in bone tissue engineering are evaluated, and the application of physical stimulation to promote osteogenesis and its related mechanisms are reviewed in detail; in particular, how physical stimulation alleviates inflammatory responses during transplantation when employing a bone scaffolding strategy is discussed. It is concluded that physical stimulation (e.g., ultrasound and cyclic stress) helps to promote osteogenesis while reducing the inflammatory response. In addition, apart from 2D cell culture, more consideration should be given to the mechanical stimuli applied to 3D scaffolds and the effects of different force moduli while evaluating inflammatory responses. This will facilitate the application of physiotherapy in bone tissue engineering.
Collapse
Affiliation(s)
- Junjie Wang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Bo Yuan
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Ruixue Yin
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Hongbo Zhang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
9
|
Qin H, Du L, Luo Z, He Z, Wang Q, Chen S, Zhu YL. The therapeutic effects of low-intensity pulsed ultrasound in musculoskeletal soft tissue injuries: Focusing on the molecular mechanism. Front Bioeng Biotechnol 2022; 10:1080430. [PMID: 36588943 PMCID: PMC9800839 DOI: 10.3389/fbioe.2022.1080430] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Musculoskeletal soft tissue injuries are very common and usually occur during both sporting and everyday activities. The intervention of adjuvant therapies to promote tissue regeneration is of great importance to improving people's quality of life and extending their productive lives. Though many studies have focused on the positive results and effectiveness of the LIPUS on soft tissue, the molecular mechanisms standing behind LIPUS effects are much less explored and reported, especially the intracellular signaling pathways. We incorporated all research on LIPUS in soft tissue diseases since 2005 and summarized studies that uncovered the intracellular molecular mechanism. This review will also provide the latest evidence-based research progress in this field and suggest research directions for future experiments.
Collapse
Affiliation(s)
- Haocheng Qin
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Liang Du
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhong He
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Wang
- Department of Orthopedics, Kunshan Hospital of Chinese Medicine, Suzhou, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu-Lian Zhu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
da Silva ANG, de Oliveira JRS, Madureira ÁNDM, Lima WA, Lima VLDM. Biochemical and Physiological Events Involved in Responses to the Ultrasound Used in Physiotherapy: A Review. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:2417-2429. [PMID: 36115728 DOI: 10.1016/j.ultrasmedbio.2022.07.009] [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: 09/30/2021] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
Therapeutic ultrasound (TUS) is the ultrasound modality widely used in physical therapy for the treatment of acute and chronic injuries of various biological tissues. Its thermal and mechanical effects modify the permeability of the plasma membrane, the flow of ions and molecules and cell signaling and, in this way, promote the cascade of physiological events that culminate in the repair of injuries. This article is a review of the biochemical and physiological effects of TUS with parameters commonly used by physical therapists. Integrins can translate the mechanical signal of the TUS into a cellular biochemical signal for protein synthesis and modification of the active site of enzymes, so cell function and metabolism are modified. TUS also alters the permeability of the plasma membrane, allowing the influx of ions and molecules that modulate the cellular electrochemical signaling pathways. With biochemical and electrochemical signals tampered with, the cellular response to damage is then modified or enhanced. Greater release of pro-inflammatory factors, cytokines and growth factors, increased blood flow and activation of protein kinases also seem to be involved in the therapeutic response of TUS. Although a vast number of publications describe the mechanisms by which TUS can interact with the biological system, little is known about the metabolic possibilities of TUS because of the lack of standardization in its application.
Collapse
Affiliation(s)
- Ayala Nathaly Gomes da Silva
- Laboratório de Lipídios e Aplicaçães de Biomoléculas em Doenças Prevalentes e Negligenciadas, Universidade Federal de Pernambuco, Recife, Brazil
| | - João Ricardhis Saturnino de Oliveira
- Laboratório de Lipídios e Aplicaçães de Biomoléculas em Doenças Prevalentes e Negligenciadas, Universidade Federal de Pernambuco, Recife, Brazil
| | - Álvaro Nóbrega de Melo Madureira
- Laboratório de Lipídios e Aplicaçães de Biomoléculas em Doenças Prevalentes e Negligenciadas, Universidade Federal de Pernambuco, Recife, Brazil
| | - Wildberg Alencar Lima
- Laboratório de Lipídios e Aplicaçães de Biomoléculas em Doenças Prevalentes e Negligenciadas, Universidade Federal de Pernambuco, Recife, Brazil
| | - Vera Lúcia de Menezes Lima
- Laboratório de Lipídios e Aplicaçães de Biomoléculas em Doenças Prevalentes e Negligenciadas, Universidade Federal de Pernambuco, Recife, Brazil.
| |
Collapse
|
11
|
Wang Y, Lu J, Liu Y. Skeletal Muscle Regeneration in Cardiotoxin-Induced Muscle Injury Models. Int J Mol Sci 2022; 23:ijms232113380. [PMID: 36362166 PMCID: PMC9657523 DOI: 10.3390/ijms232113380] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Skeletal muscle injuries occur frequently in daily life and exercise. Understanding the mechanisms of regeneration is critical for accelerating the repair and regeneration of muscle. Therefore, this article reviews knowledge on the mechanisms of skeletal muscle regeneration after cardiotoxin-induced injury. The process of regeneration is similar in different mouse strains and is inhibited by aging, obesity, and diabetes. Exercise, microcurrent electrical neuromuscular stimulation, and mechanical loading improve regeneration. The mechanisms of regeneration are complex and strain-dependent, and changes in functional proteins involved in the processes of necrotic fiber debris clearance, M1 to M2 macrophage conversion, SC activation, myoblast proliferation, differentiation and fusion, and fibrosis and calcification influence the final outcome of the regenerative activity.
Collapse
|
12
|
Ueno M, Maeshige N, Hirayama Y, Yamaguchi A, Ma X, Uemura M, Kondo H, Fujino H. Pulsed ultrasound prevents lipopolysaccharide-induced muscle atrophy through inhibiting p38 MAPK phosphorylation in C2C12 myotubes. Biochem Biophys Res Commun 2021; 570:184-190. [PMID: 34293592 DOI: 10.1016/j.bbrc.2021.07.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Inflammation contributes to skeletal muscle atrophy via protein degradation induced by p38 mitogen-activated protein kinase (MAPK) phosphorylation. Meanwhile, pulsed ultrasound irradiation provides the mechanical stimulation to the target tissue, and has been reported to show anti-inflammatory effects. This study investigated the preventive effects of pulsed ultrasound irradiation on muscle atrophy induced by lipopolysaccharide (LPS) in C2C12 myotubes. METHODS C2C12 myotubes were used in this research. The pulsed ultrasound (a frequency of 3 MHz, duty cycle of 20%, intensity of 0.5 W/cm2) was irradiated to myotube before LPS administration. RESULTS The LPS increased phosphorylation of p38 MAPK and decreased the myofibril and myosin heavy chain protein (P < 0.05), followed by atrophy in C2C12 myotubes. The pulsed ultrasound irradiation attenuated p38 MAPK phosphorylation and myotube atrophy induced by LPS (P < 0.05). CONCLUSIONS Pulsed ultrasound irradiation has the preventive effects on inflammation-induced muscle atrophy through inhibiting phosphorylation of p38 MAPK.
Collapse
Affiliation(s)
- Mizuki Ueno
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Noriaki Maeshige
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Yusuke Hirayama
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Atomu Yamaguchi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Xiaoqi Ma
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Mikiko Uemura
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Hiroyo Kondo
- Department of Food Science and Nutrition, Nagoya Women's University, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.
| |
Collapse
|
13
|
Wang ZG, Zhu ZQ, He ZY, Cheng P, Liang S, Chen AM, Yang Q. Endogenous conversion of n-6 to n-3 polyunsaturated fatty acids facilitates the repair of cardiotoxin-induced skeletal muscle injury in fat-1 mice. Aging (Albany NY) 2021; 13:8454-8466. [PMID: 33714197 PMCID: PMC8034919 DOI: 10.18632/aging.202655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022]
Abstract
In this study, we investigated the beneficial effects of high endogenous levels of n-3 polyunsaturated fatty acids (PUFAs) on skeletal muscle repair and regeneration using a mouse cardiotoxin (CTX, 20 μM/200 μL) -induced gastrocnemius muscle injury model. Transgenic fat-1 mice expressing the Caenorhabditis elegans fat-1 gene, encoding n-3 fatty acid desaturase, showed higher n-3 PUFA levels and lower n-6/n-3 PUFA ratios in gastrocnemius muscle tissues. Hematoxylin and eosin and Masson’s trichrome staining of gastrocnemius sections revealed increased muscle fiber size and reduced fibrosis in fat-1 mice on days 7 and 14 after CTX injections. Gastrocnemius muscle tissues from fat-1 mice showed reduced inflammatory responses and increased muscle fiber regeneration reflecting enhanced activation of satellite cells on day 3 after cardiotoxin injections. Gastrocnemius muscle tissues from cardiotoxin-treated fat-1 mice showed reduced levels of pro-apoptotic proteins (Caspase 3 and Bax) and increased levels of anti-apoptotic proteins (Bcl-2 and Survivin). Moreover, eicosapentaenoic acid (EPA) reduced the incidence of apoptosis among cardiotoxin-treated C2C12 mouse myoblasts. These findings demonstrate that higher endogenous n-3 PUFA levels in fat-1 mice enhances skeletal muscle repair and regeneration following cardiotoxin-induced injury.
Collapse
Affiliation(s)
- Zheng-Gang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, PR China
| | - Zi-Qing Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, PR China
| | - Zhi-Yi He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, PR China
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, PR China
| | - Shuang Liang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, PR China
| | - An-Min Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, PR China
| | - Qing Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, PR China
| |
Collapse
|
14
|
Sakamoto M. Effects of Physical Agents on Muscle Healing with a Focus on Animal Model Research. Phys Ther Res 2021; 24:1-8. [PMID: 33981522 PMCID: PMC8111410 DOI: 10.1298/ptr.r0011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/24/2020] [Indexed: 11/23/2022]
Abstract
Skeletal muscle injury is caused by a variety of events, such as muscle laceration, contusions, or strain. Muscle fibers respond to minor damage with immediate repair mechanisms that reseal the cell membrane. On the other hand, repair of irreversibly damaged fibers is achieved by activation of muscle precursor cells. Muscle repair is not always perfect, especially after severe damage, and can lead to excessive fibroblast proliferation that results in the formation of scar tissue within muscle fibers. Remaining scar tissue can impair joint movement, reduce muscular strength, and inhibit exercise ability; therefore, to restore muscle function, minimizing the extent of injury and promoting muscle regeneration are necessary. Various physical agents, such as cold, thermal, electrical stimulation, and low-intensity pulsed ultrasound therapy, have been reported as treatments for muscle healing. Although approaches based on the muscle regeneration process have been under development, the most efficacious physiological treatment for muscle injury remains unclear. In this review, the influence of these physical agents on muscle injury is described with a focus on research using animal models.
Collapse
Affiliation(s)
- Miki Sakamoto
- Department of Physical Therapy, School of Allied Health Sciences, Kitasato University, Japan
| |
Collapse
|
15
|
Maeshige N, Langston PK, Yuan ZM, Kondo H, Fujino H. High-intensity ultrasound irradiation promotes the release of extracellular vesicles from C2C12 myotubes. ULTRASONICS 2021; 110:106243. [PMID: 32961400 DOI: 10.1016/j.ultras.2020.106243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 08/19/2020] [Accepted: 08/28/2020] [Indexed: 06/11/2023]
Abstract
Skeletal muscle is an important secretory organ in mammals, producing myriad chemical mediators ("myokines") with distinct biological action in different tissues, including anti-inflammatory activity. Extracellular vesicles (EVs) have recently been identified as a mode of myokine transport from muscle, facilitating such anti-inflammatory activity. In this report, we have demonstrated that high-intensity ultrasound (US) strongly induces EV secretion from cultured myotubes without a reduction in cell viability. High-intensity US of 3.0 W/cm2 with 20% duty cycle increased the number of EVs by 2-fold compared to control at 6 h. This effect was specific to EVs in the 100-150 nm size range. Thus, high-intensity US is a novel modality for inducing myocellular EV release and may hold therapeutic value.
Collapse
Affiliation(s)
- Noriaki Maeshige
- Department of Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.
| | - P Kent Langston
- Department of Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, MA, USA
| | - Zhi-Min Yuan
- Department of Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Hiroyo Kondo
- Department of Food Sciences and Nutrition, Nagoya Women's University, Nagoya, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
16
|
Xu M, Wang L, Wu S, Dong Y, Chen X, Wang S, Li X, Zou C. Review on experimental study and clinical application of low-intensity pulsed ultrasound in inflammation. Quant Imaging Med Surg 2021; 11:443-462. [PMID: 33392043 DOI: 10.21037/qims-20-680] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Low-intensity pulsed ultrasound (LIPUS), as physical therapy, is widely used in both research and clinical settings. It induces multiple bioeffects, such as alleviating pain, promoting tissue repair, and shortening disease duration. LIPUS can also mediate inflammation. This paper reviews the application of LIPUS in inflammation and discusses the underlying mechanism. In basic experiments, LIPUS can regulate inflammatory responses at the cellular level by affecting some signaling pathways. In a clinical trial, LIPUS has been shown to alleviate inflammatory responses efficiently. As a cheap, safe, and convenient physical method, LIPUS is promising as anti-inflammatory therapy.
Collapse
Affiliation(s)
- Maosheng Xu
- Department of Ultrasonography, The Second Affiliated Hospital, and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liang Wang
- Department of Ultrasonography, The Second Affiliated Hospital, and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Senmin Wu
- Department of Ultrasonography, The Second Affiliated Hospital, and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yanyan Dong
- Department of Ultrasonography, The Second Affiliated Hospital, and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiu Chen
- Department of Ultrasonography, The Second Affiliated Hospital, and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shijia Wang
- Department of Ultrasonography, The Second Affiliated Hospital, and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiuyun Li
- Department of Ultrasonography, The Second Affiliated Hospital, and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chunpeng Zou
- Department of Ultrasonography, The Second Affiliated Hospital, and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
17
|
Madzia A, Agrawal C, Jarit P, Petterson S, Plancher K, Ortiz R. Sustained Acoustic Medicine Combined with A Diclofenac Ultrasound Coupling Patch for the Rapid Symptomatic Relief of Knee Osteoarthritis: Multi-Site Clinical Efficacy Study. Open Orthop J 2020; 14:176-185. [PMID: 33408796 PMCID: PMC7784557 DOI: 10.2174/1874325002014010176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/07/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Sustained Acoustic Medicine (SAM) is an emerging, non-invasive, non-narcotic, home-use ultrasound therapy for the daily treatment of joint pain. The aim of this multi-site clinical study was to examine the efficacy of long-duration continuous ultrasound combined with a 1% diclofenac ultrasound gel patch in treating pain and improving function in patients with knee osteoarthritis. METHODS The Consolidated Standards of Reporting Trials (CONSORT) were followed. Thirty-two (32) patients (18-males, 14-females) 54 years of average age with moderate to severe knee pain and radiographically confirmed knee osteoarthritis (Kellgren-Lawrence (KL) grade II/III) were enrolled for treatment with the SAM device and diclofenac patch applied daily to the treated knee. SAM ultrasound (3 MHz, 0.132 W/cm2, 1.3 W) and 6 grams of 1% diclofenac were applied with a wearable device for 4 hours daily for 1 week, delivering 18,720 Joules of ultrasound energy per treatment. The primary outcome was the daily change in pain intensity using a numeric rating scale (NRS 0-10), which was assessed prior to intervention (baseline, day 1), before and after each daily treatment, and after 1 week of daily treatment (day 7). Rapid responders were classified as those patients exhibiting greater than a 1-point reduction in pain following the first treatment. Change in Western Ontario McMaster Osteoarthritis Questionnaire (WOMAC) score from baseline to day 7 was the secondary functional outcome measure. Additionally, a series of daily usability and user experience questions related to devising ease of use, functionality, safety, and effectiveness, were collected. Data were analyzed using t-tests and repeated measure ANOVAs. RESULTS The study had a 94% retention rate, and there were no adverse events or study-related complaints across 224 unique treatment sessions. Rapid responders included 75% of the study population. Patients exhibited a significant mean NRS pain reduction over the 7-day study of 2.06-points (50%) for all subjects (n=32, p<0.001) and 2.96-points (70%) for rapid responders (n=24, p<0.001). The WOMAC functional score significantly improved by 351 points for all subjects (n=32, p<0.001), and 510 points for rapid responders (n=24, p<0.001). Over 95% of patients found the device safe, effective and easy to use, and would continue treatment for their knee OA symptoms. CONCLUSION Sustained Acoustic Medicine combined with 1% topical diclofenac rapidly reduced pain and improved function in patients with moderate to severe osteoarthritis-related knee pain. The clinical findings suggest that this treatment approach may be used as a conservative, non-invasive treatment option for patients with knee osteoarthritis. Additional research is warranted on non-weight bearing joints of the musculoskeletal system as well as different topical drugs that could benefit from improved localized delivery.Clinical Trial Registry Number: (NCT04391842).
Collapse
Affiliation(s)
- Alex Madzia
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45219, USA
| | - Chirag Agrawal
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45219, USA
| | - Paddy Jarit
- Sport and Orthopaedic Physical Therapy, Fairfield, CT 06824, USA
| | | | - Kevin Plancher
- Albert Einstein College of Medicine, Bronx, NY, New York, USA
- Weill Cornell Medical College, New York, NY 13053, USA
- Plancher Orthopaedics & Sports Medicine, New York, NY 13053, USA
| | - Ralph Ortiz
- Medical Pain Consultants, Dryden, NY 13053, USA
| |
Collapse
|
18
|
IL-1β and TNF-α Modulation of Proliferated and Committed Myoblasts: IL-6 and COX-2-Derived Prostaglandins as Key Actors in the Mechanisms Involved. Cells 2020; 9:cells9092005. [PMID: 32882817 PMCID: PMC7564831 DOI: 10.3390/cells9092005] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/11/2022] Open
Abstract
In this study, we investigated the effects and mechanisms of the pro-inflammatory cytokines IL-1β and TNF-α on the proliferation and commitment phases of myoblast differentiation. C2C12 mouse myoblast cells were cultured to reach a proliferated or committed status and were incubated with these cytokines for the evaluation of cell proliferation, cyclooxygenase 2 (COX-2) expression, release of prostaglandins (PGs) and myokines, and activation of myogenic regulatory factors (MRFs). We found that inhibition of the IL-6 receptor reduced IL-1β- and TNF-α-induced cell proliferation, and that the IL-1β effect also involved COX-2-derived PGs. Both cytokines modulated the release of the myokines myostatin, irisin, osteonectin, and IL-15. TNF-α and IL-6 reduced the activity of Pax7 in proliferated cells and reduced MyoD and myogenin activity at both proliferative and commitment stages. Otherwise, IL-1β increased myogenin activity only in committed cells. Our data reveal a key role of IL-6 and COX-2-derived PGs in IL-1β and TNF-α-induced myoblast proliferation and support the link between TNF-α and IL-6 and the activation of MRFs. We concluded that IL-1β and TNF-α induce similar effects at the initial stages of muscle regeneration but found critical differences between their effects with the progression of the process, bringing new insights into inflammatory signalling in skeletal muscle regeneration.
Collapse
|
19
|
Effectiveness of low-intensity pulsed ultrasound on osteoarthritis of the temporomandibular joint: A review. Ann Biomed Eng 2020; 48:2158-2170. [PMID: 32514932 DOI: 10.1007/s10439-020-02540-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/26/2020] [Indexed: 01/15/2023]
Abstract
Loading is indispensable for the growth, development, and maintenance of joint tissues, including mandibular condylar cartilage, but excessive loading or reduced host adaptive capacity can considerably damage the temporomandibular joint (TMJ), leading to temporomandibular joint osteoarthritis (TMJ-OA). TMJ-OA, associated with other pathological conditions and aging processes, is a highly degenerative disease affecting the articular cartilage. Many treatment modalities for TMJ-OA have been developed. Traditional clinical treatment includes mainly nonsurgical options, such as occlusal splints. However, non-invasive therapy does not achieve joint tissue repair and regeneration. Growing evidence suggests that low-intensity pulsed ultrasound (LIPUS) accelerates bone fracture healing and regeneration, as well as having extraordinary effects in terms of soft tissue repair and regeneration. The latter have received much attention, and various studies have been performed to evaluate the potential role of LIPUS in tissue regeneration including that applied to articular cartilage. The present article provides an overview of the status of LIPUS stimulation used to prevent the onset and progression of TMJ-OA and enhance the tissue regeneration of mandibular condylar cartilage. The etiology and management of TMJ-OA are explained briefly, animal models of TMJ-OA are described, and the effectiveness of LIPUS on cell metabolism and tissue regeneration in the TMJ is discussed.
Collapse
|
20
|
Jiang X, Savchenko O, Li Y, Qi S, Yang T, Zhang W, Chen J. A Review of Low-Intensity Pulsed Ultrasound for Therapeutic Applications. IEEE Trans Biomed Eng 2019; 66:2704-2718. [DOI: 10.1109/tbme.2018.2889669] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
21
|
Fujita M, Sato-Shigeta M, Mori H, Iwasa A, Kawai N, Hassan AH, Tanaka E. Protective Effects of Low-Intensity Pulsed Ultrasound on Mandibular Condylar Cartilage Exposed to Mechanical Overloading. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:944-953. [PMID: 30732913 DOI: 10.1016/j.ultrasmedbio.2018.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/04/2018] [Accepted: 12/17/2018] [Indexed: 06/09/2023]
Abstract
The aim of this study was to assess the effect of low-intensity pulsed ultrasound (LIPUS) application on rat temporomandibular joints (TMJs) with early-stage of osteoarthritis-like conditions induced by mechanical overloading. Fifteen-week-old male Wistar rats were divided into two experimental groups and a control group (n = 10 each). Both TMJs of all rats in one experimental group were subjected to mechanical overloading for 5 d, and those in the other experimental group were exposed to LIPUS for 20 min/d after overloading. Condyles were assessed using micro-computed tomography, histology and histomorphometry. LIPUS treatment attenuated cartilage degeneration, decreased the number of osteoclastic cells and restored the expression of aggrecan after an initial decrease induced by mechanical overloading. These results indicate that LIPUS may have a protective effect on the early progression of TMJ osteoarthritis.
Collapse
Affiliation(s)
- Mutsumi Fujita
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Oral Sciences, Tokushima, Japan.
| | - Minami Sato-Shigeta
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Hospital, Tokushima, Japan
| | - Hiroki Mori
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Hospital, Tokushima, Japan
| | - Akihiko Iwasa
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Nobuhiko Kawai
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Ali H Hassan
- Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Eiji Tanaka
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
22
|
Ohno Y, Egawa T, Yokoyama S, Fujiya H, Sugiura T, Ohira Y, Yoshioka T, Goto K. MENS-associated increase of muscular protein content via modulation of caveolin-3 and TRIM72. Physiol Res 2019; 68:265-273. [PMID: 30628834 DOI: 10.33549/physiolres.933992] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Microcurrent electrical neuromuscular stimulation (MENS) is known as an extracellular stimulus for the regeneration of injured skeletal muscle in sports medicine. However, the effects of MENS-associated increase in muscle protein content are not fully clarified. The purpose of this study was to investigate the effects of MENS on the muscular protein content, intracellular signals, and the expression level of caveolin-3 (Cav-3), tripartite motif-containing 72 (TRIM72) and MM isoenzyme of creatine kinase (CK-MM) in skeletal muscle using cell culture system. C2C12 myotubes on the 7th day of differentiation phase were treated with MENS (intensity: 10-20 microA, frequency: 0.3 Hz, pulse width: 250 ms, stimulation time: 15-120 min). MENS-associated increase in the protein content of myotubes was observed, compared to the untreated control level. MENS upregulated the expression of Cav-3, TRIM72, and CK-MM in myotubes. A transient increase in phosphorylation level of Akt was also observed. However, MENS had no effect on the phosphorylation level of p42/44 extracellular signal-regulated kinase-1/2 and 5'AMP-activated protein kinase. MENS may increase muscle protein content accompanied with a transient activation of Akt and the upregulation of Cav-3 and TRIM72.
Collapse
Affiliation(s)
- Y Ohno
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Aichi, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Low-intensity focused ultrasound stimulator using focal depth controller for improved targeting in neuromuscular rehabilitation. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2018; 2017:209-212. [PMID: 29059847 DOI: 10.1109/embc.2017.8036799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Low-intensity ultrasound can be used for noninvasive neuromuscular stimulation to enhance the recovery of injured peripheral nerves and muscles. In order to improve the stimulation efficiency, focused ultrasound (FUS) with a high energy density should be used. Because the FUS is concentrated at a miniaturized focal area, deliberate targeting with focal depth control is required to achieve the accurate stimulation. In this study, a low-intensity focused ultrasound (LIFU) stimulator with a focal depth controller is presented as an effective ultrasound stimulation system for the neuromuscular rehabilitation. In the developed stimulator, the target depth is adjusted by the focal depth controller with the brief structure. The experimental results show that the developed stimulator can generate the high quality LIFU stimulation with the spatial average-pulse average intensity (ISAPA) of 496 mW/cm2, which is sufficient to improve the rehabilitation of injured peripheral nerves and muscles. In the in vivo experiment, a rat's sciatic nerve was successfully targeted by the developed stimulator. These results indicate that the developed stimulator can be used to improve the quality of the ultrasound stimulation for neuromuscular rehabilitation by targeting a specific area.
Collapse
|
24
|
Why is Skeletal Muscle Regeneration Impaired after Myonecrosis Induced by Viperid Snake Venoms? Toxins (Basel) 2018; 10:toxins10050182. [PMID: 29723952 PMCID: PMC5983238 DOI: 10.3390/toxins10050182] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/22/2018] [Accepted: 04/25/2018] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle regeneration after myonecrosis involves the activation, proliferation and fusion of myogenic cells, and a coordinated inflammatory response encompassing phagocytosis of necrotic cell debris, and the concerted synthesis of cytokines and growth factors. Myonecrosis often occurs in snakebite envenomings. In the case of venoms that cause myotoxicity without affecting the vasculature, such as those of many elapid snakes, regeneration proceeds successfully. In contrast, in envenomings by most viperid snakes, which affect the vasculature and extracellular matrix in addition to muscle fibers, regeneration is largely impaired and, therefore, the muscle mass is reduced and replaced by fibro-adipose tissue. This review discusses possible causes for such poor regenerative outcome including: (a) damage to muscle microvasculature, which causes tissue hypoxia and affects the inflammatory response and the timely removal of necrotic tissue; (b) damage to intramuscular nerves, which results in atrophy of regenerating fibers; (c) degradation of muscle cell basement membrane, compromising the spatial niche for proliferating myoblasts; (d) widespread degradation of the extracellular matrix; and (e) persistence of venom components in the damaged tissue, which may affect myogenic cells at critical points in the regenerative process. Understanding the causes of poor muscle regeneration may pave the way for the development of novel therapeutic interventions aimed at fostering the regenerative process in envenomed patients.
Collapse
|
25
|
Li Y, Feng Q, Guo M, Wang Y, Jiang Y, Xing J. Genome-wide survey reveals dynamic effects of folate supplement on DNA methylation and gene expression during C2C12 differentiation. Physiol Genomics 2018; 50:158-168. [PMID: 29341861 DOI: 10.1152/physiolgenomics.00094.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Folic acid supplements taken during pregnancy can prevent neural tube defects and other developmental abnormalities. Here, we explored the effects of folate supplementation on gene expression and DNA methylation during C2C12 differentiation. Based on the folic acid concentration, this study comprised three groups: low folate (L), normal folate (N), and high-folate supplement (H). Our analyses revealed that differentiation and the mRNA expression of the gene myogenin in C2C12 cell were enhanced by folic acid; however, the overall methylation percentage in myogenin promoter between different treatment groups was not significantly different ( P > 0.05). The results of MeDIP-chip showed that hundreds of differentially methylated regions (DMRs) were identified between every two groups in both promoter and CpG islands, respectively. Genes with DMRs between N and L groups were mainly enriched in the processes of cell differentiation and cell development, whereas those with DMRs between H and N groups were frequently enriched in cellular process/cycle and cell metabolic processes. In addition, correlation analysis between methylation profile and expression profile revealed that some genes were regulated by methylation status directly. Together, these analyses suggest that folate deficiency and supplementation can influence the differentiation, genome-wide DNA methylation level and the expression of myogenesis-related genes including myogenin in the C2C12 cell line.
Collapse
Affiliation(s)
- Yi Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Qiang Feng
- The Central Hospital of Taian, Tai'an, Shandong , China
| | - Miao Guo
- Taishan Medical University, Tai'an, Shandong , China
| | - Yuding Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Yunliang Jiang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Jinyi Xing
- School of Life Science, Linyi University, Linyi, Shandong , China
| |
Collapse
|
26
|
Zhang X, Hu B, Sun J, Li J, Liu S, Song J. Inhibitory Effect of Low-Intensity Pulsed Ultrasound on the Expression of Lipopolysaccharide-Induced Inflammatory Factors in U937 Cells. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2017; 36:2419-2429. [PMID: 28600899 DOI: 10.1002/jum.14239] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/01/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVES Low-intensity pulsed ultrasound (US) has been reported to promote periodontal tissue regeneration and reduce inflammation in soft tissues and in bone infectious diseases. Here we investigated the effect of low-intensity pulsed US on the expression of lipopolysaccharide (LPS)-induced inflammatory factors in U937 macrophage cells. METHODS U937 cells were stimulated with different concentrations of LPS and exposed to different intensities of low-intensity pulsed US. Cell viability and apoptosis of U937 cells were determined by cell-counting kit assays and flow cytometry. A real-time polymerase chain reaction and an enzyme-linked immunosorbent assay were used to test the expression of inflammatory factors. The expression levels of toll-like receptor 4, p65, p-IκBα, and IκBα were assessed by western blots. RESULTS Tumor necrosis factor α began to increase in U937 cells on induction with 1-μg/mL LPS. Low-intensity pulsed US at the intensity of 60 mW/cm2 was more effective in reducing interleukin 8 (IL-8) expression. Furthermore, LPS inhibited the viability and increased apoptosis of U937 cells, whereas low-intensity pulsed US significantly reversed these effects (P < .05). Low-intensity pulsed US reduced the protein expression of IL-6 and IL-8 at both gene and protein levels in U937 cells. The western blot and immunofluorescence showed that low-intensity pulsed US primarily suppressed the degradation and phosphorylation of IκBα and the translocation of p65 into the nuclei. CONCLUSIONS Low-intensity pulsed US alleviated the expression of inflammatory factors induced by LPS in U937 cells. This process was modulated by suppressing the toll-like receptor 4-nuclear factor κB signaling pathway. Therefore, low-intensity pulsed US might be a potential immunomodulatory therapy for the treatment of periodontitis.
Collapse
Affiliation(s)
- Xuan Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Bo Hu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jicheng Sun
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Shan Liu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
27
|
Salgarella AR, Cafarelli A, Ricotti L, Capineri L, Dario P, Menciassi A. Optimal Ultrasound Exposure Conditions for Maximizing C2C12 Muscle Cell Proliferation and Differentiation. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:1452-1465. [PMID: 28433437 DOI: 10.1016/j.ultrasmedbio.2017.03.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 02/27/2017] [Accepted: 03/06/2017] [Indexed: 05/07/2023]
Abstract
Described here is an in vitro systematic investigation of the effects on C2C12 myoblasts of exposure to finely controlled and repeatable low-intensity pulsed ultrasound of different frequencies (500 kHz, 1 MHz, 3 MHz and 5 MHz) and different intensities (250, 500 and 1000 mW/cm2). An in-house stimulation system and an ultrasound-transparent cell culture well minimized reflections and attenuations, allowing precise control of ultrasound delivery. Results indicated that a 3 MHz stimulation at 1 W/cm2 intensity maximized cell proliferation in comparison with the other exposure conditions and untreated controls. In contrast, cell differentiation and the consequent formation of multinucleated myotubes were maximized by 1 MHz stimulation at 500 mW/cm2 intensity. The highly controlled exposure conditions employed allowed precise correlation of the ultrasound delivery to the bio-effects produced, thus overcoming the inconsistency of some results available in the literature and contributing to the potential of ultrasound treatment for muscle therapy and regeneration.
Collapse
Affiliation(s)
| | - Andrea Cafarelli
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Pontedera (Pisa), Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Pontedera (Pisa), Italy
| | - Lorenzo Capineri
- Department of Information Engineering, University of Florence, Florence, Italy
| | - Paolo Dario
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Pontedera (Pisa), Italy
| | - Arianna Menciassi
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Pontedera (Pisa), Italy
| |
Collapse
|
28
|
Nieminen HJ, Barreto G, Finnilä MA, García-Pérez A, Salmi A, Ranjan S, Eklund KK, Pritzker KPH, Saarakkala S, Hæggström E. Laser-ultrasonic delivery of agents into articular cartilage. Sci Rep 2017; 7:3991. [PMID: 28638116 PMCID: PMC5479804 DOI: 10.1038/s41598-017-04293-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 05/12/2017] [Indexed: 11/09/2022] Open
Abstract
Research is ongoing to develop drug therapies to manage osteoarthritis (OA) and articular cartilage (AC) injuries. However, means to deliver drug to localized AC lesions are highly limited and not clinically available. This study investigates the capability of laser ultrasound (laser-induced plasma sound source) to deliver agents (methylene blue, MB, in PBS) into bovine AC. Treatment samples (n = 10) were immersed in MB solution simultaneously with LU exposure, while adjacent control 1 tissue (n = 10) was pre-treated with LU followed by immersion in MB and adjacent control 2 tissue (n = 10) was only immersed in MB. AC exposed (n = 22) or not exposed (n = 27) to LU were characterized for anomalies in structure, composition, viability or RNA expression. Optically detected MB content was significantly (p < 0.01) higher in treatment samples up to a depth of 500 µm from AC surface as compared to controls. No major unwanted short-term effects on AC structure, proteoglycan or collagen contents, chondrocyte viability or RNA expression levels were detected. In conclusion, LU can deliver agents into AC without major short-term concerns on safety. LU could reveal new strategies for the development of localized drug therapies in AC.
Collapse
Affiliation(s)
- Heikki J Nieminen
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland.
- Research Group of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
| | - Gonçalo Barreto
- Orton Orthopaedic Hospital and Research Institute, Invalid Foundation, Helsinki, Finland
- Department of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko A Finnilä
- Research Group of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Alejandro García-Pérez
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland
- Department of Electronic Engineering, Higher Technological Institute of Poza Rica, Poza Rica, México, USA
| | - Ari Salmi
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland
| | - Sanjeev Ranjan
- Laboratory of Radiochemistry, Department of Chemistry, University of Helsinki, Helsinki, Finland
| | - Kari K Eklund
- Department of Rheumatology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kenneth P H Pritzker
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, Mount Sinai Hospital, Toronto, Canada
| | - Simo Saarakkala
- Research Group of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Edward Hæggström
- Electronics Research Laboratory, Department of Physics, University of Helsinki, Helsinki, Finland
| |
Collapse
|
29
|
Best TM, Wilk KE, Moorman CT, Draper DO. Low Intensity Ultrasound for Promoting Soft Tissue Healing: A Systematic Review of the Literature and Medical Technology. ACTA ACUST UNITED AC 2016; 2. [PMID: 30198009 DOI: 10.18103/imr.v2i11.271] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Therapeutic ultrasound has been studied and used for the past seven decades to treat musculoskeletal injuries. Recently, a significant body of animal and human research has focused on the biomechanical effects of daily-applied, low intensity therapeutic ultrasound (LITUS) on soft tissue recovery. We performed a systematic review of the last two decades of LITUS literature to examine the effects on tendon, skeletal muscle, ligament, and tendon-bone junction injuries. LITUS facilitated tendon healing, with increased tensile strength and improved collagen alignment. For skeletal muscle and ligament injuries, LITUS increased cell proliferation during myoregeneration and improved tissue biomechanics (ultimate load, stiffness, energy absorption). LITUS aided tendon-bone junction healing through improved tissue function. Scientific evidence supports the use of LITUS to treat soft tissue injuries, and improve outcomes for musculoskeletal injuries and post-operative recovery. Lastly, we discuss the use of LITUS devices facilitating daily applied therapy in the home setting.
Collapse
|
30
|
Chongsatientam A, Yimlamai T. Therapeutic Pulsed Ultrasound Promotes Revascularization and Functional Recovery of Rat Skeletal Muscle after Contusion Injury. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:2938-2949. [PMID: 27665217 DOI: 10.1016/j.ultrasmedbio.2016.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 07/26/2016] [Accepted: 08/02/2016] [Indexed: 06/06/2023]
Abstract
The mechanism by which therapeutic pulsed ultrasound (TPU) promotes the repair of damaged gastrocnemius muscle was investigated. Male Wistar rats were divided into uninjured, sham-treated injured and TPU-treated injured (TPU) groups. Injury was induced by mass-drop technique. TPU was applied to the injured muscle for 5 min, daily, started at day 1 post-injury and continuing for 3, 7 and 14 d. For 3 d post-injury, a significant reduction in muscle force was observed in both the sham-treated injured and TPU groups. TPU treatment significantly increased recovery force of the injured muscle after day 7 post-injury. This effect of TPU is associated with increased centronucleated fibers and cross-sectional area, mRNA expression of the vascular endothelial growth factor and capillary density of the regenerated fibers, but not with mRNA expression of nitric oxide synthase. We conclude that TPU hastens muscle recovery, at least in part, by upregulating angiogenesis.
Collapse
Affiliation(s)
| | - Tossaporn Yimlamai
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
31
|
Sato M, Kuroda S, Mansjur KQ, Khaliunaa G, Nagata K, Horiuchi S, Inubushi T, Yamamura Y, Azuma M, Tanaka E. Low-intensity pulsed ultrasound rescues insufficient salivary secretion in autoimmune sialadenitis. Arthritis Res Ther 2015; 17:278. [PMID: 26445930 PMCID: PMC4596462 DOI: 10.1186/s13075-015-0798-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/24/2015] [Indexed: 11/17/2022] Open
Abstract
Introduction Low-intensity pulsed ultrasound (LIPUS) has been known to promote bone healing by nonthermal effects. In recent studies, LIPUS has been shown to reduce inflammation in injured soft tissues. Xerostomia is one of the most common symptoms in Sjögren syndrome (SS). It is caused by a decrease in the quantity or quality of saliva. The successful treatment of xerostomia is still difficult to achieve and often unsatisfactory. The aim of this study is to clarify the therapeutic effects of LIPUS on xerostomia in SS. Methods Human salivary gland acinar (NS-SV-AC) and ductal (NS-SV-DC) cells were cultured with or without tumor necrosis factor-α (TNF-α; 10 ng/ml) before LIPUS or sham exposure. The pulsed ultrasound signal was transmitted at a frequency of 1.5 MHz or 3 MHz with a spatial average intensity of 30 mW/cm2 and a pulse rate of 20 %. Cell number, net fluid secretion rate, and expression of aquaporin 5 (AQP5) and TNF-α were subsequently analyzed. Inhibitory effects of LIPUS on the nuclear factor κB (NF-κB) pathway were determined by Western blot analysis. The effectiveness of LIPUS in recovering salivary secretion was also examined in a MRL/MpJ/lpr/lpr (MRL/lpr) mouse model of SS with autoimmune sialadenitis. Results TNF-α stimulation of NS-SV-AC and NS-SV-DC cells resulted in a significant decrease in cell number and net fluid secretion rate (p < 0.01), whereas LIPUS treatment abolished them (p < 0.05). The expression changes of AQP5 and TNF-α were also inhibited in LIPUS treatment by blocking the NF-κB pathway. Furthermore, we found that mRNA expression of A20, a negative feedback regulator, was significantly increased by LIPUS treatment after TNF-α or interleukin 1β stimulation (NS-SV-AC, p < 0.01; NS-SV-DC, p < 0.05). In vivo LIPUS exposure to MRL/lpr mice exhibited a significant increase in both salivary flow and AQP5 expression by reducing inflammation in salivary glands (p < 0.01). Conclusions These results suggest that LIPUS upregulates expression of AQP5 and inhibits TNF-α production. Thus, LIPUS may restore secretion by inflamed salivary glands. It may synergistically activate negative feedback of NF-κB signaling in response to inflammatory stimulation. Collectively, LIPUS might be a new strategic therapy for xerostomia in autoimmune sialadenitis with SS. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0798-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Minami Sato
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Shingo Kuroda
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Karima Qurnia Mansjur
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Ganzorig Khaliunaa
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Kumiko Nagata
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Shinya Horiuchi
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Toshihiro Inubushi
- Genetic Disease Program, Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA.
| | - Yoshiko Yamamura
- Department of Oral Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Masayuki Azuma
- Department of Oral Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Eiji Tanaka
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan. .,Department of Orthodontics, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia.
| |
Collapse
|
32
|
Ganzorig K, Kuroda S, Maeda Y, Mansjur K, Sato M, Nagata K, Tanaka E. Low-intensity pulsed ultrasound enhances bone formation around miniscrew implants. Arch Oral Biol 2015; 60:902-10. [PMID: 25824309 DOI: 10.1016/j.archoralbio.2015.02.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 02/10/2015] [Accepted: 02/18/2015] [Indexed: 11/17/2022]
Abstract
UNLABELLED Miniscrew implants (MSIs) are currently used to provide absolute anchorage in orthodontics; however, their initial stability is an issue of concern. Application of low-intensity pulsed ultrasound (LIPUS) can promote bone healing. Therefore, LIPUS application may stimulate bone formation around MSIs and enhance their initial stability. AIM To investigate the effect of LIPUS exposure on bone formation after implantation of titanium (Ti) and stainless steel (SS) MSIs. METHODS MSIs made of Ti-6Al-4V and 316L SS were placed on rat tibiae and treated with LIPUS. The bone morphology around MSIs was evaluated by scanning electron microscopy and three-dimensional micro-computed tomography. MC3T3-E1 cells cultured on Ti and SS discs were treated with LIPUS, and the temporary expression of alkaline phosphatase (ALP) was examined. RESULTS Bone-implant contact increased gradually from day 3 to day 14 after MSI insertion. LIPUS application increased the cortical bone density, cortical bone thickness, and cortical bone rate after implantation of Ti and SS MSIs (P<0.05). LIPUS exposure induced ALP upregulation in MC3T3-E1 cells at day 3 (P<0.05). CONCLUSION LIPUS enhanced bone formation around Ti and SS MSIs, enhancing the initial stability of MSIs.
Collapse
Affiliation(s)
- Khaliunaa Ganzorig
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Oral Sciences, Tokushima, Japan
| | - Shingo Kuroda
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Health Biosciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yuichi Maeda
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Oral Sciences, Tokushima, Japan
| | - Karima Mansjur
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Oral Sciences, Tokushima, Japan
| | - Minami Sato
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Oral Sciences, Tokushima, Japan
| | - Kumiko Nagata
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Health Biosciences, Tokushima University Graduate School, Tokushima, Japan
| | - Eiji Tanaka
- Department of Orthodontics and Dentofacial Orthopedics, Institute of Health Biosciences, Tokushima University Graduate School, Tokushima, Japan.
| |
Collapse
|
33
|
Rossato DD, Dal Lago P, Hentschke VS, Rucatti AL, Signori LU, Silveira MN, Plentz RDM. Ultrasound modulates skeletal muscle cytokine levels in rats with heart failure. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:797-805. [PMID: 25619785 DOI: 10.1016/j.ultrasmedbio.2014.11.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 11/11/2014] [Accepted: 11/24/2014] [Indexed: 06/04/2023]
Abstract
Heart failure is a multisystemic disorder that leads to an imbalance between pro- and anti-inflammatory cytokines. Therapeutic ultrasound (TU) has been reported to modulate the inflammatory process. The aim of this study was to evaluate the effect of TU on pro- and anti-inflammatory cytokine levels in soleus muscle and plasma of rats with heart failure. Thirty male Wistar rats (230-260 g) were submitted to ligation of the left coronary artery or sham surgery. Six weeks after surgery, TU was administered directly to the right lower limb. The results indicate that TU promotes reduction of pro-inflammatory cytokine levels (tumor necrosis factor α, interleukin-6) and increases anti-inflammatory cytokine levels (interleukin-10) in the soleus muscle of rats with heart failure. This is the first study to find that TU can modulate cytokine levels in rats with heart failure. Additionally, this is a first report that TU can modulate interleukin-10 levels in the soleus muscle.
Collapse
Affiliation(s)
- Douglas Dalcin Rossato
- Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Fisiologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Centro Universitário Franciscano, Santa Maria, Brazil
| | - Pedro Dal Lago
- Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Fisiologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Vítor Scotta Hentschke
- Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Fisiologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Ananda Lazzarotto Rucatti
- Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Fisiologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Luis Ulisses Signori
- Departamento de Fisioterapia e Reabilitação, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Matheus Noronha Silveira
- Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Fisiologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Rodrigo Della Méa Plentz
- Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Fisiologia, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
34
|
Low-intensity pulsed ultrasound in dentofacial tissue engineering. Ann Biomed Eng 2015; 43:871-86. [PMID: 25672801 DOI: 10.1007/s10439-015-1274-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/04/2015] [Indexed: 02/04/2023]
Abstract
Oral and maxillofacial diseases affect millions of people worldwide and hence tissue engineering can be considered an interesting and clinically relevant approach to regenerate orofacial tissues after being affected by different diseases. Among several innovations for tissue regeneration, low-intensity pulsed ultrasound (LIPUS) has been used extensively in medicine as a therapeutic, operative, and diagnostic tool. LIPUS is accepted to promote bone fracture repair and regeneration. Furthermore, the effect of LIPUS on soft tissues regeneration has been paid much attention, and many studies have performed to evaluate the potential use of LIPUS to tissue engineering soft tissues. The present article provides an overview about the status of LIPUS stimulation as a tool to be used to enhance regeneration/tissue engineering. This review consists of five parts. Part 1 is a brief introduction of the acoustic description of LIPUS and mechanical action. In Part 2, biological problems in dentofacial tissue engineering are proposed. Part 3 explores biologic mechanisms of LIPUS to cells and tissues in living body. In Part 4, the effectiveness of LIPUS on cell metabolism and tissue regeneration in dentistry are summarized. Finally, Part 5 relates the possibility of clinical application of LIPUS in orthodontics. The present review brings out better understanding of the bioeffect of LIPUS therapy on orofacial tissues which is essential to the successful integration of management remedies for tissue regeneration/engineering. To develop an evidence-based approach to clinical management and treatment of orofacial degenerative diseases using LIPUS, we would like to be in full pursuit of LIPUS biotherapy. Still, there are many challenges for this relatively new strategy, but the up to date achievements using it promises to go far beyond the present possibilities.
Collapse
|
35
|
Abrunhosa VM, Soares CP, Batista Possidonio AC, Alvarenga AV, Costa-Felix RPB, Costa ML, Mermelstein C. Induction of skeletal muscle differentiation in vitro by therapeutic ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2014; 40:504-512. [PMID: 24412173 DOI: 10.1016/j.ultrasmedbio.2013.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/11/2013] [Accepted: 10/15/2013] [Indexed: 06/03/2023]
Abstract
Therapeutic ultrasound (TU) has been used for the last 50 y in rehabilitation, including treatment of soft tissues. Ultrasound waves can be employed in two different modes of operation, continuous and pulsed, which produce both thermal and non-thermal effects. Despite the large-scale use of TU, there are few scientific studies on its biologic effects during skeletal muscle differentiation. To better analyze the cellular effects of TU, we decided to follow cells in vitro. The main purpose of this study was to evaluate the effects of TU in primary chick myogenic cell cultures using phase contrast optical microscopy and immunofluorescence microscopy, followed by image analysis and quantification. Our results indicate that TU can stimulate the differentiation of skeletal muscle cells in vitro, as measured by the thickness of multinucleated myotubes, the ratio of mononucleated cells to multinucleated cells and expression of the muscle-specific protein desmin. This study is a first step toward a metrologic and science-based protocol for cell treatment under different ultrasound field exposures.
Collapse
Affiliation(s)
- Viviane Mendes Abrunhosa
- Laboratório de Ultrassom, Diretoria de Metrologia Científica e Industrial (DIMCI), Instituto Nacional de Metrologia, Qualidade e Tecnologia (INMETRO), Rio de Janeiro, RJ, Brazil; Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carolina Pontes Soares
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - André Victor Alvarenga
- Laboratório de Ultrassom, Diretoria de Metrologia Científica e Industrial (DIMCI), Instituto Nacional de Metrologia, Qualidade e Tecnologia (INMETRO), Rio de Janeiro, RJ, Brazil
| | - Rodrigo P B Costa-Felix
- Laboratório de Ultrassom, Diretoria de Metrologia Científica e Industrial (DIMCI), Instituto Nacional de Metrologia, Qualidade e Tecnologia (INMETRO), Rio de Janeiro, RJ, Brazil
| | - Manoel Luis Costa
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Claudia Mermelstein
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
36
|
Negishi Y, Ishii Y, Shiono H, Akiyama S, Sekine S, Kojima T, Mayama S, Kikuchi T, Hamano N, Endo-Takahashi Y, Suzuki R, Maruyama K, Aramaki Y. Bubble liposomes and ultrasound exposure improve localized morpholino oligomer delivery into the skeletal muscles of dystrophic mdx mice. Mol Pharm 2014; 11:1053-61. [PMID: 24433046 DOI: 10.1021/mp4004755] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder that is caused by mutations in the DMD gene that lead to an absence of functional protein. The mdx dystrophic mouse contains a nonsense mutation in exon 23 of the dystrophin gene; a phosphorodiamidate morpholino oligomer (PMO) designed to skip this mutated exon in the mRNA induces dystrophin expression. However, an efficient PMO delivery method is needed to improve treatment strategies for DMD. We previously developed polyethylene glycol (PEG)-modified liposomes (Bubble liposomes) that entrap ultrasound contrast gas and demonstrated that the combination of Bubble liposomes with ultrasound exposure is an effective gene delivery tool in vitro and in vivo. In this study, to evaluate the ability of Bubble liposomes as a PMO delivery tool, we tested the potency of the Bubble liposomes combined with ultrasound exposure to boost the delivery of PMO and increase the skipping of the mutated exon in the mdx mouse. The results indicated that the combination of Bubble liposomes and ultrasound exposure increased the uptake of the PMO targeting a nonsense mutation in exon 23 of the dystrophin gene and consequently increased the PMO-mediated exon-skipping efficiency compared with PMO injection alone, leading to significantly enhanced dystrophin expression. This increased efficiency indicated the potential of the combination of Bubble liposomes with ultrasound exposure to enhance PMO delivery for treating DMD. Thus, this ultrasound-mediated Bubble liposome technique may provide an effective, noninvasive, nonviral method for PMO therapy for DMD muscle as well as for other muscular dystrophies.
Collapse
Affiliation(s)
- Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences , 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|