1
|
Liao Q, Chen J, Liu G. Low intensity pulsed ultrasound alleviates synovial fibrosis in osteoarthritis via the PI3K/AKT pathway. Sci Rep 2025; 15:9644. [PMID: 40113833 PMCID: PMC11926212 DOI: 10.1038/s41598-025-92413-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025] Open
Abstract
Previous studies have reported that low-intensity pulsed ultrasound (LIPUS) can alleviate cartilage degradation in osteoarthritis (OA). However, the functions and mechanisms of LIPUS in synovial fibrosis with OA require further study. To investigate the role of the PI3K/AKT signaling pathway in synovial fibrosis and in LIPUS treatment in synovial fibrosis, a TGF-β stimulated rat FLS cell model and a rat OA animal model based on anterior cruciate ligament transection (ACLT) and partial medial meniscectomy (MMx) were used. The results revealed that LIPUS delayed the progression of OA. Masson staining revealed that LIPUS reduced the collagen deposition of synovial tissue in OA rats. Correspondingly, immunofluorescence demonstrated that LIPUS significantly downregulated the expression of α-SMA, Col1a1 and Col3a1 in OA rats. Moreover, TGF-β stimulation upregulated fibrosis markers at the mRNA and protein levels in FLS, as well as increased phosphorylation-dependent activation of the PI3K/Akt pathway. 740Y-P was found to promote the fibrotic change of FLS induced by TGF-β, but LY294002 reduced its expression. However, LIPUS inhibits the fibrotic change and activation of the PI3K/Akt pathway in FLS under stimulation of TGF-β. In conclusion, LIPUS alleviates synovial fibrosis by blocking the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Qing Liao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China
| | - Jun Chen
- Department of Rehabilitation Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, 510000, China
- Taihe Hospital, Hubei University of Medicine, Shiyan City, 442000, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
2
|
Zhang K, Wang T, Huang X, Wu P, Shen L, Yang Y, Wan W, Sun S, Zhang Z. Ultrasound-mediated nanomaterials for the treatment of inflammatory diseases. ULTRASONICS SONOCHEMISTRY 2025; 114:107270. [PMID: 39961217 DOI: 10.1016/j.ultsonch.2025.107270] [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: 12/12/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
Sterile and infection-associated inflammatory diseases are becoming increasingly prevalent worldwide. Conventional drug therapies often entail significant drawbacks, such as the risk of drug overdose, the development of drug resistance in pathogens, and systemic adverse reactions, all of which can undermine the effectiveness of treatments for these conditions. Nanomaterials (NMs) have emerged as a promising tool in the treatment of inflammatory diseases due to their precise targeting capabilities, tunable characteristics, and responsiveness to external stimuli. Ultrasound (US), a non-invasive and effective treatment method, has been explored in combination with NMs to achieve enhanced therapeutic outcomes. This review provides a comprehensive overview of the recent advances in the use of US-mediated NMs for treating inflammatory diseases. A comprehensive introduction to the application and classification of US was first presented, emphasizing the advantages of US-mediated NMs and the mechanisms through which US and NMs interact to enhance anti-inflammatory therapy. Subsequently, specific applications of US-mediated NMs in sterile and infection-associated inflammation were summarized. Finally, the challenges and prospects of US-mediated NMs in clinical translation were discussed, along with an outline of future research directions. This review aims to provide insights to guide the development and improvement of US-mediated NMs for more effective therapeutic interventions in inflammatory diseases.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China
| | - Tingting Wang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Xingyong Huang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Lufan Shen
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Yuanyuan Yang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Wenyu Wan
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, PR China; Key Laboratory of Immunodermatology, National Health Commission of the People's Republic of China, The First Hospital of China Medical University, PR China; National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, PR China.
| | - Siyu Sun
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China.
| | - Zhan Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, PR China; Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
3
|
Liang C, Zhang Y, Yan Y, Geng W, Li J, Liu X. LIPUS promotes osteogenic differentiation of rat BMSCs and osseointegration of dental implants by regulating ITGA11 and focal adhesion pathway. BMC Oral Health 2025; 25:22. [PMID: 39755586 DOI: 10.1186/s12903-024-05411-2] [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: 10/07/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Low-intensity pulsed ultrasound (LIPUS) has been used as an effective noninvasive method for treating fractures and osteoarthrosis, but the application in the field of oral implantation is in its infancy. This study aimed to clarify the effect and mechanism of LIPUS on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and implant osseointegration, and to provide an experimental basis for future clinical applications. METHODS Dental implants were inserted into Wistar rat femurs, and LIPUS was performed for 4 weeks. Micro-CT and toluidine blue staining were used to assess implant osseointegration. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were used to identify enriched functional terms and signalling pathways for differentially expressed genes from LIPUS-treated rat BMSC RNAseq data obtained from the GEO database. The random forest method was used to identify key risk genes according to the mean decrease Gini (MDG) coefficient. Then, LIPUS was applied to treat rat BMSCs, and alkaline phosphatase (ALP) staining, alizarin red staining, RT-PCR and western blotting were used to determine whether LIPUS could promote BMSC osteogenic differentiation via integrin α11 (ITGA11) and the focal adhesion pathway. RESULTS Our in vivo experimentations verified that LIPUS significantly increased new bone formation and osseointegration around the implant in rats. Bioinformatics analysis of RNA-seq data revealed that the upregulated genes in BMSCs after LIPUS treatment were significantly enriched in osteoblast differentiation-related functions and focal adhesion-related pathways. Random forest analysis revealed that ITGA11 was the most significant factor affecting BMSC osteogenic differentiation among the differentially expressed genes. In addition, LIPUS significantly increased ALP expression and mineralized nodule formation in rat BMSCs by upregulating ITGA11 and increasing the activity of FAK/PI3K/AKT/GSK3β/β-catenin pathway. CONCLUSIONS LIPUS can effectively promote implant osseointegration in rats and improve rat BMSC osteogenic differentiation by upregulating ITGA11 and increasing the activity of the downstream focal adhesion pathway.
Collapse
Affiliation(s)
- Chao Liang
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yuqing Zhang
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yuwei Yan
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Wei Geng
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Jun Li
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Xiu Liu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
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
|
5
|
Wu D, Zhao X, Xie J, Yuan R, Li Y, Yang Q, Cheng X, Wu C, Wu J, Zhu N. Physical modulation of mesenchymal stem cell exosomes: A new perspective for regenerative medicine. Cell Prolif 2024; 57:e13630. [PMID: 38462759 PMCID: PMC11294442 DOI: 10.1111/cpr.13630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/29/2024] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Mesenchymal stem cell-derived exosomes (MSC-Exo) offer promising therapeutic potential for various refractory diseases, presenting a novel therapeutic strategy. However, their clinical application encounters several obstacles, including low natural secretion, uncontrolled biological functions and inherent heterogeneity. On the one hand, physical stimuli can mimic the microenvironment dynamics where MSC-Exo reside. These factors influence not only their secretion but also, significantly, their biological efficacy. Moreover, physical factors can also serve as techniques for engineering exosomes. Therefore, the realm of physical factors assumes a crucial role in modifying MSC-Exo, ultimately facilitating their clinical translation. This review focuses on the research progress in applying physical factors to MSC-Exo, encompassing ultrasound, electrical stimulation, light irradiation, intrinsic physical properties, ionizing radiation, magnetic field, mechanical forces and temperature. We also discuss the current status and potential of physical stimuli-affected MSC-Exo in clinical applications. Furthermore, we address the limitations of recent studies in this field. Based on this, this review provides novel insights to advance the refinement of MSC-Exo as a therapeutic approach in regenerative medicine.
Collapse
Affiliation(s)
- Dan Wu
- Department of DermatologyHuashan Hospital, Fudan UniversityShanghaiChina
| | - Xiansheng Zhao
- Department of DermatologyHuashan Hospital, Fudan UniversityShanghaiChina
| | - Jiaheng Xie
- Department of Plastic SurgeryXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Ruoyue Yuan
- Department of DermatologyHuashan Hospital, Fudan UniversityShanghaiChina
| | - Yue Li
- Department of DermatologyHuashan Hospital, Fudan UniversityShanghaiChina
| | - Quyang Yang
- Department of DermatologyHuashan Hospital, Fudan UniversityShanghaiChina
| | - Xiujun Cheng
- Department of DermatologyHuashan Hospital, Fudan UniversityShanghaiChina
| | - Changyue Wu
- Department of DermatologyHuashan Hospital, Fudan UniversityShanghaiChina
| | - Jinyan Wu
- Department of DermatologyChongzhou People's HospitalChengduChina
| | - Ningwen Zhu
- Department of DermatologyHuashan Hospital, Fudan UniversityShanghaiChina
- Department of PlasticReconstructive and Burns Surgery, Huashan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
6
|
Liu Y, Lin R, Fang H, Li L, Zhang M, Lu L, Gao X, Song J, Wei J, Xiao Q, Zhang F, Wu K, Cui L. Sargassum polysaccharide attenuates osteoarthritis in rats and is associated with the up-regulation of the ITGβ1-PI3K-AKT signaling pathway. J Orthop Translat 2024; 47:176-190. [PMID: 39040490 PMCID: PMC11260896 DOI: 10.1016/j.jot.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/06/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
Background Osteoarthritis (OA) presents a formidable challenge, characterized by as-yet-unclear mechanical intricacies within cartilage and the dysregulation of bone homeostasis. Our preliminary data revealed the encouraging potential of a Sargassum polysaccharide (SP), in promoting chondrogenesis. The aim of our study is to comprehensively assess the therapeutic effects of SP on OA models and further elucidate its potential mechanism. Methods The protective effects of SP were initially evaluated in an inflammation-induced human chondrocyte (C28) cell model. CCK-8 assays, Alcian blue staining, RT-qPCR and Western blotting were used to verify the chondrogenesis of SP in vitro. To assess the efficacy of SP in vivo, surgically induced medial meniscus destabilization (DMM) OA rats underwent an 8-week SP treatment. The therapeutic effects of SP in OA rats were comprehensively evaluated using X-ray imaging, micro-computed tomography (μ-CT), histopathological analysis, as well as immunohistochemical and immunofluorescent staining. Following these assessments, we delved into the potential signaling pathways of SP in inflammatory chondrocytes utilizing RNA-seq analysis. Validation of these findings was conducted through RT-qPCR and western blotting techniques. Results SP significantly enhance the viability of C28 chondrocytes, and increased the secretion of acidic glycoproteins. Moreover, SP stimulated the expression of chondrogenic genes (Aggrecan, Sox9, Col2a1) and facilitated the synthesis of Collagen II protein in C28 inflammatory chondrocytes. In vivo experiments revealed that SP markedly ameliorated knee joint stenosis, alleviated bone and cartilage injuries, and reduced the histopathological scores in the OA rats. μ-CT analysis confirmed that SP lessened bone impairments in the medial femoral condyle and the subchondral bone of the tibial plateau, significantly improving the microarchitectural parameters of the subchondral bone. Histopathological analyses indicated that SP notably enhanced cartilage quality on the surface of the tibial plateau, leading to increased cartilage thickness and area. Immunohistochemistry staining and immunofluorescence staining corroborated these findings by showing a significant promotion of Collagen II expression in OA joints treated with SP. RNA-seq analysis suggest that SP's effects were mediated through the regulation of the ITGβ1-PI3K-AKT signaling axis, thereby stimulating chondrogenesis. Verification through RT-qPCR and Western blot analyses confirmed that SP significantly upregulated the expression of ITGβ1, p110δ, AKT1, ACAN, and Col2a1. Notably, knock-down of ITGβ1 using siRNA in C28 chondrocytes inhibited the expression of ITGβ1, p110δ, AKT1, and ACAN. However, these inhibitory effects were not completely reversed by supplemental SP intervention. Conclusions In summary, our findings reveal that SP significantly enhances chondrogenesis both in vitro and in vivo, alleviating OA progression both in bone and cartilage. The observed beneficial effects are intricately linked to the activation of the ITGβ1-PI3K-AKT signaling axis. The translational potential of this article Our research marks the first instance unveiling the advantageous effects and underlying mechanisms of SP in OA treatment. With its clinical prospects, SP presents compelling new evidence for the advancement of a next-generation polysaccharide drug for OA therapy.
Collapse
Affiliation(s)
- Yanzhi Liu
- Corresponding author. Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524045, China.
| | | | | | - Lixian Li
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Min Zhang
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Lujiao Lu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Xiang Gao
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Jintong Song
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Jinsong Wei
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Qixian Xiao
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Fucheng Zhang
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Kefeng Wu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| | - Liao Cui
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, School of Ocean and Tropical Medicine, The Affiliated Hospital, The Second Affiliated Hospital, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
7
|
Feng X, Li S, Wang S, Meng Y, Zheng S, Liu C, Chang B, Shi C, Sun H. Piezo1 mediates the degradation of cartilage extracellular matrix in malocclusion-induced TMJOA. Oral Dis 2024; 30:2425-2438. [PMID: 37184045 DOI: 10.1111/odi.14615] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 04/20/2023] [Accepted: 04/22/2023] [Indexed: 05/16/2023]
Abstract
OBJECTIVES To evaluate the role of Piezo1 in the malocclusion-induced osteoarthritic cartilage of the temporomandibular joint. METHODS A temporomandibular joint osteoarthritis model was established using a unilateral anterior crossbite in vivo, and cartilage degeneration and Piezo1 expression were observed by histological and immunohistochemical staining. ATDC5 cells were loaded with 24 dyn/cm2 fluid flow shear stress using the Flexcell device in vitro and expression and function of Piezo1 were evaluated. After identifying the function of Piezo1 in YAP translocation under FFSS conditions, the influence of Piezo1 and YAP on metabolism-related enzymes under FFSS was detected through a real-time polymerase chain reaction analysis and western blotting. A UAC-TMJ injection model was established to observe the therapeutic effect of intra-articular injection of a Piezo1 inhibitor on osteoarthritic cartilage matrix loss. RESULTS Piezo1 was overexpressed in the osteoarthritic cartilage and cultured chondrocytes under shear stress. Piezo1 Silencing inhibited the nuclear translocation of YAP and subsequently downregulated the expression of MMP13 and ADAMTS5. Intra-articular injection of the Piezo1 inhibitor, GsMTx4, could ameliorate proteoglycan degradation in malocclusion-induced TMJOA and suppressed MMP13 and ADAMTS5 expression. CONCLUSIONS Our results revealed that the activation of Piezo1 promotes mechanical-induced cartilage degradation through the YAP-MMP13/ADAMTS5 signaling pathway.
Collapse
Affiliation(s)
- Xu Feng
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Shenyang, China
- Department of Oral Pathology, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Siwen Li
- Department of Oral Pathology, School and Hospital of Stomatology, China Medical University, Shenyang, China
- Department of Prosthodontics, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Shuangshuang Wang
- Department of Oral Pathology, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Yuan Meng
- Department of Oral Pathology, School and Hospital of Stomatology, China Medical University, Shenyang, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Shize Zheng
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Cangwei Liu
- Department of Oral Pathology, School and Hospital of Stomatology, China Medical University, Shenyang, China
- Department of Prosthodontics, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Bei Chang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Ce Shi
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Hongchen Sun
- Department of Oral Pathology, School and Hospital of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
8
|
Zhou J, Ning E, Lu L, Zhang H, Yang X, Hao Y. Effectiveness of low-intensity pulsed ultrasound on osteoarthritis: molecular mechanism and tissue engineering. Front Med (Lausanne) 2024; 11:1292473. [PMID: 38695024 PMCID: PMC11061361 DOI: 10.3389/fmed.2024.1292473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/05/2024] [Indexed: 05/04/2024] Open
Abstract
Osteoarthritis (OA) is distinguished by pathological alterations in the synovial membrane, articular cartilage, and subchondral bone, resulting in physical symptoms such as pain, deformity, and impaired mobility. Numerous research studies have validated the effectiveness of low-intensity pulsed ultrasound (LIPUS) in OA treatment. The periodic mechanical waves generated by LIPUS can mitigate cellular ischemia and hypoxia, induce vibration and collision, produce notable thermal and non-thermal effects, alter cellular metabolism, expedite tissue repair, improve nutrient delivery, and accelerate the healing process of damaged tissues. The efficacy and specific mechanism of LIPUS is currently under investigation. This review provides an overview of LIPUS's potential role in the treatment of OA, considering various perspectives such as the synovial membrane, cartilage, subchondral bone, and tissue engineering. It aims to facilitate interdisciplinary scientific research and further exploration of LIPUS as a complementary technique to existing methods or surgery. Ongoing research is focused on determining the optimal dosage, frequency, timing, and treatment strategy of LIPUS for OA. Additional research is required to clarify the precise mechanism of action and potential impacts on cellular, animal, and human systems prior to its integration into therapeutic applications.
Collapse
Affiliation(s)
- Jing Zhou
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Eryu Ning
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Lingfeng Lu
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Huili Zhang
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
9
|
Huang Y, Liao J, Vlashi R, Chen G. Focal adhesion kinase (FAK): its structure, characteristics, and signaling in skeletal system. Cell Signal 2023; 111:110852. [PMID: 37586468 DOI: 10.1016/j.cellsig.2023.110852] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/29/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase and distributes important regulatory functions in skeletal system. Mesenchymal stem cell (MSC) possesses significant migration and differentiation capacity, is an important source of distinctive bone cells production and a prominent bone development pathway. MSC has a wide range of applications in tissue bioengineering and regenerative medicine, and is frequently employed for hematopoietic support, immunological regulation, and defect repair, although current research is insufficient. FAK has been identified to cross-link with many other keys signaling pathways in bone biology and is considered as a fundamental "crossroad" on the signal transduction pathway and a "node" in the signal network to mediate MSC lineage development in skeletal system. In this review, we summarized the structure, characteristics, cellular signaling, and the interactions of FAK with other signaling pathways in the skeletal system. The discovery of FAK and its mediated molecules will lead to a new knowledge of bone development and bone construction as well as considerable potential for therapeutic use in the treatment of bone-related disorders such as osteoporosis, osteoarthritis, and osteosarcoma.
Collapse
Affiliation(s)
- Yuping Huang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Junguang Liao
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Rexhina Vlashi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
10
|
Da Z, Guo R, Sun J, Wang A. Identification of osteoarthritis-characteristic genes and immunological micro-environment features through bioinformatics and machine learning-based approaches. BMC Med Genomics 2023; 16:236. [PMID: 37805587 PMCID: PMC10559406 DOI: 10.1186/s12920-023-01672-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/23/2023] [Indexed: 10/09/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a multifaceted chronic joint disease characterized by complex mechanisms. It has a detrimental impact on the quality of life for individuals in the middle-aged and elderly population while also imposing a significant socioeconomic burden. At present, there remains a lack of comprehensive understanding regarding the pathophysiology of OA. The objective of this study was to examine the genes, functional pathways, and immune infiltration characteristics associated with the development and advancement of OA. METHODS The Gene Expression Omnibus (GEO) database was utilized to acquire gene expression profiles. The R software was employed to conduct the screening of differentially expressed genes (DEGs) and perform enrichment analysis on these genes. The OA-characteristic genes were identified using the Weighted Gene Co-expression Network Analysis (WGCNA) and the Lasso algorithm. In addition, the infiltration levels of immune cells in cartilage were assessed using single-sample gene set enrichment analysis (ssGSEA). Subsequently, a correlation analysis was conducted to examine the relationship between immune cells and the OA-characteristic genes. RESULTS A total of 80 DEGs were identified. As determined by functional enrichment, these DEGs were associated with chondrocyte metabolism, apoptosis, and inflammation. Three OA-characteristic genes were identified using WGCNA and the lasso algorithm, and their expression levels were then validated using the verification set. Finally, the analysis of immune cell infiltration revealed that T cells and B cells were primarily associated with OA. In addition, Tspan2, HtrA1 demonstrated a correlation with some of the infiltrating immune cells. CONCLUSIONS The findings of an extensive bioinformatics analysis revealed that OA is correlated with a variety of distinct genes, functional pathways, and processes involving immune cell infiltration. The present study has successfully identified characteristic genes and functional pathways that hold potential as biomarkers for guiding drug treatment and facilitating molecular-level research on OA.
Collapse
Affiliation(s)
- Zheng Da
- Xingtai People's Hospital Affiliated to Hebei Medical University, Xingtai City, Hebei Province, China
| | - Rui Guo
- Xingtai People's Hospital Affiliated to Hebei Medical University, Xingtai City, Hebei Province, China.
| | - Jianjian Sun
- Ningbo Huamei Hospital, University of Chinese Academy of Sciences, Ningbo City, Zhejiang Province, China
| | - Ai Wang
- Zhongshan Hospital Affiliated to Fudan University, Shanghai City, China
| |
Collapse
|
11
|
Chu G, Niu H. Knowledge mapping and global trends in the field of low-intensity pulsed ultrasound and endocrine and metabolic diseases: a bibliometric and visual analysis from 2012 to 2022. Front Endocrinol (Lausanne) 2023; 14:1237864. [PMID: 37732128 PMCID: PMC10508976 DOI: 10.3389/fendo.2023.1237864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Background Low-intensity pulsed ultrasound (LIPUS) is a highly promising therapeutic method that has been widely used in rehabilitation, orthopedics, dentistry, urology, gynecology, and other multidisciplinary disease diagnoses and treatments. It has attracted extensive attention worldwide. However, there is currently a lack of comprehensive and systematic research on the current status and future development direction of the LIPUS field. Therefore, this study comprehensively analyzed LIPUS-related reports from the past decade using bibliometrics methods, and further conducted research specifically focusing on its application in endocrine and metabolic diseases. Methods We downloaded LIPUS literature from 2012 to 2022 reported in the Web of Science Core Collection Science Citation Index-Expanded and Social Sciences Citation Index, and used bibliometric analysis software such as VOSviewer and CiteSpace to execute the analysis and visualize the results. Results We searched for 655 English articles published on LIPUS from 2012 to 2022. China had the highest number of published articles and collaborations between China and the United States were the closest in this field. Chongqing Medical University was the institution with the highest output, and ULTRASOUND IN MEDICINE AND BIOLOGY was the journal with the most related publications. In recent years, research on the molecular mechanisms of LIPUS has continued to deepen, and its clinical applications have also continued to expand. The application of LIPUS in major diseases such as oxidative stress, regeneration mechanism, and cancer is considered to be a future research direction, especially in the field of endocrinology and metabolism, where it has broad application value. Conclusion Global research on LIPUS is expected to continue to increase, and future research will focus on its mechanisms of action and clinical applications. This study comprehensively summarizes the current development status and global trends in the field of LIPUS, and its research progress in the field of endocrine and metabolic diseases, providing valuable reference for future research in this field.
Collapse
Affiliation(s)
| | - Haitao Niu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Tran NT, Truong MD, Yun HW, Min BH. Potential of secretome of human fetal cartilage progenitor cells as disease modifying agent for osteoarthritis. Life Sci 2023; 324:121741. [PMID: 37149084 DOI: 10.1016/j.lfs.2023.121741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023]
Abstract
AIMS Osteoarthritis (OA) is caused by an imbalance in the synthesis and degradation of cartilage tissue by chondrocytes. Therefore, a therapeutic agent for OA patients that can positively affect both synthesis and degradation is needed. However, current nonsurgical treatments for OA can barely achieve satisfactory long-term outcomes in cartilage repair. Human fetal cartilage progenitor cells-secretome (ShFCPC) has shown potent anti-inflammatory and tissue-repair effects; however, its underlying mechanisms and effects on OA have rarely been systematically elucidated. This study aims to analyze and evaluate the potency of ShFCPC in modifying OA process. MAIN METHODS Herein, secreted proteins enriched in ShFCPC have been characterized, and their biological functions both in vitro and in vivo in an OA model are compared with those of human bone marrow-derived mesenchymal stem cells-secretome (ShBMSC) and hyaluronan (HA). KEY FINDINGS Secretome analysis has shown that ShFCPC is significantly enriched with extracellular matrix molecules involved in many effects of cellular processes required for homeostasis during OA progression. Biological validation in vitro has shown that ShFCPC protects chondrocyte apoptosis by suppressing the expression of inflammatory mediators and matrix-degrading proteases and promotes the secretion of pro-chondrogenic cytokines in lipopolysaccharide-induced coculture of human chondrocytes and SW982 synovial cells compared with ShBMSC. Moreover, in a rat OA model, ShFCPC protects articular cartilage by reducing inflammatory cell infiltration and M1/M2 macrophage ratio in the synovium, which directly contributes to an increase in immunomodulatory atmosphere and enhances cartilage repair compared to ShBMSC and HA. SIGNIFICANCE Our findings support clinical translations of ShFCPC as a novel agent for modifying OA process.
Collapse
Affiliation(s)
- Ngoc-Trinh Tran
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea; Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Minh-Dung Truong
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Hee-Woong Yun
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Byoung-Hyun Min
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Republic of Korea; Institute of Regenerative Medicine, Wake Forest University, NC, USA; Advanced Translational Engineering & Medical Science, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Effects and mechanotransduction pathways of therapeutic ultrasound on healthy and osteoarthritic chondrocytes: a systematic review of in vitro studies. Osteoarthritis Cartilage 2023; 31:317-339. [PMID: 36481451 DOI: 10.1016/j.joca.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate the effects and mechanotransduction pathways of therapeutic ultrasound on chondrocytes. METHOD PubMed, EMBASE and Web of Science databases were searched up to 19th September 2021 to identify in vitro studies exploring ultrasound to stimulate chondrocytes for osteoarthritis (OA) treatment. Study characteristics, ultrasound parameters, in vitro setup, and mechanotransduction pathways were collected. Risk of bias was judged using the Risk of Bias Assessment for Non-randomized Studies (RoBANS) tool. RESULTS Thirty-one studies were included comprising healthy and OA chondrocytes and explants. Most studies had high risk of performance, detection and pseudoreplication bias due to lack of temperature control, setup calibration, inadequate semi-quantitatively analyzes and independent experiments. Ultrasound was applied to the culture plate via acoustic gel, water bath or culture media. Regardless of the setup used, ultrasound stimulated the cartilage production and suppressed its degradation, although the effect size was nonsignificant. Ultrasound inhibited p38, c-Jun N-terminal kinases (JNK) and factor nuclear kappa B (NFκB) pathways in OA chondrocytes to reduce apoptosis, inflammation and matrix degradation, while triggered phosphoinositide-3-kinase/akt (PI3K/Akt), extracellular signal-regulated kinase (ERK), p38 and JNK pathways in healthy chondrocytes to promote matrix synthesis. CONCLUSION The included studies suggest that ultrasound application induces therapeutic effects on chondrocytes. However, these results should be interpreted with caution because high risk of performance, detection and pseudoreplication bias were identified. Future studies should explore the application of ultrasound on human OA chondrocytes cultures to potentiate the applicability of ultrasound towards cartilage regeneration of knee with OA.
Collapse
|
14
|
Okur S, Okumuş Z. Effects of low-level laser therapy and therapeutic ultrasound on Freund's complete adjuvant-induced knee arthritis model in rats. Arch Rheumatol 2023; 38:32-43. [PMID: 37235114 PMCID: PMC10208612 DOI: 10.46497/archrheumatol.2022.9409] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/05/2022] [Indexed: 08/08/2023] Open
Abstract
OBJECTIVES The aim of this study was to evaluate and monitor the effect of low-level laser therapy (LLLT) and therapeutic ultrasound (TU) alone, or combined with intra-articular prednisolone (P) in Freund's complete adjuvant (FCA)-induced knee arthritis model in rats. MATERIALS AND METHODS A total of 56 adult male Wistar rats were divided into seven groups: control (C), disease control (RA), P, TU, LLLT (L), P + TU (P+TU), P + LLLT (P+L) groups. The skin temperature, radiography, joint volume, serum rheumatoid factor (RF), interleukin (IL)-1β, serum tumor necrosis factor-alpha (TNF-α), and histopathological evaluation of joint were performed. RESULTS Thermal imaging and radiographic examination provided results consistent with the severity of the disease. The mean joint temperature (°C) was the highest in the RA (36.2±1.6) group on Day 28. The P+TU and P+L groups significantly decreased radiological scores at the end of the study. The rat serum TNF-α, IL-1β, and RF levels in all groups were significantly higher compared to the C group (p<0.05). Compared to the RA group, serum TNF-α, IL-1β, and RF levels were significantly lower in the treatment groups (p<0.05). The P+TU and P+L group was showed minimal chondrocyte degeneration and cartilage erosion and mild cartilage fibrillation and mononuclear cell infiltration of synovial membrane compared to the P, TU, and L group. CONCLUSION The LLLT and TU effectively reduced inflammation. In addition, a more effective result was obtained from the use of LLLT and TU combined with intra-articular P. This result may be due to insufficient dose of LLLT and TU, thus further studies should be focus on at higher dose ranges on FCA arthritis model in rats.
Collapse
Affiliation(s)
- Sıtkıcan Okur
- Department of Veterinary Surgery, Atatürk University Faculty of Veterinary Medicine, Erzurum, Türkiye
| | - Zafer Okumuş
- Department of Veterinary Surgery, Atatürk University Faculty of Veterinary Medicine, Erzurum, Türkiye
| |
Collapse
|
15
|
Mo Q, Kulyar MFEA, Quan C, Ding Y, Zhang Y, Zhang L, Pan H, Li J. Thiram-induced hyperglycemia causes tibial dyschondroplasia by triggering aberrant ECM remodeling via the gut-pancreas axis in broiler chickens. JOURNAL OF HAZARDOUS MATERIALS 2023; 444:130368. [PMID: 36423455 DOI: 10.1016/j.jhazmat.2022.130368] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/28/2022] [Accepted: 11/08/2022] [Indexed: 06/16/2023]
Abstract
Pesticide thiram is widely used in agriculture and has been demonstrated to cause tibial dyschondroplasia (TD) in birds. However, the underlying mechanism remains unclear. This work used multi-omics analysis to evaluate the molecular pathways of TD in broilers that were exposed to low level of thiram. Integrative analysis of transcriptomic, proteomic, and metabolomic revealed thiram activity in enhancing pathological ECM remodeling via attenuating the glycolysis pathway and activating the hexosamine and glucuronic acid pathways. Intriguingly, we found hyperglycemia as a crucial factor for ECM overproduction, which resulted in the development of TD. We further demonstrated that high glucose levels are caused by islet secretion dysfunction in thiram-treated broilers. A combination of factors, including lipid disorder, low-grade inflammation, and gut flora disturbance, might contribute to the dysregulation of insulin secretion. The current work revealed the underlying toxicological mechanisms of thiram-induced tibial dyschondroplasia through blood glucose disorder via the gut-pancreas axis in chickens for the first time, which makes it easier to figure out the health risks of pesticides for worldwide policy decisions.
Collapse
Affiliation(s)
- Quan Mo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Muhammad Fakhar-E-Alam Kulyar
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Chuxian Quan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yanmei Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yan Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Lihong Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Huachun Pan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
16
|
Kamatsuki Y, Aoyama E, Furumatsu T, Ozaki T, Takigawa M. The Evaluation of Meniscus Regenerative Effects of LIPUS-Induced CCN Proteins: Induction by LIPUS of CCN2 and Meniscus-Related Genes in Cultured Meniscus Cells and Meniscus Tissues. Methods Mol Biol 2023; 2582:223-235. [PMID: 36370353 DOI: 10.1007/978-1-0716-2744-0_15] [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] [Indexed: 06/16/2023]
Abstract
Menisci are a pair of crescent-shaped fibrocartilages and composed primarily of type I collagen. Inner region of the meniscus has similar characteristics to articular cartilage. Low-intensity pulsed ultrasound (LIPUS) has been reported to have chondroprotective effects on chondrocytes by inducing the expression of chondrocyte differentiation markers and CCN2/CTGF production. Here, we describe an experimental approach that investigates the distinct cellular behavior of human inner and outer meniscus cells in response to LIPUS stimulation. Our experimental model can analyze the relationships between LIPUS-induced CCN2 and its repairing role in the meniscus.
Collapse
Affiliation(s)
- Yusuke Kamatsuki
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kitaku, Okayama, Japan
- Advanced Research Center for Oral and Craniofacial Sciences (ARCOCS), Okayama University Dental School/Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kitaku, Okayama, Japan
| | - Eriko Aoyama
- Advanced Research Center for Oral and Craniofacial Sciences (ARCOCS), Okayama University Dental School/Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kitaku, Okayama, Japan
| | - Takayuki Furumatsu
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kitaku, Okayama, Japan
| | - Toshifumi Ozaki
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kitaku, Okayama, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences (ARCOCS), Okayama University Dental School/Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Kitaku, Okayama, Japan.
| |
Collapse
|
17
|
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] [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
|
18
|
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
|
19
|
Sumsuzzman DM, Khan ZA, Choi J, Hong Y. Assessment of functional roles and therapeutic potential of integrin receptors in osteoarthritis: A systematic review and meta-analysis of preclinical studies. Ageing Res Rev 2022; 81:101729. [PMID: 36087701 DOI: 10.1016/j.arr.2022.101729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/22/2022] [Accepted: 09/03/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Integrins are heterodimeric transmembrane receptors that mediate a variety of biological function and plays a critical role in osteoarthritis (OA) pathogenesis, which may provide new targets for the development of OA therapies. However, the roles of integrins in different stages of OA remain elusive. OBJECTIVES This study aimed to synthesize all published preclinical evidence on the roles of integrin receptors in different stages of OA to identify the potential target for drug development in alleviating OA pathogenesis. METHODS Major electronic databases were used to identify related original articles. The methodological quality of all included studies was appraised using the SYRCLE risk of bias tool. We used the generic inverse variance with random effects model to calculate standardized mean differences (SMDs) and 95% confidence interval (CI). RESULTS Seventeen studies were included in this systematic review. Integrin α5β1 activation increases the histopathological score both in early [SMD, 6.39; 95%CI (2.90, 9.87); p = 0.0003] and late [SMD, 3.41; 95%CI (2.44, 4.38); p < 0.00001] stage of OA. Integrin α5β1 also increased the core catabolic factors like MMP-3, IL-1β, and TNF-α. Interestingly, the inactivation of α5β1 integrin did not change the histopathological score (p = 0.84). Similarly, β1 integrin notably increased histopathological score at both stages of OA [early; SMD, 7.13; 95%CI (2.01, 12.24); p = 0.006]; [late; SMD, 10.25; 95%CI (5.11, 15.39); p < 0.0001], and increased the MMP-13 levels. However, integrin β1 was upregulated at the early stage and downregulated at the late stage of OA. Furthermore, α2β1 integrin significantly increased histopathological score [SMD, 3.14; 95%CI (2.18, 4.10); p < 0.00001] and MMP-13 [SMD, 2.24; 95%CI (0.07, 4.41); p = 0.04]. Deactivating integrin α1β1 increased histopathological score in late [SMD, 1.53; 95%CI (0.80, 2.26); p < 0.0001], but not in early [SMD, 0.90; 95%CI (-1.65, 3.45); p = 0.49] stage of OA. CONCLUSION This study provides evidence that α5β1, α2β1, and α1β1 integrin might be the potential target for future drug development in alleviating OA pathogenesis. Further work is required to establish our findings through activating/deactivating these receptors in different stages of OA.
Collapse
Affiliation(s)
- Dewan Md Sumsuzzman
- Department of Physical Therapy, College of Healthcare Medical Science & Engineering, Gimhae 50834, Republic of Korea; Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae 50834, Republic of Korea; Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Republic of Korea.
| | - Zeeshan Ahmad Khan
- Department of Physical Therapy, College of Healthcare Medical Science & Engineering, Gimhae 50834, Republic of Korea; Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae 50834, Republic of Korea; Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Republic of Korea.
| | - Jeonghyun Choi
- Department of Physical Therapy, College of Healthcare Medical Science & Engineering, Gimhae 50834, Republic of Korea; Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae 50834, Republic of Korea; Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Republic of Korea.
| | - Yonggeun Hong
- Department of Physical Therapy, College of Healthcare Medical Science & Engineering, Gimhae 50834, Republic of Korea; Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae 50834, Republic of Korea; Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Republic of Korea; Department of Rehabilitation Science, Graduate School of Inje University, Gimhae 50834, Republic of Korea.
| |
Collapse
|
20
|
Liang C, Liu X, Yan Y, Sun R, Li J, Geng W. Effectiveness and Mechanisms of Low-Intensity Pulsed Ultrasound on Osseointegration of Dental Implants and Biological Functions of Bone Marrow Mesenchymal Stem Cells. Stem Cells Int 2022; 2022:7397335. [PMID: 36199628 PMCID: PMC9529500 DOI: 10.1155/2022/7397335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/09/2022] [Indexed: 11/27/2022] Open
Abstract
Dental implant restoration is the preferred choice for patients with dentition defects or edentulous patients, and obtaining stable osseointegration is the determining factor for successful implant healing. The risk of implant failure during the healing stage is still an urgent problem in clinical practice due to differences in bone quality at different implant sites and the impact of some systemic diseases on bone tissue metabolism. Low-intensity pulsed ultrasound (LIPUS) is a noninvasive physical intervention method widely recognized in the treatment of bone fracture and joint damage repair. Moreover, many studies indicated that LIPUS could effectively promote the osseointegration of dental implants and improve the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). This review is aimed at investigating the research progress on the use of LIPUS in dental implant medicine from three aspects: (1) discuss the promoting effects of LIPUS on osseointegration and peri-implant bone regeneration, (2) summarize the effects and associated mechanisms of LIPUS on the biological functions of BMSCs, and (3) introduce the application and prospects of LIPUS in the clinical work of dental implantation. Although many challenges need to be overcome in the future, LIPUS is bound to be an efficient and convenient therapeutic method to improve the dental implantation success rate and expand clinical implant indications.
Collapse
Affiliation(s)
- Chao Liang
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Xiu Liu
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Yuwei Yan
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Rongxin Sun
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Jun Li
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
- Beijing Institute of Dental Research, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| | - Wei Geng
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing 100050, China
| |
Collapse
|
21
|
Germain P, Delalande A, Pichon C. Role of Muscle LIM Protein in Mechanotransduction Process. Int J Mol Sci 2022; 23:ijms23179785. [PMID: 36077180 PMCID: PMC9456170 DOI: 10.3390/ijms23179785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/14/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022] Open
Abstract
The induction of protein synthesis is crucial to counteract the deconditioning of neuromuscular system and its atrophy. In the past, hormones and cytokines acting as growth factors involved in the intracellular events of these processes have been identified, while the implications of signaling pathways associated with the anabolism/catabolism ratio in reference to the molecular mechanism of skeletal muscle hypertrophy have been recently identified. Among them, the mechanotransduction resulting from a mechanical stress applied to the cell appears increasingly interesting as a potential pathway for therapeutic intervention. At present, there is an open question regarding the type of stress to apply in order to induce anabolic events or the type of mechanical strain with respect to the possible mechanosensing and mechanotransduction processes involved in muscle cells protein synthesis. This review is focused on the muscle LIM protein (MLP), a structural and mechanosensing protein with a LIM domain, which is expressed in the sarcomere and costamere of striated muscle cells. It acts as a transcriptional cofactor during cell proliferation after its nuclear translocation during the anabolic process of differentiation and rebuilding. Moreover, we discuss the possible opportunity of stimulating this mechanotransduction process to counteract the muscle atrophy induced by anabolic versus catabolic disorders coming from the environment, aging or myopathies.
Collapse
Affiliation(s)
- Philippe Germain
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
| | - Anthony Delalande
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
| | - Chantal Pichon
- UFR Sciences and Techniques, University of Orleans, 45067 Orleans, France
- Center for Molecular Biophysics, CNRS Orleans, 45071 Orleans, France
- Institut Universitaire de France, 1 Rue Descartes, 75231 Paris, France
- Correspondence:
| |
Collapse
|
22
|
Xia P, Shi Y, Wang X, Li X. Advances in the application of low-intensity pulsed ultrasound to mesenchymal stem cells. Stem Cell Res Ther 2022; 13:214. [PMID: 35619156 PMCID: PMC9137131 DOI: 10.1186/s13287-022-02887-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/03/2022] [Indexed: 11/10/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are stem cells that exhibit self-renewal capacity and multi-directional differentiation potential. They can be extracted from the bone marrow and umbilical cord, as well as adipose, amnion, and other tissues. They are widely used in tissue engineering and are currently considered an important source of cells in the field of regenerative medicine. Since certain limitations, such as an insufficient cell source, mature differentiation, and low transplantation efficiency, are still associated with MSCs, researchers have currently focused on improving the efficacy of MSCs. Low-intensity pulsed ultrasound (LIPUS) has mechanical, cavitation, and thermal effects that can produce different biological effects on organs, tissues, and cells. It can be used for fracture treatment, cartilage repair, and stem cell applications. An in-depth study of the role and mechanism of action of LIPUS in MSC treatment would promote our understanding of LIPUS and promote research in this field. In this article, we have reviewed the progress in research on the use of LIPUS with various MSCs and comprehensively discussed the progress in the use of LIPUS for promoting the proliferation, differentiation, and migration of MSCs, as well as its future prospects.
Collapse
Affiliation(s)
- Peng Xia
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Yi Shi
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaoju Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xueping Li
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
23
|
Liao YH, Chen MX, Chen SC, Luo KX, Wang B, Ao LJ, Liu Y. Low-Intensity Focused Ultrasound Alleviates Spasticity and Increases Expression of the Neuronal K-Cl Cotransporter in the L4–L5 Sections of Rats Following Spinal Cord Injury. Front Cell Neurosci 2022; 16:882127. [PMID: 35634464 PMCID: PMC9133482 DOI: 10.3389/fncel.2022.882127] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Low-intensity focused ultrasound (LIFU) has been shown to provide effective activation of the spinal cord neurocircuits. The aim of this study was to investigate the effects of LIFU in order to alleviate spasticity following spinal cord injury (SCI) by activating the spinal neurocircuits and increasing the expression of the neuronal K-Cl cotransporter KCC2. Adult male Sprague Dawley (SD) rats (220–300 g) were randomly divided into a sham control group, a LIFU− group, and a LIFU+ group. The mechanical threshold hold (g) was used to evaluate the behavioral characteristics of spasm. Electromyography (EMG) was used to assess activation of the spinal cord neurocircuits and muscle spontaneous contraction. Spasticity was assessed by frequency-dependent depression (FDD). The expression of KCC2 of the lumbar spinal cord was determined via western blot (WB) and immunofluorescence (IF) staining. The spinal cord neurocircuits were activated by LIFU simulation, which significantly reduced the mechanical threshold (g), FDD, and EMG recordings (s) after 4 weeks of treatment. WB and IF staining both demonstrated that the expression of KCC2 was reduced in the LIFU− group (P < 0.05). After 4 weeks of LIFU stimulation, expression of KCC2 had significantly increased (P < 0.05) in the LIFU+ group compared with the LIFU− group. Thus, we hypothesized that LIFU treatment can alleviate spasticity effectively and upregulate the expression of KCC2 in the L4–L5 section of SCI rats.
Collapse
Affiliation(s)
- Ye-Hui Liao
- School of Rehabilitation, Kunming Medical University, Kunming, China
- Department of Orthopaedics, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Mo-Xian Chen
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | - Shao-Chun Chen
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | - Kai-Xuan Luo
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | - Bing Wang
- School of Rehabilitation, Kunming Medical University, Kunming, China
| | - Li-Juan Ao
- School of Rehabilitation, Kunming Medical University, Kunming, China
- *Correspondence: Li-Juan Ao
| | - Yao Liu
- School of Rehabilitation, Kunming Medical University, Kunming, China
- Yao Liu
| |
Collapse
|
24
|
Acheta J, Stephens SBZ, Belin S, Poitelon Y. Therapeutic Low-Intensity Ultrasound for Peripheral Nerve Regeneration – A Schwann Cell Perspective. Front Cell Neurosci 2022; 15:812588. [PMID: 35069118 PMCID: PMC8766802 DOI: 10.3389/fncel.2021.812588] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/09/2021] [Indexed: 01/22/2023] Open
Abstract
Peripheral nerve injuries are common conditions that can arise from trauma (e.g., compression, severance) and can lead to neuropathic pain as well as motor and sensory deficits. Although much knowledge exists on the mechanisms of injury and nerve regeneration, treatments that ensure functional recovery following peripheral nerve injury are limited. Schwann cells, the supporting glial cells in peripheral nerves, orchestrate the response to nerve injury, by converting to a “repair” phenotype. However, nerve regeneration is often suboptimal in humans as the repair Schwann cells do not sustain their repair phenotype long enough to support the prolonged regeneration times required for successful nerve regrowth. Thus, numerous strategies are currently focused on promoting and extending the Schwann cells repair phenotype. Low-intensity ultrasound (LIU) is a non-destructive therapeutic approach which has been shown to facilitate peripheral nerve regeneration following nerve injury in rodents. Still, clinical trials in humans are scarce and limited to small population sizes. The benefit of LIU on nerve regeneration could possibly be mediated through the repair Schwann cells. In this review, we discuss the known and possible molecular mechanisms activated in response to LIU in repair Schwann cells to draw support and attention to LIU as a compelling regenerative treatment for peripheral nerve injury.
Collapse
|
25
|
Du S, Liang C, Sun Y, Ma B, Gao W, Geng W. The Attenuating Effect of Low-Intensity Pulsed Ultrasound on Hypoxia-Induced Rat Chondrocyte Damage in TMJ Osteoarthritis Based on TMT Labeling Quantitative Proteomic Analysis. Front Pharmacol 2022; 12:752734. [PMID: 34970140 PMCID: PMC8712703 DOI: 10.3389/fphar.2021.752734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Temporomandibular joint osteoarthritis (TMJOA) is a degenerative disease with a complex and multifactorial etiology. An increased intrajoint pressure or weakened penetration can exacerbate the hypoxic state of the condylar cartilage microenvironment. Our group previously simulated the hypoxic environment of TMJOA in vitro. Low-intensity pulsed ultrasound (LIPUS) stimulation attenuates chondrocyte matrix degradation via a hypoxia-inducible factor (HIF) pathway-associated mechanism, but the mode of action of LIPUS is currently poorly understood. Moreover, most recent studies investigated the pathological mechanisms of osteoarthritis, but no biomarkers have been established for assessing the therapeutic effect of LIPUS on TMJOA with high specificity, which results in a lack of guidance regarding clinical application. Here, tandem mass tag (TMT)-based quantitative proteomic technology was used to comprehensively screen the molecular targets and pathways affected by the action of LIPUS on chondrocytes under hypoxic conditions. A bioinformatic analysis identified 902 and 131 differentially expressed proteins (DEPs) in the <1% oxygen treatment group compared with the control group and in the <1% oxygen + LIPUS stimulation group compared with the <1% oxygen treatment group, respectively. The DEPs were analyzed by gene ontology (GO), KEGG pathway and protein-protein interaction (PPI) network analyses. By acting on extracellular matrix (ECM)-associated proteins, LIPUS increases energy production and activates the FAK signaling pathway to regulate cell biological behaviors. DEPs of interest were selected to verify the reliability of the proteomic results. In addition, this experiment demonstrated that LIPUS could upregulate chondrogenic factors (such as Sox9, Collagen Ⅱ and Aggrecan) and increase the mucin sulfate content. Moreover, LIPUS reduced the hydrolytic degradation of the ECM by decreasing the MMP3/TIMP1 ratio and vascularization by downregulating VEGF. Interestingly, LIPUS improved the migration ability of chondrocytes. In summary, LIPUS can regulate complex biological processes in chondrocytes under hypoxic conditions and alter the expression of many functional proteins, which results in reductions in hypoxia-induced chondrocyte damage. ECM proteins such as thrombospondin4, thrombospondin1, IL1RL1, and tissue inhibitors of metalloproteinase 1 play a central role and can be used as specific biomarkers determining the efficacy of LIPUS and viable clinical therapeutic targets of TMJOA.
Collapse
Affiliation(s)
- Sa Du
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Chao Liang
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yujie Sun
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Bowen Ma
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Wenmo Gao
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Wei Geng
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Wang N, Zhang J, Lv H, Liu Z. Regulation of COL1A2, AKT3 genes, and related signaling pathway in the pathology of congenital talipes equinovarus. Front Pediatr 2022; 10:890109. [PMID: 35935376 PMCID: PMC9355787 DOI: 10.3389/fped.2022.890109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Congenital talipes equinovarus (CTEV) is one of the most common congenital limb defects in children, which is a multifactorial and complex disease that associates with many unknown genetic, social-demographic, and environmental risk factors. Emerging evidence proved that gene expression or mutation might play an important role in the occurrence and development of CTEV. However, the underlying reasons and involved mechanisms are still not clear. Herein, to probe the potential genes and related signaling pathways involved in CTEV, we first identified the differentially expressed genes (DEGs) by mRNA sequencing in pediatric patients with CTEV compared with normal children. The gene of COL1A2 was upregulated, and AKT3 was downregulated at the transcriptional level. Western blot and quantitative polymerase chain reaction (qRT-PCR) results also showed that the expression of COL1A2 in CTEV was enhanced, and the AKT3 was decreased. Furthermore, the COL1A2 Knock-in (+COL1A2) and AKT3 Knock-out (-AKT3) transgenic mice were used to verify the effects of these two genes in the CTEV, and the results of which showed that both COL1A2 and AKT3 were closely related to the CTEV. We also investigated the effect of the PI3K-AKT3 signaling pathway in CTEV by measuring the relative expression of several key genes using Western blot and qRT-PCR. In line with the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis data, the PI3K-AKT3 signaling pathway might play a potentially important role in the regulation of pathological changes of CTEV. This study will provide new ideas for the mechanism investigation and prenatal diagnosis of CTEV.
Collapse
Affiliation(s)
- Ningqing Wang
- Department of Orthopedics, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Jiangchao Zhang
- Department of Orthopedics, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Haixiang Lv
- Department of Orthopedics, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Zhenjiang Liu
- Department of Orthopedics, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
27
|
Jin H, Jiang S, Wang R, Zhang Y, Dong J, Li Y. Mechanistic Insight Into the Roles of Integrins in Osteoarthritis. Front Cell Dev Biol 2021; 9:693484. [PMID: 34222261 PMCID: PMC8250141 DOI: 10.3389/fcell.2021.693484] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 05/19/2021] [Indexed: 01/13/2023] Open
Abstract
Osteoarthritis (OA), one of the most common degenerative diseases, is characterized by progressive degeneration of the articular cartilage and subchondral bone, as well as the synovium. Integrins, comprising a family of heterodimeric transmembrane proteins containing α subunit and β subunit, play essential roles in various physiological functions of cells, such as cell attachment, movement, growth, differentiation, and mechanical signal conduction. Previous studies have shown that integrin dysfunction is involved in OA pathogenesis. This review article focuses on the roles of integrins in OA, especially in OA cartilage, subchondral bone and the synovium. A clear understanding of these roles may influence the future development of treatments for OA.
Collapse
Affiliation(s)
- Hongfu Jin
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Shigang Jiang
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ruomei Wang
- Department of Endocrinology and Metabolic Diseases, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi Zhang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jiangtao Dong
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
28
|
Santana-Rodríguez N, Clavo B, Llontop P, Fiuza MD, Calatayud-Gastardi J, López D, López-Fernández D, Aguiar-Santana IA, Ayub A, Alshehri K, Jordi NA, Zubeldia J, Bröering DC. Pulsed Ultrasounds Reduce Pain and Disability, Increasing Rib Fracture Healing, in a Randomized Controlled Trial. PAIN MEDICINE 2020; 20:1980-1988. [PMID: 30496510 DOI: 10.1093/pm/pny224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Rib fractures are an important health issue worldwide, with significant, pain, morbidity, and disability for which only symptomatic treatment exists. OBJECTIVES Based on our previous experimental model, the objective of the current study was to assess for the first time whether pulsed ultrasound (PUS) application could have beneficial effects on humans. METHODS Prospective, double-blinded, randomized, controlled trial of 51 patients. Four were excluded, and 47 were randomized into the control group (N = 23) or PUS group (N = 24). The control group received a PUS procedure without emission, and the PUS group received 1 Mhz, 0.5 W/cm2 for 1 min/cm2. Pain level, bone callus healing rate, physical and work activity, pain medication intake, and adverse events were blindly evaluated at baseline and one, three, and six months. RESULTS There were no significant differences at baseline between groups. PUS treatment significantly decreased pain by month 1 (P = 0.004), month 3 (P = 0.005), and month 6 (P = 0.025), significantly accelerated callus healing by month 1 (P = 0.013) and month 3 (P < 0.001), accelerated return to physical activity by month 3 (P = 0.036) and work activity (P = 0.001) by month 1, and considerably reduced pain medication intake by month 1 (P = 0.057) and month 3 (P = 0.017). No related adverse events were found in the PUS group. CONCLUSIONS This study is the first evidence that PUS treatment is capable of improving rib fracture outcome, significantly accelerating bone callus healing, and decreasing pain, time off due to both physical activity and convalescence period, and pain medication intake. It is a safe, efficient, and low-cost therapy that may become a new treatment for patients with stable rib fractures.
Collapse
Affiliation(s)
- Norberto Santana-Rodríguez
- Section of Thoracic Surgery, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia.,Department of Surgery, College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia.,Instituto Universitario de Investigaciones Biomédicas y Sanitarias IUIBS-BioPharm Group, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Bernardino Clavo
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias IUIBS-BioPharm Group, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain.,Experimental Surgery Group, Research Unit, Hospital Dr. Negrín, Las Palmas de Gran Canaria, Las Palmas, Spain.,Chronic Pain Unit, Hospital Dr. Negrín, Las Palmas de Gran Canaria, Las Palmas, Spain.,Department of Radiation Oncology, Hospital Dr. Negrín, Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Pedro Llontop
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias IUIBS-BioPharm Group, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain.,Experimental Medicine and Surgery Unit of Hospital Gregorio Marañón and the Health Research Institute of Hospital Gregorio Marañón IiSGM, Madrid, Spain
| | - María D Fiuza
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias IUIBS-BioPharm Group, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain.,Experimental Surgery Group, Research Unit, Hospital Dr. Negrín, Las Palmas de Gran Canaria, Las Palmas, Spain
| | | | - Daniel López
- Experimental Surgery Group, Research Unit, Hospital Dr. Negrín, Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Daniel López-Fernández
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias IUIBS-BioPharm Group, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain.,Experimental Surgery Group, Research Unit, Hospital Dr. Negrín, Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Ione A Aguiar-Santana
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias IUIBS-BioPharm Group, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain.,Experimental Surgery Group, Research Unit, Hospital Dr. Negrín, Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Adil Ayub
- Department of Surgery, University of Texas Medical Branch Galveston, Galveston, Texas, USA
| | - Khalid Alshehri
- Section of Thoracic Surgery, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Nagib A Jordi
- Department of Orthopedic Surgery and Upper Extremity Unit, Healthpoint Hospital, Abu Dhabi, UAE
| | - José Zubeldia
- Experimental Surgery Group, Research Unit, Hospital Dr. Negrín, Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Dieter C Bröering
- Section of Thoracic Surgery, Department of Surgery, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia.,Department of Surgery, College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
29
|
Yanoshita M, Hirose N, Sumi C, Takano M, Nishiyama S, Tsuboi E, Onishi A, Yamauchi Y, Asakawa Y, Ryo K, Tanimoto K. FAK inhibition protects condylar cartilage under excessive mechanical stress. Oral Dis 2020; 26:1736-1746. [DOI: 10.1111/odi.13494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/28/2020] [Accepted: 06/08/2020] [Indexed: 11/28/2022]
Affiliation(s)
- Makoto Yanoshita
- Department of Orthodontics and Craniofacial Developmental Biology Hiroshima University Graduate School of Biomedical and Health Sciences Hiroshima prefecture Japan
| | - Naoto Hirose
- Department of Orthodontics and Craniofacial Developmental Biology Hiroshima University Graduate School of Biomedical and Health Sciences Hiroshima prefecture Japan
| | - Chikako Sumi
- Department of Orthodontics and Craniofacial Developmental Biology Hiroshima University Graduate School of Biomedical and Health Sciences Hiroshima prefecture Japan
| | - Mami Takano
- Department of Orthodontics and Craniofacial Developmental Biology Hiroshima University Graduate School of Biomedical and Health Sciences Hiroshima prefecture Japan
| | - Sayuri Nishiyama
- Department of Orthodontics and Craniofacial Developmental Biology Hiroshima University Graduate School of Biomedical and Health Sciences Hiroshima prefecture Japan
| | - Eri Tsuboi
- Department of Orthodontics and Craniofacial Developmental Biology Hiroshima University Graduate School of Biomedical and Health Sciences Hiroshima prefecture Japan
| | - Azusa Onishi
- Department of Orthodontics and Craniofacial Developmental Biology Hiroshima University Graduate School of Biomedical and Health Sciences Hiroshima prefecture Japan
| | - Yuka Yamauchi
- Department of Orthodontics and Craniofacial Developmental Biology Hiroshima University Graduate School of Biomedical and Health Sciences Hiroshima prefecture Japan
| | - Yuki Asakawa
- Department of Orthodontics and Craniofacial Developmental Biology Hiroshima University Graduate School of Biomedical and Health Sciences Hiroshima prefecture Japan
| | - Kunimatsu Ryo
- Department of Orthodontics and Craniofacial Developmental Biology Hiroshima University Graduate School of Biomedical and Health Sciences Hiroshima prefecture Japan
| | - Kotaro Tanimoto
- Department of Orthodontics and Craniofacial Developmental Biology Hiroshima University Graduate School of Biomedical and Health Sciences Hiroshima prefecture Japan
| |
Collapse
|
30
|
Liu Q, Zeng H, Yuan Y, Wang Z, Wu Z, Luo W. Osteopontin inhibits osteoarthritis progression via the OPN/CD44/PI3K signal axis. Genes Dis 2020; 9:128-139. [PMID: 35005113 PMCID: PMC8720673 DOI: 10.1016/j.gendis.2020.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/26/2020] [Accepted: 06/12/2020] [Indexed: 12/23/2022] Open
Abstract
Chondrocyte degeneration and extracellular matrix component loss are the primary causes of osteoarthritis (OA). OA can be treated by inhibiting chondrocyte degeneration and increasing extracellular matrix component secretion. Osteopontin (OPN), a multifunctional protein, has gained immense attention with regard to its involvement in OA. This study aimed to explore the therapeutic value and mechanism of action of OPN in OA treatment. Results of the histomorphological analysis revealed a worn-off OA cartilage tissue surface, cartilage matrix layer deterioration, and calcium salt deposition. Compared to that in normal chondrocytes, in OA chondrocytes, the OPN, CD44, and PI3K protein and mRNA expression was upregulated. Further, siOPN, rhOPN, and rhOPN plus LS-C179404 interfered with OA chondrocytes. As verified in mice, OPN directly inhibited the expression level of PI3K in OA chondrocytes by binding with CD44. Morphological analysis of the knee joints demonstrated that OPN effectively inhibited OA progression via the OPN/CD44/PI3K signal axis. In conclusion, OPN activates intracellular PI3K signaling molecules by binding to CD44 on the cell surface to cause downstream cascading effects, thereby delaying chondrocyte degeneration and reducing cartilage matrix component loss; therefore, OPN is a potential therapeutic agent for OA.
Collapse
Affiliation(s)
- Qing Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87th Xiangya Road, Changsha, Hunan 410008, PR China.,Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139th Renmin Middle Road, Changsha, Hunan 410011, PR China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87th Xiangya Road, Changsha, Hunan 410008, PR China
| | - Yuhao Yuan
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87th Xiangya Road, Changsha, Hunan 410008, PR China
| | - Zhiwei Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87th Xiangya Road, Changsha, Hunan 410008, PR China
| | - Ziyi Wu
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87th Xiangya Road, Changsha, Hunan 410008, PR China
| | - Wei Luo
- Department of Orthopaedics, Xiangya Hospital, Central South University, 87th Xiangya Road, Changsha, Hunan 410008, PR China
| |
Collapse
|
31
|
Lei J, Amhare AF, Wang L, Lv Y, Deng H, Gao H, Guo X, Han J, Lammi MJ. Proteomic analysis of knee cartilage reveals potential signaling pathways in pathological mechanism of Kashin-Beck disease compared with osteoarthritis. Sci Rep 2020; 10:6824. [PMID: 32322000 PMCID: PMC7176695 DOI: 10.1038/s41598-020-63932-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 04/03/2020] [Indexed: 12/31/2022] Open
Abstract
The pathological mechanism of Kashin-Beck disease (KBD), an endemic osteoarthritic disease, remains to be poorly understood. This study was designed to identify signaling pathways and crucial proteins involved in the pathological mechanism of KBD compared with osteoarthritis (OA). The knee cartilage samples were collected from gender- and age-matched KBD (n = 9) and OA (n = 9) patients. After pre-processing, samples were labeled with Tamdem Mass Tags 6plex multiplex kit, and analyzed by liquid chromatography-tandem mass spectrometry. Proteomic results were analyzed with gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and protein-protein interactions (PPI). The differential abundance proteins from KBD and OA were validated using western blot analysis. As a result, A total number of 375 proteins were identified to have differential abundance between KBD and OA, of which 121 and 254 proteins were observed to be up-regulated or down-regulated in KBD group. GO analysis shows that the differential abundant proteins are associated with cell junction and signal transducer activity from extracellular to intracellular. KEGG pathways enrichment and PPI network indicate four major pathways, including extracellular matrix -receptor interaction, focal adhesion, phosphatidylinositol 3-kinase (PI3K)-Protein kinase B (Akt), and Ras signaling pathways were involved in the degeneration of cartilage. Moreover, integrins, laminins, NF-κB and other regulative molecules were found as crucial proteins. In conclusion, our results demonstrated that compared with OA, the differential abundance proteins and signaling pathways may contribute to the occurrence and development of joint damage in KBD. Further investigation of their regulative roles and interaction may provide new insights into the pathological mechanisms and therapeutic targets for KBD.
Collapse
Affiliation(s)
- Jian Lei
- School of Public Health, Health Science Center; Key Laboratory of Environment and Gene Related Diseases of Ministry Education; Key Laboratory of Trace Elements and Endemic Diseases, Ministry of Health, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China.,Shenzhen Institute, Xi'an Jiaotong University, Shenzhen, Guangdong, 518057, P. R. China
| | - Abebe Feyissa Amhare
- School of Public Health, Health Science Center; Key Laboratory of Environment and Gene Related Diseases of Ministry Education; Key Laboratory of Trace Elements and Endemic Diseases, Ministry of Health, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China
| | - Liyun Wang
- School of Public Health, Health Science Center; Key Laboratory of Environment and Gene Related Diseases of Ministry Education; Key Laboratory of Trace Elements and Endemic Diseases, Ministry of Health, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China.,Shenzhen Institute, Xi'an Jiaotong University, Shenzhen, Guangdong, 518057, P. R. China
| | - Yizhen Lv
- School of Public Health, Health Science Center; Key Laboratory of Environment and Gene Related Diseases of Ministry Education; Key Laboratory of Trace Elements and Endemic Diseases, Ministry of Health, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China
| | - Huan Deng
- School of Public Health, Health Science Center; Key Laboratory of Environment and Gene Related Diseases of Ministry Education; Key Laboratory of Trace Elements and Endemic Diseases, Ministry of Health, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China
| | - Hang Gao
- Laboratory of Resource Biology and Biotechnology in Western China (Ministry of Education), Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, Shaanxi, 710069, P. R. China
| | - Xiong Guo
- School of Public Health, Health Science Center; Key Laboratory of Environment and Gene Related Diseases of Ministry Education; Key Laboratory of Trace Elements and Endemic Diseases, Ministry of Health, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China
| | - Jing Han
- School of Public Health, Health Science Center; Key Laboratory of Environment and Gene Related Diseases of Ministry Education; Key Laboratory of Trace Elements and Endemic Diseases, Ministry of Health, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China. .,Shenzhen Institute, Xi'an Jiaotong University, Shenzhen, Guangdong, 518057, P. R. China.
| | - Mikko J Lammi
- School of Public Health, Health Science Center; Key Laboratory of Environment and Gene Related Diseases of Ministry Education; Key Laboratory of Trace Elements and Endemic Diseases, Ministry of Health, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P. R. China.,Department of Integrative Medical Biology, Umeå University, Umeå, 90187, Sweden
| |
Collapse
|
32
|
Uddin SMZ, Komatsu DE. Therapeutic Potential Low-Intensity Pulsed Ultrasound for Osteoarthritis: Pre-clinical and Clinical Perspectives. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:909-920. [PMID: 31959508 DOI: 10.1016/j.ultrasmedbio.2019.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/27/2019] [Accepted: 12/06/2019] [Indexed: 06/10/2023]
Abstract
Osteoarthritis (OA), degeneration of cartilage associated with aging, lifestyle, and trauma, is one of the most common diseases that leads to lower quality of life and socioeconomic burden in the United States. Clinically, OA is initially managed by non-steroidal anti-inflammatory drugs, but eventually requires surgical intervention to reduce pain and increase function. Cartilage is a mechanotransductive tissue and requires a mechanical stimulus to sustain its mechanical and physiologic properties. Low-intensity pulsed ultrasound (LIPUS) is a cyclic acoustic wave that can provide essential mechanical stimuli to activate molecular and cellular pathways leading to chondrocyte proliferation, differentiation and activity, as well as to inhibit inflammatory pathways associated with OA. The activation of chondrocyte proliferation and inhibition of anti-inflammatory cytokines make LIPUS a potential therapy for mild to moderate OA. Although a few review articles have described the effects of ultrasound on chondrocytes and cartilage, there remains a need for a comprehensive analysis of our current understanding of the basic science and clinical status of the effects of low-intensity ultrasound on chondrocytes and cartilage and the implications of these studies on LIPUS as a therapeutic option for OA. This review analyzes recent literature describing the results of LIPUS using in vitro and in vivo pre-clinical models and clinical studies, as well as future directions for research.
Collapse
Affiliation(s)
- Sardar M Z Uddin
- Department of Orthopaedics, Stony Brook University, Stony Brook, New York, USA.
| | - David E Komatsu
- Department of Orthopaedics, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
33
|
Martínez-Moreno D, Jiménez G, Gálvez-Martín P, Rus G, Marchal JA. Cartilage biomechanics: A key factor for osteoarthritis regenerative medicine. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1067-1075. [PMID: 30910703 DOI: 10.1016/j.bbadis.2019.03.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 03/18/2019] [Accepted: 03/20/2019] [Indexed: 11/26/2022]
Abstract
Osteoarthritis (OA) is a joint disorder that is highly extended in the global population. Several researches and therapeutic strategies have been probed on OA but without satisfactory long-term results in joint replacement. Recent evidences show how the cartilage biomechanics plays a crucial role in tissue development. This review describes how physics alters cartilage and its extracellular matrix (ECM); and its role in OA development. The ECM of the articular cartilage (AC) is widely involved in cartilage turnover processes being crucial in regeneration and joint diseases. We also review the importance of physicochemical pathways following the external forces in AC. Moreover, new techniques probed in cartilage tissue engineering for biomechanical stimulation are reviewed. The final objective of these novel approaches is to create a cellular implant that maintains all the biochemical and biomechanical properties of the original tissue for long-term replacements in patients with OA.
Collapse
Affiliation(s)
- D Martínez-Moreno
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada E-18100, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain
| | - G Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada E-18100, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain; Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada E-18071, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada E-18016, Spain
| | - P Gálvez-Martín
- Advanced Therapies Area, Pharmascience Division, Bioibérica S.A.U., E-08029 Barcelona, Spain; Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Granada E-18071, Spain
| | - G Rus
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada E-18100, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain; Department of Structural Mechanics, University of Granada, Politécnico de Fuentenueva, Granada E-18071, Spain.
| | - J A Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada E-18100, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain; Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada E-18071, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada E-18016, Spain.
| |
Collapse
|
34
|
Xia B, Chen G, Zou Y, Yang L, Pan J, Lv Y. Low-intensity pulsed ultrasound combination with induced pluripotent stem cells-derived neural crest stem cells and growth differentiation factor 5 promotes sciatic nerve regeneration and functional recovery. J Tissue Eng Regen Med 2019; 13:625-636. [PMID: 30770650 DOI: 10.1002/term.2823] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 01/13/2019] [Accepted: 02/13/2019] [Indexed: 12/22/2022]
Abstract
The treatment of lengthy peripheral nerve defect is challenging in the field of nerve regeneration. Our previous studies have shown that low-intensity pulsed ultrasound (LIPUS) could promote the proliferation, cell viability, and neural differentiation of induced pluripotent stem cells-derived neural crest stem cells (iPSCs-NCSCs) and improve the regeneration of damaged peripheral nerve. In this study, the mechanical signal transduction pathway of LIPUS promoting iPSCs-NCSCs proliferation and differentiation was further explored, and the effects of LIPUS combined with iPSCs-NCSCs, perfluorotributylamine (PFTBA), and growth differentiation factor 5 (GDF5) on the repair of peripheral nerve injury were evaluated. Results showed LIPUS may regulate the proliferation and differentiation of iPSCs-NCSCs through FAK-ERK1/2 signal pathway. PFTBA could supply sufficient oxygen to promote the viability of iPSCs-NCSCs under 5% hypoxia culture condition and provide a favourable microenvironment for nerve regeneration. The addition of GDF5 could promote the neural differentiation of iPSCs-NCSCs in vitro. LIPUS treatment of allogeneic decellularized nerve conduit containing iPSCs-NCSCs, PFTBA, and GDF5 has very good effect on the repair of sciatic nerve injury. Taken together, these results provide functional evidence that LIPUS might be a useful tool to explore alternative approaches in the field of nerve regeneration.
Collapse
Affiliation(s)
- Bin Xia
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China.,Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| | - Guobao Chen
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China.,Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| | - Yang Zou
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China.,Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China.,Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| | - Jun Pan
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China.,Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| | - Yonggang Lv
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Bioengineering College, Chongqing University, Chongqing, China.,Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| |
Collapse
|
35
|
Wang X, Lin Q, Zhang T, Wang X, Cheng K, Gao M, Xia P, Li X. Low-intensity pulsed ultrasound promotes chondrogenesis of mesenchymal stem cells via regulation of autophagy. Stem Cell Res Ther 2019; 10:41. [PMID: 30670079 PMCID: PMC6343259 DOI: 10.1186/s13287-019-1142-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 12/28/2018] [Accepted: 01/08/2019] [Indexed: 02/07/2023] Open
Abstract
Background Low-intensity pulsed ultrasound (LIPUS) can induce mesenchymal stem cell (MSC) differentiation, although the mechanism of its potential effects on chondrogenic differentiation is unknown. Since autophagy is known to regulate the differentiation of MSCs, the aim of our study was to determine whether LIPUS induced chondrogenesis via autophagy regulation. Methods MSCs were isolated from the rat bone marrow, cultured in either standard or chondrogenic medium, and stimulated with 3 MHz of LIPUS given in 20% on–off cycles, with or without prior addition of an autophagy inhibitor or agonist. Chondrogenesis was evaluated on the basis of aggrecan (AGG) organization and the amount of type II collagen (COL2) and the mRNA expression of AGG, COL2, and SRY-related high mobility group-box gene 9 (SOX9) genes. Results LIPUS promoted the chondrogenic differentiation of MSCs, as shown by the changes in the extracellular matrix (ECM) proteins and upregulation of chondrogenic genes, and these effects were respectively augmented and inhibited by the autophagy inhibitor and agonist. Conclusions Taken together, these results indicate that LIPUS promotes MSC chondrogenesis by inhibiting autophagy.
Collapse
Affiliation(s)
- Xiaoju Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Qiang Lin
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Tingting Zhang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xinwei Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Kai Cheng
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Mingxia Gao
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Peng Xia
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Xueping Li
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
36
|
Li X, Sun Y, Zhou Z, Zhang D, Jiao J, Hu M, Hassan CR, Qin YX. Mitigation of Articular Cartilage Degeneration and Subchondral Bone Sclerosis in Osteoarthritis Progression Using Low-Intensity Ultrasound Stimulation. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:148-159. [PMID: 30322672 PMCID: PMC6289639 DOI: 10.1016/j.ultrasmedbio.2018.08.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 08/24/2018] [Accepted: 08/28/2018] [Indexed: 06/08/2023]
Abstract
The purpose of this study was to evaluate the effect of low-intensity ultrasound on articular cartilage and subchondral bone alterations in joints under normal and functional disuse conditions during osteoarthritis (OA) progression. Total of thirty 5-mo-old female Sprague-Dawley rats were randomly assigned to six groups (n = 5/group): age-matched group, OA group, OA + ultrasound (US) group, hindlimb suspension (HLS) group, HLS + OA group and HLS + OA + US group. The surgical anterior cruciate ligament was used to induce OA in the right knee joints. After 2 wk of OA induction, low-intensity ultrasound generated with a 3-MHz transducer with 20% pulse duty cycle and 30 mW/cm2 acoustic intensity was delivered to the right knee joints for 20 min a day, 5 d a week for a total of 6 wk. Then, the right tibias were harvested for micro-computed tomography, histologic and mechanical analysis. Micro-computed tomography results indicated that the thickness and sulfated glycosaminoglycan content of cartilage decreased, but the thickness of the subchondral cortical bone plate and the formation of subchondral trabecular bone increased in the OA group under the normal joint use condition. Furthermore, histologic results revealed that chondrocyte density and arrangement in cartilage corrupted and the underlying subchondral bone increased during OA progression. These changes were accompanied by reductions in mechanical parameters in OA cartilage. However, fewer OA symptoms were observed in the HLS + OA group under the joint disuse condition. The cartilage degeneration and subchondral bone sclerosis were alleviated in the US treatment group, especially under normal joint use condition. In conclusion, low-intensity ultrasound could improve cartilage degeneration and subchondral sclerosis during OA progression. Also, it could provide a promising strategy for future clinical treatment for OA patients.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Yueli Sun
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Zhilun Zhou
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Dongye Zhang
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Jian Jiao
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Minyi Hu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Chaudhry Raza Hassan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Yi-Xian Qin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA.
| |
Collapse
|
37
|
Kamatsuki Y, Aoyama E, Furumatsu T, Miyazawa S, Maehara A, Yamanaka N, Nishida T, Kubota S, Ozaki T, Takigawa M. Possible reparative effect of low-intensity pulsed ultrasound (LIPUS) on injured meniscus. J Cell Commun Signal 2018; 13:193-207. [PMID: 30460593 DOI: 10.1007/s12079-018-0496-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022] Open
Abstract
Menisci are a pair of crescent-shaped fibrocartilages, particularly of which their inner region of meniscus is an avascular tissue. It has characteristics similar to those of articular cartilage, and hence is inferior in healing. We previously reported that low-intensity pulsed ultrasound (LIPUS) treatment stimulates the production of CCN2/CTGF, a protein involved in repairing articular cartilage, and the gene expression of major cartilage matrices such as type II collagen and aggrecan in cultured chondrocytes. Therefore, in this present study, we investigated whether LIPUS has also favorable effect on meniscus cells and tissues. LIPUS applied with a 60 mW/cm2 intensity for 20 min stimulated the gene expression and protein production of CCN2 via ERK and p38 signaling pathways, as well as gene expression of SOX9, aggrecan, and collagen type II in human inner meniscus cells in culture, and slightly stimulated the gene expression of CCN2 and promoted the migration in human outer meniscus cells in culture. LIPUS also induced the expression of Ccn2, Sox9, Col2a1, and Vegf in rat intact meniscus. Furthermore, histological evaluations showed that LIPUS treatment for 1 to 4 weeks promoted healing of rat injured lateral meniscus, as evidenced by better and earlier angiogenesis and extracellular matrix synthesis. The data presented indicate that LIPUS treatment might prevent meniscus from degenerative change and exert a reparative effect on injured meniscus via up-regulation of repairing factors such as CCN2 and that it might thus be useful for treatment of an injured meniscus as a non-invasive therapy.
Collapse
Affiliation(s)
- Yusuke Kamatsuki
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kitaku, Okayama, 700-8558, Japan.,Advanced Research Center for Oral and Craniofacial Sciences (ARCOCS), Okayama University Dental School/Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Eriko Aoyama
- Advanced Research Center for Oral and Craniofacial Sciences (ARCOCS), Okayama University Dental School/Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Takayuki Furumatsu
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kitaku, Okayama, 700-8558, Japan
| | - Shinichi Miyazawa
- Department of Intelligent Orthopedic System, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama, Japan
| | - Ami Maehara
- Department of Intelligent Orthopedic System, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama, Japan
| | | | - Takashi Nishida
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Satoshi Kubota
- Advanced Research Center for Oral and Craniofacial Sciences (ARCOCS), Okayama University Dental School/Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan.,Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kitaku, Okayama, 700-8558, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences (ARCOCS), Okayama University Dental School/Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan.
| |
Collapse
|
38
|
Kaur H, Siraki AG, Sharma M, Uludağ H, Dederich DN, Flood P, El-Bialy T. Reactive Oxygen Species Mediate Therapeutic Ultrasound-Induced, Mitogen-Activated Protein Kinase Activation in C28/I2 Chondrocytes. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:2105-2114. [PMID: 30037475 DOI: 10.1016/j.ultrasmedbio.2018.05.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 04/03/2018] [Accepted: 05/29/2018] [Indexed: 06/08/2023]
Abstract
Low-intensity pulsed ultrasound (LIPUS) has been used for the treatment of non-healing fractures because of its therapeutic properties of stimulating enhancing endochondral bone formation. However, its mechanism of action remains unclear. In this study, we hypothesized that LIPUS activates mitogen-activated protein kinases through generation of reactive oxygen species. C28/I2 cells were stimulated with LIPUS for 10 and 20 min, while the control group was treated using a sham LIPUS transducer. Through quantitative reverse transcription polymerase chain reaction and immunoblot analyses, we determined that LIPUS application increased reactive oxygen species generation and cell viability in C28/I2 cells. There were increases in the phosphorylation level of ERK1/2 and in expression of SOX9, COL2 A1 and ACAN genes. These effects were reversed when cells were treated with diphenylene iodonium, which is known to inhibit NADPH oxidase. It was concluded that exposure of chondrocytes to LIPUS led to reactive oxygen species generation, which activated MAPK signaling and further increased chondrocyte-specific gene markers involved in chondrocyte differentiation and extracellular matrix formation.
Collapse
Affiliation(s)
- Harmanpreet Kaur
- Department of Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| | - Arno G Siraki
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Monika Sharma
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - Hasan Uludağ
- Department of Chemical and Material Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Douglas N Dederich
- Department of Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Patrick Flood
- Department of Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Tarek El-Bialy
- Department of Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
39
|
Shen J, Xie Y, Liu Z, Zhang S, Wang Y, Jia L, Wang Y, Cai Z, Ma H, Xiang M. Increased myocardial stiffness activates cardiac microvascular endothelial cell via VEGF paracrine signaling in cardiac hypertrophy. J Mol Cell Cardiol 2018; 122:140-151. [PMID: 30138627 DOI: 10.1016/j.yjmcc.2018.08.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 11/21/2022]
Abstract
When the heart is subjected to an increased workload, mechanical stretch together with neurohumoral stimuli activate the "fetal gene program" and induce cardiac hypertrophy to optimize output. Due to a lack of effective methods/models to quantify and modulate cardiac mechanical properties, the connection between these properties and the development of cardiac hypertrophy remains largely unexplored. Here, we utilized an atomic force microscope (AFM) to directly measure the elastic modulus of the hypertrophic myocardium induced by pressure overload. Additionally, we investigated the effects of extracellular elasticity on angiogenesis, which provides blood and nutrition to support cardiomyocyte hypertrophic growth in this process. In response to pressure overload, the myocardium rapidly developed hypertrophy and correspondingly demonstrated a high elastic modulus property. This mechanical feature correlated with enhanced angiogenesis. Mechanistically, we found that a high elastic modulus promoted cultured cardiomyocytes to synthesize and paracrine vascular endothelial growth factor (VEGF) to activate cardiac microvascular endothelial cells. Further analysis showed that the increased elastic modulus enhanced the interaction between Talin1 and integrin β1 to activate the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/hypoxia-inducible factor 1α (Hif-1α) pathway, which contributed to VEGF production. Thus, our study revealed a critical role of the elastic modulus in regulating angiogenesis during the development of cardiac hypertrophy.
Collapse
Affiliation(s)
- Jian Shen
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Yao Xie
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Zhenjie Liu
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Shuning Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200000, China
| | - Yaping Wang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Liangliang Jia
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Yidong Wang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Zhejun Cai
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Hong Ma
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China.
| | - Meixiang Xiang
- Department of Cardiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China.
| |
Collapse
|
40
|
Yanoshita M, Hirose N, Okamoto Y, Sumi C, Takano M, Nishiyama S, Asakawa-Tanne Y, Horie K, Onishi A, Yamauchi Y, Mitsuyoshi T, Kunimatsu R, Tanimoto K. Cyclic Tensile Strain Upregulates Pro-Inflammatory Cytokine Expression Via FAK-MAPK Signaling in Chondrocytes. Inflammation 2018; 41:1621-1630. [DOI: 10.1007/s10753-018-0805-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
41
|
Xia P, Wang X, Qu Y, Lin Q, Cheng K, Gao M, Ren S, Zhang T, Li X. TGF-β1-induced chondrogenesis of bone marrow mesenchymal stem cells is promoted by low-intensity pulsed ultrasound through the integrin-mTOR signaling pathway. Stem Cell Res Ther 2017; 8:281. [PMID: 29237506 PMCID: PMC5729425 DOI: 10.1186/s13287-017-0733-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 11/19/2017] [Accepted: 11/23/2017] [Indexed: 02/06/2023] Open
Abstract
Background Low-intensity pulsed ultrasound (LIPUS) is a mechanical stimulus that plays a key role in regulating the differentiation of bone marrow mesenchymal stem cells (BMSCs). However, the way in which it affects the chondrogenic differentiation of BMSCs remains unknown. In this study, we aimed to investigate whether LIPUS is able to influence TGF-β1-induced chondrogenesis of BMSCs through the integrin-mechanistic target of the Rapamycin (mTOR) signaling pathway. Methods BMSCs were isolated from rat bone marrow and cultured in either standard or TGF-β1-treated culture medium. BMSCs were then subjected to LIPUS at a frequency of 3 MHz and a duty cycle of 20%, and integrin and mTOR inhibitors added in order to analyze their influence on cell differentiation. BMSCs were phenotypically analyzed by flow cytometry and the degree of chondrogenesis evaluated through toluidine blue staining, immunofluorescence, and immunocytochemistry. Furthermore, expression of COL2, aggrecan, SOX9, and COL1 was assessed by qRT-PCR and western blot analysis. Results We found that LIPUS promoted TGF-β1-induced chondrogenesis of BMSCs, represented by increased expression of COL2, aggrecan and SOX9 genes, and decreased expression of COL1. Notably, these effects were prevented following addition of integrin and mTOR inhibitors. Conclusions Taken together, these results indicate that mechanical stimulation combined with LIPUS promotes TGF-β1-induced chondrogenesis of BMSCs through the integrin-mTOR signaling pathway.
Collapse
Affiliation(s)
- Peng Xia
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xiaoju Wang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yanping Qu
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Qiang Lin
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Kai Cheng
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Mingxia Gao
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Shasha Ren
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Tingting Zhang
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xueping Li
- Department of Rehabilitation Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
42
|
Ali ER, Mohamad AM. Evaluation of the efficiency of anthropometric parameters and submental ultrasonographic indices as predictors for screening of obstructive sleep apnea and its severity. THE EGYPTIAN JOURNAL OF BRONCHOLOGY 2017. [DOI: 10.4103/ejb.ejb_17_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
43
|
Yang Q, Nanayakkara GK, Drummer C, Sun Y, Johnson C, Cueto R, Fu H, Shao Y, Wang L, Yang WY, Tang P, Liu LW, Ge S, Zhou XD, Khan M, Wang H, Yang X. Low-Intensity Ultrasound-Induced Anti-inflammatory Effects Are Mediated by Several New Mechanisms Including Gene Induction, Immunosuppressor Cell Promotion, and Enhancement of Exosome Biogenesis and Docking. Front Physiol 2017; 8:818. [PMID: 29109687 PMCID: PMC5660123 DOI: 10.3389/fphys.2017.00818] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 10/05/2017] [Indexed: 12/18/2022] Open
Abstract
Background: Low-intensity ultrasound (LIUS) was shown to be beneficial in mitigating inflammation and facilitating tissue repair in various pathologies. Determination of the molecular mechanisms underlying the anti-inflammatory effects of LIUS allows to optimize this technique as a therapy for the treatment of malignancies and aseptic inflammatory disorders. Methods: We conducted cutting-edge database mining approaches to determine the anti-inflammatory mechanisms exerted by LIUS. Results: Our data revealed following interesting findings: (1) LIUS anti-inflammatory effects are mediated by upregulating anti-inflammatory gene expression; (2) LIUS induces the upregulation of the markers and master regulators of immunosuppressor cells including MDSCs (myeloid-derived suppressor cells), MSCs (mesenchymal stem cells), B1-B cells and Treg (regulatory T cells); (3) LIUS not only can be used as a therapeutic approach to deliver drugs packed in various structures such as nanobeads, nanospheres, polymer microspheres, and lipidosomes, but also can make use of natural membrane vesicles as small as exosomes derived from immunosuppressor cells as a novel mechanism to fulfill its anti-inflammatory effects; (4) LIUS upregulates the expression of extracellular vesicle/exosome biogenesis mediators and docking mediators; (5) Exosome-carried anti-inflammatory cytokines and anti-inflammatory microRNAs inhibit inflammation of target cells via multiple shared and specific pathways, suggesting exosome-mediated anti-inflammatory effect of LIUS feasible; and (6) LIUS-mediated physical effects on tissues may activate specific cellular sensors that activate downstream transcription factors and signaling pathways. Conclusions: Our results have provided novel insights into the mechanisms underlying anti-inflammatory effects of LIUS, and have provided guidance for the development of future novel therapeutic LIUS for cancers, inflammatory disorders, tissue regeneration and tissue repair.
Collapse
Affiliation(s)
- Qian Yang
- Department of Ultrasound, Xijing Hospital and Fourth Military Medical University, Xi'an, China.,Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Gayani K Nanayakkara
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Charles Drummer
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yu Sun
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Candice Johnson
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ramon Cueto
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hangfei Fu
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ying Shao
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Luqiao Wang
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Cardiovascular Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - William Y Yang
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Peng Tang
- Department of Orthopedics, Beijing Charity Hospital of China Rehabilitation Research Center, Beijing, China
| | - Li-Wen Liu
- Department of Ultrasound, Xijing Hospital and Fourth Military Medical University, Xi'an, China
| | - Shuping Ge
- Heart Center, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, PA, United States.,Deborah Heart and Lung Center, Browns Mills, NJ, United States
| | - Xiao-Dong Zhou
- Department of Ultrasound, Xijing Hospital and Fourth Military Medical University, Xi'an, China
| | - Mohsin Khan
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Departments of Pharmacology, Microbiology and Immunology, Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
44
|
Zhao L, Feng Y, Shi A, Zhang L, Guo S, Wan M. Neuroprotective Effect of Low-Intensity Pulsed Ultrasound Against MPP +-Induced Neurotoxicity in PC12 Cells: Involvement of K2P Channels and Stretch-Activated Ion Channels. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:1986-1999. [PMID: 28583325 DOI: 10.1016/j.ultrasmedbio.2017.04.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/18/2017] [Accepted: 04/20/2017] [Indexed: 06/07/2023]
Abstract
Parkinson's disease is the second most common neurodegenerative disease. It is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta. 1-Methyl-4-phenylpyridinium (MPP+) is a dopaminergic neuronal toxin that is widely used in constructing Parkinson's disease models in vitro. Low-intensity pulsed ultrasound (LIPUS) is a non-invasive therapeutic approach that has neuromodulation and neuroprotective effects in the central neural system; however, whether LIPUS can provide protection for dopaminergic neurons against MPP+-induced neurocytotoxicity remains unknown. In this study, we found that pre-treatment with LIPUS (1 MHz, 50 mW/cm2, 20% duty cycle and 100-Hz pulse repetition frequency, 10 min) inhibited MPP+-induced neurotoxicity and mitochondrial dysfunction in PC12 cells. LIPUS decreased MPP+-induced oxidative stress by modulating antioxidant proteins, including thioredoxin-1 and heme oxygenase-1, and prevented neurocytotoxicity via the phosphoinositide 3-kinase (PI3K)-Akt and ERK1/2 pathways. Furthermore, these beneficial effects were attributed to the activation of K2P channels and stretch-activated ion channels by LIPUS. These data indicate that LIPUS protects neuronal cells from MPP+-induced cell death through the K2P channel- and stretch-activated ion channel-mediated downstream pathways. The data also suggest that LIPUS could be a promising therapeutic method in halting or retarding the degeneration of dopaminergic neurons in Parkinson's disease in a non-invasive manner.
Collapse
Affiliation(s)
- Lu Zhao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Yi Feng
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.
| | - Aiwei Shi
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Lei Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Shifang Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Mingxi Wan
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
45
|
The Role of Low-Intensity Pulsed Ultrasound on Cartilage Healing in Knee Osteoarthritis: A Review. PM R 2017; 9:1268-1277. [PMID: 28606838 DOI: 10.1016/j.pmrj.2017.05.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 04/05/2017] [Accepted: 05/25/2017] [Indexed: 10/19/2022]
Abstract
Ultrasound (US) is a therapeutic modality that has been used in the treatment of musculoskeletal conditions for decades. In recent years, there have been technological advancements using low-intensity pulsed ultrasound (LIPUS) as a clinical modality. The purpose of this review was to critically examine the medical literature to determine the effects of LIPUS on the chondrogenic properties of knee osteoarthritis. A literature search of 3 major databases (PubMed, Scopus, and EMBASE) was performed. Two independent physician reviewers screened titles and abstracts, yielding a total of 18 relevant articles after the inclusion and exclusion criteria were applied. Results favored that LIPUS has a promising effect on the cellular elements in articular cartilage, specifically on chondrocytes in knee osteoarthritis. Although the use of LIPUS is encouraging based on basic science and preclinical data, there is a paucity of evidence with respect to humans. Consequently, there is insufficient evidence to recommend for or against LIPUS in clinical OA populations. We suggest future directions for research centered on LIPUS in both human and animal models to delineate the effect on the biologic properties of cartilage in knee osteoarthritis. LEVEL OF EVIDENCE III.
Collapse
|
46
|
Atherton P, Lausecker F, Harrison A, Ballestrem C. Low-intensity pulsed ultrasound promotes cell motility through vinculin-controlled Rac1 GTPase activity. J Cell Sci 2017; 130:2277-2291. [PMID: 28576970 DOI: 10.1242/jcs.192781] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 05/29/2017] [Indexed: 12/16/2022] Open
Abstract
Low-intensity pulsed ultrasound (LIPUS) is a therapy used clinically to promote healing. Using live-cell imaging we show that LIPUS stimulation, acting through integrin-mediated cell-matrix adhesions, rapidly induces Rac1 activation associated with dramatic actin cytoskeleton rearrangements. Our study demonstrates that the mechanosensitive focal adhesion (FA) protein vinculin, and both focal adhesion kinase (FAK, also known as PTK2) and Rab5 (both the Rab5a and Rab5b isoforms) have key roles in regulating these effects. Inhibiting the link of vinculin to the actin-cytoskeleton abolished LIPUS sensing. We show that this vinculin-mediated link was not only critical for Rac1 induction and actin rearrangements, but was also important for the induction of a Rab5-dependent increase in the number of early endosomes. Expression of dominant-negative Rab5, or inhibition of endocytosis with dynasore, also blocked LIPUS-induced Rac1 signalling events. Taken together, our data show that LIPUS is sensed by cell matrix adhesions through vinculin, which in turn modulates a Rab5-Rac1 pathway to control ultrasound-mediated endocytosis and cell motility. Finally, we demonstrate that a similar FAK-Rab5-Rac1 pathway acts to control cell spreading upon fibronectin.
Collapse
Affiliation(s)
- Paul Atherton
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, USA
| | - Franziska Lausecker
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, USA
| | - Andrew Harrison
- Bioventus Cooperatief, Taurusavenue 31, 2132 LS Hoofddorp, The Netherlands
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, USA
| |
Collapse
|
47
|
Cao Z, Dou C, Li J, Tang X, Xiang J, Zhao C, Zhu L, Bai Y, Xiang Q, Dong S. Cordycepin inhibits chondrocyte hypertrophy of mesenchymal stem cells through PI3K/Bapx1 and Notch signaling pathway. BMB Rep 2017; 49:548-553. [PMID: 27439604 PMCID: PMC5227296 DOI: 10.5483/bmbrep.2016.49.10.071] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Indexed: 12/25/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are widely used in cartilage tissue engineering to repair articular cartilage defects. However, hypertrophy of chondrocytes derived from MSCs might hinder the stabilization of hyaline cartilage. Thus, it is very important to find a suitable way to maintain the chondrogenic phenotype of chondrocytes. It has been reported that cordycepin has anti-inflammatory and anti-tumor functions. However, the role of cordycepin in chondrocyte hypertrophy remains unclear. Therefore, the objective of this study was to determine the effect of cordycepin on chondrogenesis and chondrocyte hypertrophy in MSCs and ATDC5 cells. Cordycepin upregulated chondrogenic markers including Sox9 and collagen type II while down-regulated hypertrophic markers including Runx2 and collagen type X. Further exploration showed that cordycepin promoted chondrogenesis through inhibiting Nrf2 while activating BMP signaling. Besides, cordycepin suppressed chondrocyte hypertrophy through PI3K/Bapx1 pathway and Notch signaling. Our results indicated cordycepin had the potential to maintain chondrocyte phenotype and reconstruct engineered cartilage. [BMB Reports 2016; 49(10): 548-553]
Collapse
Affiliation(s)
- Zhen Cao
- Department of Anatomy, Third Military Medical University and National & Regional United Engineering Laboratory of Tissue Engineering, Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, China
| | - Ce Dou
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, China
| | - Jianmei Li
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, China
| | - Xiangyu Tang
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, China
| | - Junyu Xiang
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, China
| | - Chunrong Zhao
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, China
| | - Lingyu Zhu
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, China
| | - Yun Bai
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, China
| | - Qiang Xiang
- Department of Emergency, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Shiwu Dong
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
48
|
Chondro-protective effects of low intensity pulsed ultrasound. Osteoarthritis Cartilage 2016; 24:1989-1998. [PMID: 27364595 PMCID: PMC5071131 DOI: 10.1016/j.joca.2016.06.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/26/2016] [Accepted: 06/13/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Cartilage is a highly mechano-responsive tissue. Chondrocytes undergo a series of complex changes, including proliferation and metabolic alteration as the target of external biomechanical and biochemical stimuli. IL-1β is known to regulate chondrocyte metabolism and plays an important role in the pathogenesis of osteoarthritis (OA). The objective of this study was to employ low-intensity pulsed ultrasound (LIPUS) as a localized mechanical stimulus and assess its effects on chondrocyte migration, proliferation, metabolism, and differentiation, as well as its ability to suppress IL-1β mediated catabolism in cartilage. METHODS Human cartilage explants and chondrocytes were stimulated by LIPUS in the presence and absence of IL-1β to asses cartilage degradation, chondrocytes metabolism, migration, and proliferation. Western blot analyses were conducted to study IL-1β the associated NFκB pathway in chondrocytes. RESULTS LIPUS stimulation increased the proteoglycan content in human cartilage explants and inhibited IL-1β induced loss of proteoglycans. LIPUS stimulation increased rates of chondrocyte migration and proliferation, and promoted chondrogenesis in mesenchymal stem cells (MSC). Further, LIPUS suppressed IL-1β induced activation of phosphorylation of NFκB-p65 and IĸBα leading to reduced expression of MMP13 and ADAMT5 in chondrocytes. CONCLUSIONS Collectively, these data demonstrate the potential therapeutic effects of LIPUS in preventing cartilage degradation and treating OA via a mechanical stimulation that inhibits the catabolic action of IL-1β and stimulates chondrocyte migration, proliferation, and differentiation.
Collapse
|
49
|
Cho SE, Kim YM, Jeong JS, Seo YK. The effect of ultrasound for increasing neural differentiation in hBM-MSCs and inducing neurogenesis in ischemic stroke model. Life Sci 2016; 165:35-42. [PMID: 27590610 DOI: 10.1016/j.lfs.2016.08.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/21/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022]
Abstract
AIMS This study's purpose was to evaluate the effect of ultrasound in air at a frequency of 0.04MHz and an intensity of 50mW/cm2 on neural differentiation of hBM-MSCs in vitro and on neurogenesis in an ischemic stroke model. MATERIALS AND METHODS hBM-MSCs were exposed to 0.04MHz ultrasound and then compared with no exposed one in cell morphology, lactate dehydrogenase (LDH) activity, RT-PCR, and Western blot. In addition, we made stroke model mice by means of the photothrombosis (PT) method and these models were exposed to 0.04MHz ultrasound after hBM-MSCs injection. We compared with sham group in histological and immunohistochemical analysis and western blot. KEY FINDINGS Ultrasound induced neural differentiation without cell death. In stroke models, inflammatory cells were observed around the infarct area in the Cell, Cell/Ultrasound group and the brain infarct volume in the Cell/Ultrasound group was smaller than in the sham group. Further, the expression of neural proteins in the Cell/Ultrasound group was increased relative to the sham group. SIGNIFICANCE The present study showed that ultrasound promotes neural differentiation of hBM-MSC and neurogenesis in a mouse stroke model. This may be applicable as a therapeutic device with the aim of inducing neurogenesis following stroke.
Collapse
Affiliation(s)
- Sang-Eun Cho
- Department of Medical Biotechnology (BK21 plus team), Dongguk University, Seoul, Republic of Korea
| | - Yu-Mi Kim
- Department of Medical Biotechnology (BK21 plus team), Dongguk University, Seoul, Republic of Korea
| | - Jong-Seob Jeong
- Department of Medical Biotechnology (BK21 plus team), Dongguk University, Seoul, Republic of Korea
| | - Young-Kwon Seo
- Department of Medical Biotechnology (BK21 plus team), Dongguk University, Seoul, Republic of Korea.
| |
Collapse
|
50
|
Effects and Mechanisms of Low-Intensity Pulsed Ultrasound for Chronic Prostatitis and Chronic Pelvic Pain Syndrome. Int J Mol Sci 2016; 17:ijms17071057. [PMID: 27376284 PMCID: PMC4964433 DOI: 10.3390/ijms17071057] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/21/2016] [Accepted: 06/27/2016] [Indexed: 01/15/2023] Open
Abstract
Chronic Prostatitis/Chronic Pelvic Pain Syndrome (CP/CPPS) is one of the most common urologic diseases, and no curative treatments have been identified. Low-intensity pulsed ultrasound (LIPUS) has been successfully used in promoting tissue healing, inhibiting inflammation and pain, differentiating stem cells, and stimulating nerve regeneration/muscle regeneration, as well as enhancing angiogenesis. Very recently, LIPUS has been proven an effective approach for CP/CPPS. This review summarizes the possible mechanisms responsible for the therapeutic effect of LIPUS for CP/CPPS. To search publications relevant to the topics of this review, the search engine for life sciences of Entrez was used. We reviewed the available evidence from 1954 through 2015 concerning LIPUS for CP/CPPS. According to the literature, both transrectal and transperineal approaches of LIPUS are effective for CP/CPPS.
Collapse
|