1
|
Tang J, Luo Y, Wang Q, Wu J, Wei Y. Stimuli-Responsive Delivery Systems for Intervertebral Disc Degeneration. Int J Nanomedicine 2024; 19:4735-4757. [PMID: 38813390 PMCID: PMC11135562 DOI: 10.2147/ijn.s463939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024] Open
Abstract
As a major cause of low back pain, intervertebral disc degeneration is an increasingly prevalent chronic disease worldwide that leads to huge annual financial losses. The intervertebral disc consists of the inner nucleus pulposus, outer annulus fibrosus, and sandwiched cartilage endplates. All these factors collectively participate in maintaining the structure and physiological functions of the disc. During the unavoidable degeneration stage, the degenerated discs are surrounded by a harsh microenvironment characterized by acidic, oxidative, inflammatory, and chaotic cytokine expression. Loss of stem cell markers, imbalance of the extracellular matrix, increase in inflammation, sensory hyperinnervation, and vascularization have been considered as the reasons for the progression of intervertebral disc degeneration. The current treatment approaches include conservative therapy and surgery, both of which have drawbacks. Novel stimuli-responsive delivery systems are more promising future therapeutic options than traditional treatments. By combining bioactive agents with specially designed hydrogels, scaffolds, microspheres, and nanoparticles, novel stimuli-responsive delivery systems can realize the targeted and sustained release of drugs, which can both reduce systematic adverse effects and maximize therapeutic efficacy. Trigger factors are categorized into internal (pH, reactive oxygen species, enzymes, etc.) and external stimuli (photo, ultrasound, magnetic, etc.) based on their intrinsic properties. This review systematically summarizes novel stimuli-responsive delivery systems for intervertebral disc degeneration, shedding new light on intervertebral disc therapy.
Collapse
Affiliation(s)
- Jianing Tang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- First Clinic School, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yuexin Luo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- First Clinic School, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Qirui Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- First Clinic School, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Juntao Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- First Clinic School, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yulong Wei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
2
|
Du X, Liang K, Ding S, Shi H. Signaling Mechanisms of Stem Cell Therapy for Intervertebral Disc Degeneration. Biomedicines 2023; 11:2467. [PMID: 37760908 PMCID: PMC10525468 DOI: 10.3390/biomedicines11092467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/27/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Low back pain is the leading cause of disability worldwide. Intervertebral disc degeneration (IDD) is the primary clinical risk factor for low back pain and the pathological cause of disc herniation, spinal stenosis, and spinal deformity. A possible approach to improve the clinical practice of IDD-related diseases is to incorporate biomarkers in diagnosis, therapeutic intervention, and prognosis prediction. IDD pathology is still unclear. Regarding molecular mechanisms, cellular signaling pathways constitute a complex network of signaling pathways that coordinate cell survival, proliferation, differentiation, and metabolism. Recently, stem cells have shown great potential in clinical applications for IDD. In this review, the roles of multiple signaling pathways and related stem cell treatment in IDD are summarized and described. This review seeks to investigate the mechanisms and potential therapeutic effects of stem cells in IDD and identify new therapeutic treatments for IDD-related disorders.
Collapse
Affiliation(s)
| | | | | | - Haifei Shi
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (X.D.); (K.L.); (S.D.)
| |
Collapse
|
3
|
Zhang GZ, Li L, Luo ZB, Zhang CY, Wang YG, Kang XW. Identification and experimental validation of key extracellular proteins as potential targets in intervertebral disc degeneration. Bone Joint Res 2023; 12:522-535. [PMID: 37661086 PMCID: PMC10475329 DOI: 10.1302/2046-3758.129.bjr-2022-0369.r2] [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] [Indexed: 09/05/2023] Open
Abstract
Aims This study aimed, through bioinformatics analysis and in vitro experiment validation, to identify the key extracellular proteins of intervertebral disc degeneration (IDD). Methods The gene expression profile of GSE23130 was downloaded from the Gene Expression Omnibus (GEO) database. Extracellular protein-differentially expressed genes (EP-DEGs) were screened by protein annotation databases, and we used Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) to analyze the functions and pathways of EP-DEGs. STRING and Cytoscape were used to construct protein-protein interaction (PPI) networks and identify hub EP-DEGs. NetworkAnalyst was used to analyze transcription factors (TFs) and microRNAs (miRNAs) that regulate hub EP-DEGs. A search of the Drug Signatures Database (DSigDB) for hub EP-DEGs revealed multiple drug molecules and drug-target interactions. Results A total of 56 EP-DEGs were identified in the differential expression analysis. EP-DEGs were enriched in the extracellular structure organization, ageing, collagen-activated signalling pathway, PI3K-Akt signalling pathway, and AGE-RAGE signalling pathway. PPI network analysis showed that the top ten hub EP-DEGs are closely related to IDD. Correlation analysis also demonstrated a significant correlation between the ten hub EP-DEGs (p<0.05), which were selected to construct TF-gene interaction and TF-miRNA coregulatory networks. In addition, ten candidate drugs were screened for the treatment of IDD. Conclusion The findings clarify the roles of extracellular proteins in IDD and highlight their potential as promising novel therapeutic targets.
Collapse
Affiliation(s)
- Guang-Zhi Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| | - Lei Li
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| | - Zhang-Bin Luo
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| | - Cang-Yu Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| | - Yong-Gang Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| | - Xue-Wen Kang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| |
Collapse
|
4
|
Zhang G, Li L, Yang Z, Zhang C, Kang X. TMT-Based Proteomics Analysis of Senescent Nucleus Pulposus from Patients with Intervertebral Disc Degeneration. Int J Mol Sci 2023; 24:13236. [PMID: 37686041 PMCID: PMC10488253 DOI: 10.3390/ijms241713236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Lower back pain, a leading cause of disability worldwide, is associated with intervertebral disc degeneration (IDD) in approximately 40% of cases. Although nucleus pulposus (NP) cell senescence is a major contributor to IDD, the underlying mechanisms remain unclear. We collected NP samples from IDD patients who had undergone spinal surgery. Healthy and senescent NP tissues (n = 3) were screened using the Pfirrmann grading system combined with immunohistochemistry, as well as hematoxylin and eosin, Safranin O, Alcian blue, and Masson staining. Differentially expressed proteins (DEPs) were identified using quantitative TMT-based proteomics technology. Bioinformatics analyses included gene ontology (GO) annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and protein-protein interaction (PPI) analyses. In addition, immunofluorescence was used to verify protein expression. In total, 301 DEPs were identified in senescent NP tissues, including 92 upregulated and 209 downregulated proteins. In GO, DEPs were primarily associated with NF-kappaB transcription factor, extracellular regions, cellular protein metabolic processes, and post-translational protein modification. The enriched KEGG pathways included TGF-β, Wnt, RAP1, interleukin-17, extracellular matrix-receptor adhesion, and PI3K/Akt signaling pathways. PPI analysis demonstrated interactions between multiple proteins. Finally, immunofluorescence verified the expressions of MMP3, LUM, TIMP1, and CDC42 in senescent NP cells. Our study provides valuable insights into the mechanisms underlying senescent NP tissues in IDD patients. DEPs provide a basis for further investigation of the effects of senescent factors on IDD.
Collapse
Affiliation(s)
- Guangzhi Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730000, China; (G.Z.); (L.L.); (Z.Y.); (C.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou 730030, China
| | - Lei Li
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730000, China; (G.Z.); (L.L.); (Z.Y.); (C.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou 730030, China
| | - Zhili Yang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730000, China; (G.Z.); (L.L.); (Z.Y.); (C.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou 730030, China
| | - Cangyu Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730000, China; (G.Z.); (L.L.); (Z.Y.); (C.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou 730030, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730000, China; (G.Z.); (L.L.); (Z.Y.); (C.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou 730030, China
| |
Collapse
|
5
|
Chen S, He T, Zhong Y, Chen M, Yao Q, Chen D, Shao Z, Xiao G. Roles of focal adhesion proteins in skeleton and diseases. Acta Pharm Sin B 2023; 13:998-1013. [PMID: 36970189 PMCID: PMC10031257 DOI: 10.1016/j.apsb.2022.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/04/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
The skeletal system, which contains bones, joints, tendons, ligaments and other elements, plays a wide variety of roles in body shaping, support and movement, protection of internal organs, production of blood cells and regulation of calcium and phosphate metabolism. The prevalence of skeletal diseases and disorders, such as osteoporosis and bone fracture, osteoarthritis, rheumatoid arthritis, and intervertebral disc degeneration, increases with age, causing pain and loss of mobility and creating a huge social and economic burden globally. Focal adhesions (FAs) are macromolecular assemblies that are composed of the extracellular matrix (ECM), integrins, intracellular cytoskeleton and other proteins, including kindlin, talin, vinculin, paxillin, pinch, Src, focal adhesion kinase (FAK) and integrin-linked protein kinase (ILK) and other proteins. FA acts as a mechanical linkage connecting the ECM and cytoskeleton and plays a key role in mediating cell-environment communications and modulates important processes, such as cell attachment, spreading, migration, differentiation and mechanotransduction, in different cells in skeletal system by impacting distinct outside-in and inside-out signaling pathways. This review aims to integrate the up-to-date knowledge of the roles of FA proteins in the health and disease of skeletal system and focuses on the specific molecular mechanisms and underlying therapeutic targets for skeletal diseases.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yiming Zhong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
6
|
Aimaijiang M, Liu Y, Zhang Z, Qin Q, Liu M, Abulikemu P, Liu L, Zhou Y. LIPUS as a potential strategy for periodontitis treatment: A review of the mechanisms. Front Bioeng Biotechnol 2023; 11:1018012. [PMID: 36911184 PMCID: PMC9992218 DOI: 10.3389/fbioe.2023.1018012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Periodontitis is a chronic inflammatory condition triggered by oral bacteria. A sustained inflammatory state in periodontitis could eventually destroy the alveolar bone. The key objective of periodontal therapy is to terminate the inflammatory process and reconstruct the periodontal tissues. The traditional Guided tissue regeneration (GTR) procedure has unstable results due to multiple factors such as the inflammatory environment, the immune response caused by the implant, and the operator's technique. Low-intensity pulsed ultrasound (LIPUS), as acoustic energy, transmits the mechanical signals to the target tissue to provide non-invasive physical stimulation. LIPUS has positive effects in promoting bone regeneration, soft-tissue regeneration, inflammation inhibition, and neuromodulation. LIPUS can maintain and regenerate alveolar bone during an inflammatory state by suppressing the expression of inflammatory factors. LIPUS also affects the cellular behavior of periodontal ligament cells (PDLCs), thereby protecting the regenerative potential of bone tissue in an inflammatory state. However, the underlying mechanisms of the LIPUS therapy are still yet to be summarized. The goal of this review is to outline the potential cellular and molecular mechanisms of periodontitis-related LIPUS therapy, as well as to explain how LIPUS manages to transmit mechanical stimulation into the signaling pathway to achieve inflammatory control and periodontal bone regeneration.
Collapse
Affiliation(s)
- Maierhaba Aimaijiang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yiping Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhiying Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Qiuyue Qin
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Manxuan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Palizi Abulikemu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Lijun Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
7
|
Zhou N, Shen B, Bai C, Ma L, Wang S, Wu D. Nutritional deficiency induces nucleus pulposus cell apoptosis via the ATF4-PKM2-AKT signal axis. BMC Musculoskelet Disord 2022; 23:946. [PMID: 36324122 PMCID: PMC9628105 DOI: 10.1186/s12891-022-05853-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022] Open
Abstract
Background The intervertebral disc is the largest avascular tissue in the human body. The nucleus pulposus (NP) consumes glucose and oxygen to generate energy to maintain cellular metabolism via nutrients that diffuse from the cartilage endplate. The microenvironment in the intervertebral disc becomes nutritionally deficient during degeneration, and nutritional deficiency has been shown to inhibit the viability and proliferation of NP cells. Methods To investigate the molecular mechanism by which nutritional deficiency reduces viability and decreases proliferation, we created an in vitro model by using decreasing serum concentration percentages. Results In this study, we found that nutritional deficiency reduced NP cell viability and increased cell apoptosis and that the upregulation of ATF4 expression and the downregulation of PKM2 expression were involved in this process. Moreover, we found that PKM2 inhibition can reduce the cell apoptosis induced by ATF4 silence under nutritional deficiency. Conclusion Our findings revealed that PKM2 inhibition reduces the cell apoptosis induced by ATF4 silence under nutritional deficiency by inhibiting AKT phosphate. Revealing the function and mechanism of NP cell development under nutritional deficiency will provide new insights into the etiology, diagnosis, and treatment of intervertebral disc and related diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-022-05853-1.
Collapse
Affiliation(s)
- Ningfeng Zhou
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bin Shen
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chong Bai
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Ma
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shanjin Wang
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Desheng Wu
- grid.24516.340000000123704535Department of Spinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
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: 2.0] [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
|
9
|
Zhou Y, Li H, Wang L. Mechanism of miR-34a in the metabolism of extracellular matrix in fibroblasts of stress urinary incontinence via Nampt-mediated autophagy. Cell Stress Chaperones 2022; 27:369-381. [PMID: 35666377 PMCID: PMC9346036 DOI: 10.1007/s12192-022-01278-w] [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: 02/16/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 01/03/2023] Open
Abstract
Stress urinary incontinence (SUI) is a troublesome hygienic problem that afflicts the female population and is associated with extracellular matrix (ECM). Herein, we investigated the effects of microRNA (miR)-34a on ECM metabolism in fibroblasts of SUI via mediating nicotinamide phosphoribosyl transferase (Nampt/NAmPRTase) and hope to find novel insights in the treatment of SUI. Firstly, the anterior vaginal wall tissues of SUI patients and the female vaginal wall fibroblasts (FVWFs) of non-SUI subjects were collected and identified. Then, FVWFs were treated with 10 ng/mL of interleukin 1 beta (IL-1β) to establish SUI cell models. Subsequently, miR-34a and Nampt expressions in both types of cells were detected via RT-qPCR. It was found that miR-34a was poorly expressed, while Nampt was highly expressed in SUI. Subsequently, IL-1β-treated FVWFs were transfected with miR-34a-mimic and pcDNA3.1-Nampt, respectively. Thereafter, RT-qPCR and Western blot detected that miR-34a overexpression increased COL1A, ACAN, and TIMP-1; decreased MMP-2 and MMP-9; and elevated LC3 II/I ratio, Beclin-1 expression, and the autophagosome number in IL-1β-treated FVWFs, while Nampt upregulation reversed the above outcomes. Then, dual-luciferase reporter gene assay detected that Nampt is a downstream target of miR-34a. Together, miR-34a overexpression promoted autophagy, inhibited ECM degradation in IL-1β-treated FVWFs, and ameliorated SUI via suppressing Nampt.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Gynecology, Zhengzhou Central Hospital of Zhengzhou University, No. 195 Middle Tongbai Road, Zhengzhou, 450000, Henan Province, China
| | - Hongjuan Li
- Department of Gynecology, Zhengzhou Central Hospital of Zhengzhou University, No. 195 Middle Tongbai Road, Zhengzhou, 450000, Henan Province, China.
| | - Lu Wang
- Department of Gynecology, Zhengzhou Central Hospital of Zhengzhou University, No. 195 Middle Tongbai Road, Zhengzhou, 450000, Henan Province, China.
| |
Collapse
|
10
|
Acheta J, Stephens SBZ, Belin S, Poitelon Y. Therapeutic Low-Intensity Ultrasound for Peripheral Nerve Regeneration – A Schwann Cell Perspective. Front Cell Neurosci 2022; 15:812588. [PMID: 35069118 PMCID: PMC8766802 DOI: 10.3389/fncel.2021.812588] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [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
|
11
|
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: 1.0] [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
|
12
|
Yi W, Chen Q, Liu C, Li K, Tao B, Tian G, Zhou L, Li X, Shen J, Liu B, Hu Z, Wang D, Bai D. LIPUS inhibits inflammation and catabolism through the NF-κB pathway in human degenerative nucleus pulposus cells. J Orthop Surg Res 2021; 16:619. [PMID: 34663388 PMCID: PMC8522043 DOI: 10.1186/s13018-021-02739-1] [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: 07/11/2021] [Accepted: 09/21/2021] [Indexed: 11/29/2022] Open
Abstract
Background Low-intensity pulsed ultrasound (LIPUS) is a safe and noninvasive rehabilitative physical therapy with anti-inflammatory effects. The current study investigated the effect of LIPUS on the inflammation of nucleus pulposus (NP) cells and its underlying mechanism. Methods Human NP cells were acquired from lumbar disc herniation tissue samples and cultured for experiments. Human NP cells were treated with LPS and then exposed to LIPUS (15 mW/cm2, 30 mW/cm2 and 60 mW/cm2) for 20 min daily for 3 days to determine the appropriate intensity to inhibit the expression of the inflammatory factors TNF-α and IL-1β. The gene and protein expression of aggrecan, collagen II, MMP-3 and MMP-9 was measured by real‐time PCR and western blotting, respectively. The activity of the nuclear factor‐kappa B (NF‐κB) pathway was examined by western blotting and immunofluorescence. After pretreatment with the NF-κB inhibitor PDTC, the expression of TNF-α, IL-1β, MMP-3 and MMP-9 was measured by real‐time PCR. Results LIPUS at intensities of 15 mW/cm2, 30 mW/cm2 and 60 mW/cm2 inhibited LPS-induced NP cell expression of the inflammatory factors TNF-α and IL-1β, especially at 30 mW/cm2. LIPUS significantly upregulated the gene and protein expression of aggrecan and collagen II and downregulated the gene and protein expression of MMP-3 and MMP-9 in LPS-induced NP cells. The NF‐κB signaling pathway was inhibited by LIPUS through inhibiting the protein expression of p-P65 and the translocation of P65 into the nucleus in LPS-induced NP cells. In addition, LIPUS had similar effects as the NF-κB inhibitor PDTC by inhibiting the NF-κB signaling pathway, inflammation and catabolism in LPS-induced human degenerative nucleus pulposus cells. Conclusion LIPUS inhibited inflammation and catabolism through the NF‐κB pathway in human degenerative nucleus pulposus cells.
Collapse
Affiliation(s)
- Weiwei Yi
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China
| | - Qing Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China
| | - Chuan Liu
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China
| | - Kaiting Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China
| | - Bailong Tao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China
| | - Guihua Tian
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China
| | - Lu Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China
| | - Xiaohong Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China
| | - Jieliang Shen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China
| | - Bo Liu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China
| | - Zhenming Hu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China
| | - Dawu Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China.
| | - Dingqun Bai
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, No. 1 Shiyou Street, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
13
|
Chu YC, Lim J, Tseng MC, Wang JL. The responses of nucleus pulposus cells to pressure and ultrasound stimulation. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2020; 148:EL314. [PMID: 33138512 DOI: 10.1121/10.0002138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/19/2020] [Indexed: 06/11/2023]
Abstract
A cellular stimulation device with a pressurized chamber is developed to investigate the effect of ultrasound and pressure fluctuation on nucleus pulposus (NP) cells. The pressurized chamber is designed to emulate the in vivo environment of intervertebral discs, which are under dynamic pressure, and to emulate impact during sports and exercise. Both hydrostatic pressure and ultrasound stimulation increase phosphorylation of ERK (pERK) in NP cells, and promote its translocation into nucleus. This increase in pERK levels might be activated through calcium signaling pathways as intracellular calcium in NP cells was strongly elevated by pressure changes.
Collapse
Affiliation(s)
- Ya-Cherng Chu
- Department of Biomedical Engineering, Nation Taiwan University, Taipei, , , ,
| | - Jormay Lim
- Department of Biomedical Engineering, Nation Taiwan University, Taipei, , , ,
| | - Mu-Cyun Tseng
- Department of Biomedical Engineering, Nation Taiwan University, Taipei, , , ,
| | - Jaw-Lin Wang
- Department of Biomedical Engineering, Nation Taiwan University, Taipei, , , ,
| |
Collapse
|
14
|
de Lucas B, Pérez LM, Bernal A, Gálvez BG. Ultrasound Therapy: Experiences and Perspectives for Regenerative Medicine. Genes (Basel) 2020; 11:genes11091086. [PMID: 32957737 PMCID: PMC7563547 DOI: 10.3390/genes11091086] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Ultrasound has emerged as a novel tool for clinical applications, particularly in the context of regenerative medicine. Due to its unique physico-mechanical properties, low-intensity ultrasound (LIUS) has been approved for accelerated fracture healing and for the treatment of established non-union, but its utility has extended beyond tissue engineering to other fields, including cell regeneration. Cells and tissues respond to acoustic ultrasound by switching on genetic repair circuits, triggering a cascade of molecular signals that promote cell proliferation, adhesion, migration, differentiation, and extracellular matrix production. LIUS also induces angiogenesis and tissue regeneration and has anti-inflammatory and anti-degenerative effects. Accordingly, the potential application of ultrasound for tissue repair/regeneration has been tested in several studies as a stand-alone treatment and, more recently, as an adjunct to cell-based therapies. For example, ultrasound has been proposed to improve stem cell homing to target tissues due to its ability to create a transitional and local gradient of cytokines and chemokines. In this review, we provide an overview of the many applications of ultrasound in clinical medicine, with a focus on its value as an adjunct to cell-based interventions. Finally, we discuss the various preclinical and clinical studies that have investigated the potential of ultrasound for regenerative medicine.
Collapse
Affiliation(s)
- Beatriz de Lucas
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (B.d.L.); (L.M.P.)
| | - Laura M. Pérez
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (B.d.L.); (L.M.P.)
| | - Aurora Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain;
| | - Beatriz G. Gálvez
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain; (B.d.L.); (L.M.P.)
- Correspondence:
| |
Collapse
|
15
|
Lim J, Chu YS, Chu YC, Lo CM, Wang JL. Low Intensity Ultrasound Induces Epithelial Cell Adhesion Responses. J Biomech Eng 2020; 142:091014. [PMID: 32280990 DOI: 10.1115/1.4046883] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Indexed: 07/25/2024]
Abstract
In this study, we investigated the cellular mechanosensitive responses to a low intensity ultrasound (LIUS) stimulation (ISATA = 1 mW/cm2, pressure = 10 kPa). The dose and temporal effects at cell-substrate adhesion (CSA) at the basal level and cell-cell adhesion (CCA) at the apical level are reported in detail. A model of mouse mammary gland epithelial cells (EpH4) and the phosphorylation of mechanosensitive 130 kDa Crk-associated substrate (p130CAS) as an indicator for cellular responses were used. The intensity of phospho-p130CAS was found to be dependent on LIUS stress level, and the p130CAS was phosphorylated after 1 min stimulation at CSA. The phospho-p130CAS was also found to increase significantly at CCA upon LIUS stimulation. We confirmed that the cellular responses to ultrasound are immediate and dose dependent. Ultrasound affects not only CSA but also CCA. An E-cadherin knockout (EpH4ECad-/-) model also confirmed that phosphorylation of p130CAS at CCA is related to E-cadherins.
Collapse
Affiliation(s)
- Jormay Lim
- Department of Biomedical Engineering, National Taiwan University, 605 Jen-Su Hall, 1 Section 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Yeh-Shiu Chu
- Brian Research Center, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei 112, Taiwan
| | - Ya-Cherng Chu
- Department of Biomedical Engineering, National Taiwan University, 605 Jen-Su Hall, 1 Section 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Chun-Min Lo
- Department of Biomedical Engineering, National Yang-Ming University, No. 155, Sec. 2, Linong Street, Taipei 112, Taiwan
| | - Jaw-Lin Wang
- Department of Biomedical Engineering, National Taiwan University, 602 Jen-Su Hall, 1 Section 4, Roosevelt Road, Taipei 10617, Taiwan
| |
Collapse
|
16
|
Horne DA, Jones PD, Adams MS, Lotz JC, Diederich CJ. LIPUS far-field exposimetry system for uniform stimulation of tissues in-vitro: development and validation with bovine intervertebral disc cells. Biomed Phys Eng Express 2020; 6:035033. [PMID: 33438678 DOI: 10.1088/2057-1976/ab8b26] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Therapeutic Low-intensity Pulsed Ultrasound (LIPUS) has been applied clinically for bone fracture healing and has been shown to stimulate extracellular matrix (ECM) metabolism in numerous soft tissues including intervertebral disc (IVD). In-vitro LIPUS testing systems have been developed and typically include polystyrene cell culture plates (CCP) placed directly on top of the ultrasound transducer in the acoustic near-field (NF). This configuration introduces several undesirable acoustic artifacts, making the establishment of dose-response relationships difficult, and is not relevant for targeting deep tissues such as the IVD, which may require far-field (FF) exposure from low frequency sources. The objective of this study was to design and validate an in-vitro LIPUS system for stimulating ECM synthesis in IVD-cells while mimicking attributes of a deep delivery system by delivering uniform, FF acoustic energy while minimizing reflections and standing waves within target wells, and unwanted temperature elevation within target samples. Acoustic field simulations and hydrophone measurements demonstrated that by directing LIPUS energy at 0.5, 1.0, or 1.5 MHz operating frequency, with an acoustic standoff in the FF (125-350 mm), at 6-well CCP targets including an alginate ring spacer, uniform intensity distributions can be delivered. A custom FF LIPUS system was fabricated and demonstrated reduced acoustic intensity field heterogeneity within CCP-wells by up to 93% compared to common NF configurations. When bovine IVD cells were exposed to LIPUS (1.5 MHz, 200 μs pulse, 1 kHz pulse frequency, and ISPTA = 120 mW cm-2) using the FF system, sample heating was minimal (+0.81 °C) and collagen content was increased by 2.6-fold compared to the control and was equivalent to BMP-7 growth factor treatment. The results of this study demonstrate that FF LIPUS exposure increases collagen content in IVD cells and suggest that LIPUS is a potential noninvasive therapeutic for stimulating repair of tissues deep within the body such as the IVD.
Collapse
Affiliation(s)
- Devante A Horne
- Department of Orthopaedic Surgery, University of California, San Francisco, United States of America. The UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, and University of California, San Francisco, United States of America. Thermal Therapy Research Group, Radiation Oncology Department, University of California, San Francisco, United States of America
| | | | | | | | | |
Collapse
|
17
|
17β-Estradiol Prevents Extracellular Matrix Degradation by Downregulating MMP3 Expression via PI3K/Akt/FOXO3 Pathway. Spine (Phila Pa 1976) 2020; 45:292-299. [PMID: 31809475 DOI: 10.1097/brs.0000000000003263] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN In vitro studies of the role of 17β-estradiol (E2) and its possible targets in intervertebral disc degeneration (IDD). OBJECTIVE To define the regulatory role of E2 in IDD and the potential mechanisms. SUMMARY OF BACKGROUND DATA IDD has intricate etiology that is influenced by multiple risk factors. However, the underlying molecular mechanisms of occurrence and progression of IDD are not well elucidated. The degradation of extracellular matrix (ECM) has been extensively observed in IDD. E2 was found to inhibit ECM degradation in human nuleus pulposus cells (HNPCs), but the molecular mechanism remained to be determined. METHODS Western blot and qPCR was performed to quantify the expression of target proteins in HNPCs. Luciferase reporter gene assay was applied to detect the effects of E2 and forkhead box O-3 (FOXO3) on matrix metalloproteinases (MMP)-3 promoter activity. Chromatin immunoprecipitation assay analyzed the binding of FOXO3 to MMP-3 and the effect of E2 on this process. RESULTS We identified the upregulation of collagen II and aggrecan by E2 independent of time and concentration. And E2 downregulated MMP-3 expression in human nucleus pulposus cells. The phosphorylation of FOXO3 led to the reduction of MMP-3 promoter activity. Furthermore, 17β-estradiol-induced the activation of PI3K/Akt pathway is required for FOXO3 phosphorylated. CONCLUSION E2 prevents the degradation of ECM by upregulating collagen II and aggrecan expression via reducing MMP-3 expression in HNPCs, and PI3K/Akt/FOXO3 pathway is dispensable for MMP-3 downregulated. Therefore, E2 protects against IDD by preventing ECM degradation. LEVEL OF EVIDENCE 3.
Collapse
|
18
|
Wu X, Li S, Wang K, Hua W, Li S, Song Y, Zhang Y, Yang S, Yang C. TNF-α Regulates ITGβ1 and SYND4 Expression in Nucleus Pulposus Cells: Activation of FAK/PI3K Signaling. Inflammation 2020; 42:1575-1584. [PMID: 31111299 DOI: 10.1007/s10753-019-01019-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Integrins can function synergistically with syndecan-4 (SYND4) and bind to the fibronectin (FN) matrix, resulting in the regulation of tissue regeneration. This study aimed to explore the effects of TNF-α on the formation of FN/ITGβ1/SYND4 complex and the relative mechanism in NP cells. The expression of FN-ITG-SYND4 at the cellular level under TNF-α stimulation was detected by immunofluorescent staining, western blotting, and RT-PCR. ITGβ1 is a crucial component of ITG FN-induced FAK signaling, which was detected using dual mode. And, the involved signaling down stream pathways were also detected. FN is a preferred adhesion substrate for NP cells and that integrin β1 (ITGβ1) and SYND4 work synergistically during ECM engagement in a focal adhesion kinase (FAK)-dependent fashion. The PI3k/Akt pathway is obviously down-regulated, resulting in decreased adherence capacity and increased anoikis. TNF-α induction could weaken FAK activity and downstream levels of phospho-PI3K and Akt, resulting in decreased adherence capacity and increased apoptosis. Thus, TNF-α is essential for the formation of FN/ITGβ1/SYND4 complex in NP cells and further elucidates the inflammatory mechanism of NP cells degeneration.
Collapse
Affiliation(s)
- Xinghuo Wu
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Suyun Li
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Kun Wang
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Wenbin Hua
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Shuai Li
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Yu Song
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Yukun Zhang
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Shuhua Yang
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China
| | - Cao Yang
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, Hubei, China.
| |
Collapse
|
19
|
Xie S, Jiang X, Wang R, Xie S, Hua Y, Zhou S, Yang Y, Zhang J. Low-intensity pulsed ultrasound promotes the proliferation of human bone mesenchymal stem cells by activating PI3K/AKt signaling pathways. J Cell Biochem 2019; 120:15823-15833. [PMID: 31090943 DOI: 10.1002/jcb.28853] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/17/2019] [Accepted: 02/28/2019] [Indexed: 12/27/2022]
Abstract
Low-intensity pulsed ultrasound (LIPUS) is a promising therapy that is widely used in clinical applications and fundamental research. Previous research has shown that LIPUS exposure has a positive effect on stem cell proliferation. However, the impact of LIPUS exposure on human bone marrow mesenchymal stem cells (hBMSCs) remains unknown. In our study, the effect and mechanism of LIPUS exposure on the proliferation of hBMSCs were investigated, and the optimal parameters of LIPUS were determined. hBMSCs were obtained and identified by flow cytometry, and the proliferation of hBMSCs was measured using the Cell Counting Kit-8 assay to determine cell cycle and cell count. Expression levels of the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKt) pathway proteins and cyclin D1 were determined by western blot analysis. Next, hBMSCs were successfully cultured and identified as multipotent mesenchymal stem cells. We found that LIPUS could promote the proliferation of hBMSCs when the exposure time was 5 or 10 minutes per day. Furthermore, 50 or 60 mW/cm2 LIPUS had a more significant effect on cell proliferation, but if cells were irradiated by LIPUS for 20 minutes once a day, an intensity of at least 50 mW/cm2 could markedly inhibit cell growth. Cell cycle analysis demonstrated that LIPUS treatment drives cells to enter S and G2/M phases from the G0/G1 phase. LIPUS exposure increased phosphorylation of PI3K/AKt and significantly upregulated expression of cyclin D1. However, these effects were inhibited when cells were treated with PI3K inhibitor (LY294002), which in turn reduced LIPUS-mediated proliferation of hBMSCs. These results suggest that LIPUS exposure may be involved in the proliferation of hBMSCs via activation of the PI3K/AKt signaling pathway and high expression of cyclin D1, and the intensity of 50 or 60 mW/cm2 and exposure time of 5 minutes were determined to be the optimal parameters for LIPUS exposure.
Collapse
Affiliation(s)
- Shucai Xie
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Xili Jiang
- Department of Radiology, The Second People's Hospital of Hunan Province/Brain Hospital of Hunan Province, Changsha, Hunan, People's Republic of China
| | - Rui Wang
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Shaowei Xie
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Yongyong Hua
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Shuai Zhou
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Yijun Yang
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| | - Jianquan Zhang
- Department of Hepatobiliary Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, People's Republic of China
| |
Collapse
|
20
|
Xiao Q, Zeng JH, Zhou H, Qiu QH, Ke B, Deng L, Hu ZM, Roh J, Dai M. Expression and effects of leukemia inhibitory factor on nucleus pulposus degeneration. Mol Med Rep 2019; 19:2377-2385. [PMID: 30664218 PMCID: PMC6392089 DOI: 10.3892/mmr.2019.9874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022] Open
Abstract
Leukemia inhibitory factor (LIF) is a multifunctional cytokine. The present study aimed to determine the expression and effects of LIF on nucleus pulposus generation. Degenerated nucleus pulposus samples were obtained from animal models and patients with lumbar intervertebral disc herniation. Degradation scores of intervertebral discs were evaluated via magnetic resonance imaging (MRI) and histology, and the protein expression levels of LIF were detected. Furthermore, cultured primary human degenerated nucleus pulposus cells (DNPCs) were stimulated with various concentrations of recombinant human LIF protein (rhLIF), and aggrecan and collagen type II α1 (COL2α1) protein expression levels were detected by western blotting. In addition, aggrecan expression was determined by toluidine blue staining. The effects of rhLIF on proliferation and apoptosis of DNPCs were evaluated by Cell Counting Kit-8 and flow cytometry, respectively. The results revealed that the degradation scores of intervertebral discs were significantly associated with modeling time, as determined by MRI and histology. In addition, the protein expression levels of LIF were initially increased in patients with lumbar disc herniation and in rabbit models, particularly in the 2-week modeling group; however, its expression decreased with the progression of disc degeneration. Notably, LIF expression in each modeling group was higher than that in the control and 0 week modeling group. The in vitro study revealed that the protein expression levels of aggrecan and COL2α1 were significantly increased in response to rhLIF, in a dose-dependent manner, and statistical differences were identified between the treatment groups and control group. The results of toluidine blue staining were consistent with this finding. Although rhLIF had no effect on proliferation, it inhibited apoptosis of DNPCs in a concentration-dependent manner. In conclusion, LIF was upregulated during the process of intervertebral disc degeneration, and may promote the expression of extracellular matrix components. It may also be hypothesized that LIF acts as a potential protective factor by inhibiting apoptosis of DNPCs without affecting cell proliferation.
Collapse
Affiliation(s)
- Qiang Xiao
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ji-Huan Zeng
- Department of Orthopaedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Hao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Quan-He Qiu
- Department of Orthopaedics, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330029, P.R. China
| | - Bo Ke
- Department of Hematopathology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Liang Deng
- Department of Orthopaedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Zhen-Ming Hu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jeffrey Roh
- Swedish Neuroscience Institute, Swedish Medical Center, Seattle, WA 98122, USA
| | - Min Dai
- Department of Orthopaedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
21
|
Zhou H, Shen J, Hu Z, Zhong X. Leukemia inhibitory factor promotes extracellular matrix synthesis in degenerative nucleus pulposus cells via MAPK-ERK1/2 signaling pathway. Biochem Biophys Res Commun 2018; 507:253-259. [PMID: 30446227 DOI: 10.1016/j.bbrc.2018.11.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/04/2018] [Indexed: 12/15/2022]
Abstract
Extracellular matrix (ECM) anabolism and catabolism imbalance is key feature of chondrocyte and intervertebral disc nucleus pulposus (NP) cell degeneration. The role of LIF as a multifunctional cytokine in the ECM metabolism of chondrocytes is controversial, but no relevant research in the ECM metabolism of NP cells. This study aimed to explore the biofunction and related mechanisms of LIF in the degenerative NP cells. We obtained an increase in the expression of LIF in the human degenerated NP specimens. The addition of recombinant human leukemia inhibitory factor (rhLIF) to the degenerated NP cells cultured in vitro was found to stimulate the synthesis of ECM, and rhLIF could activate the ERK1/2 signaling pathway. However, coculture with PD98059, a signal inhibitor of ERK1/2, blocked the effect of rhLIF on the synthesis of ECM. To furtherly clarify the role of LIF, we carried out animal experiments and found that rhLIF treatment could successfully delay the degree of degeneration of the intervertebral disc in a rabbit model; but with the addition of PD98059, the function of rhLIF for degeneration protection disappeared. In summary, this study demonstrates that LIF plays a role in promoting ECM synthesis in the degenerated NP cells as a protective role in intervertebral disc degeneration (IDD), which is related to the activation of ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd., Chongqing, 400016, China
| | - Jieliang Shen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd., Chongqing, 400016, China
| | - Zhenming Hu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd., Chongqing, 400016, China
| | - Xiaoming Zhong
- Department of Orthopaedic Surgery, The Ninth People's Hospital of Chongqing, No.69, Jialing Village, Beibei District, Chongqing, 400799, China.
| |
Collapse
|
22
|
Shen J, Zhuo N, Xu S, Song Z, Hu Z, Hao J, Guo X. Resveratrol delivery by ultrasound-mediated nanobubbles targeting nucleus pulposus cells. Nanomedicine (Lond) 2018; 13:1433-1446. [PMID: 29658365 DOI: 10.2217/nnm-2018-0019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To improve nucleus pulposus cell-targeted therapy for intervertebral disc degeneration (IDD) by fabricating a novel kind of ultrasound (US)-mediated poly(lactic-co-glycolic acid) nanobubbles (NBs) as a means of targeted drug delivery. Materials & methods: The resveratrol (RES)-embedded NBs were synthesized using a double-emulsion method. The active NP cell-targeting biomarker CDH2 antibody (AbCDH2) was further conjugated to the NBs using a carbodiimide method. Then, this RES/AbCDH2 NBs were examined by physical properties, specifc cell-targeting ability, anticatabolism effect in vitro and in vivo. Results: RES/AbCDH2 NBs exhibited high RES-loading efficiency, and US triggered accelerated RES release. Furthermore, RES/AbCDH2 NB treatment exhibited excellent anticatabolic ability in vitro; and in an IDD rabbit model, US-mediated RES/AbCDH2 NB injection effectively retarded the degenerative process of the intervertebral disc in vivo. Conclusion: The combination of US irradiation and drug delivery through RES/AbCDH2 NBs can be considered as a novel treatment option for IDD.
Collapse
Affiliation(s)
- Jieliang Shen
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Naiqiang Zhuo
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Shenxi Xu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Zhaojun Song
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Zhenming Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Jie Hao
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Xiuming Guo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| |
Collapse
|
23
|
Zhang X, Hu B, Sun J, Li J, Liu S, Song J. Inhibitory Effect of Low-Intensity Pulsed Ultrasound on the Expression of Lipopolysaccharide-Induced Inflammatory Factors in U937 Cells. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2017; 36:2419-2429. [PMID: 28600899 DOI: 10.1002/jum.14239] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/01/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVES Low-intensity pulsed ultrasound (US) has been reported to promote periodontal tissue regeneration and reduce inflammation in soft tissues and in bone infectious diseases. Here we investigated the effect of low-intensity pulsed US on the expression of lipopolysaccharide (LPS)-induced inflammatory factors in U937 macrophage cells. METHODS U937 cells were stimulated with different concentrations of LPS and exposed to different intensities of low-intensity pulsed US. Cell viability and apoptosis of U937 cells were determined by cell-counting kit assays and flow cytometry. A real-time polymerase chain reaction and an enzyme-linked immunosorbent assay were used to test the expression of inflammatory factors. The expression levels of toll-like receptor 4, p65, p-IκBα, and IκBα were assessed by western blots. RESULTS Tumor necrosis factor α began to increase in U937 cells on induction with 1-μg/mL LPS. Low-intensity pulsed US at the intensity of 60 mW/cm2 was more effective in reducing interleukin 8 (IL-8) expression. Furthermore, LPS inhibited the viability and increased apoptosis of U937 cells, whereas low-intensity pulsed US significantly reversed these effects (P < .05). Low-intensity pulsed US reduced the protein expression of IL-6 and IL-8 at both gene and protein levels in U937 cells. The western blot and immunofluorescence showed that low-intensity pulsed US primarily suppressed the degradation and phosphorylation of IκBα and the translocation of p65 into the nuclei. CONCLUSIONS Low-intensity pulsed US alleviated the expression of inflammatory factors induced by LPS in U937 cells. This process was modulated by suppressing the toll-like receptor 4-nuclear factor κB signaling pathway. Therefore, low-intensity pulsed US might be a potential immunomodulatory therapy for the treatment of periodontitis.
Collapse
Affiliation(s)
- Xuan Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Bo Hu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jicheng Sun
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Shan Liu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
24
|
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: 62] [Impact Index Per Article: 8.9] [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
|
25
|
Horne D, Jones P, Salgaonkar V, Adams M, Ozilgen BA, Zahos P, Tang X, Liebenburg E, Coughlin D, Lotz J, Diederich C. Low intensity pulsed ultrasound (LIPUS) for the treatment of intervertebral disc degeneration. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2017; 10066. [PMID: 29104343 DOI: 10.1117/12.2255761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Discogenic back pain presents a major public health issue, with current therapeutic interventions limited to short-term symptom relief without providing regenerative remedies for diseased intervertebral discs (IVD). Many of these interventions are invasive and can diminish the biomechanical integrity of the IVDs. Low intensity pulsed ultrasound (LIPUS) is a potential treatment option that is both non-invasive and regenerative. LIPUS has been shown to be a clinically effective method for the enhancement of wound and fracture healing. Recent in vitro studies have shown that LIPUS stimulation induces an upregulation functional matrix proteins and downregulation of inflammatory factors in cultured IVD cells. However, we do not know the effects of LIPUS on an in vivo model for intervertebral disc degeneration. The objective of this study was to show technical feasibility of building a LIPUS system that can target the rat tail IVD and apply this setup to a model for acute IVD degeneration. A LIPUS exposimetry system was built using a 1.0 MHz planar transducer and custom housing. Ex vivo intensity measurements demonstrated LIPUS delivery to the center of the rat tail IVD. Using an established stab-incision model for disc degeneration, LIPUS was applied for 20 minutes daily for five days. For rats that displayed a significant injury response, LIPUS treatment caused significant upregulation of Collagen II and downregulation of Tumor Necrosis Factor - α gene expression. Our preliminary studies indicate technical feasibility of targeted delivery of ultrasound to a rat tail IVD for studies of LIPUS biological effects.
Collapse
Affiliation(s)
- Devante Horne
- UC Berkeley - UC San Francisco Graduate Program in Bioengineering, CA, USA.,Department of Radiation Oncology, UC San Francisco, CA USA
| | - Peter Jones
- Department of Radiation Oncology, UC San Francisco, CA USA
| | | | - Matt Adams
- Department of Radiation Oncology, UC San Francisco, CA USA.,Department of Orthopaedic Surgery, UC San Francisco, CA, USA
| | - B Arda Ozilgen
- UC Berkeley - UC San Francisco Graduate Program in Bioengineering, CA, USA.,Department of Radiation Oncology, UC San Francisco, CA USA
| | - Peter Zahos
- Department of Radiation Oncology, UC San Francisco, CA USA
| | - Xinyan Tang
- Department of Orthopaedic Surgery, UC San Francisco, CA, USA
| | | | - Dezba Coughlin
- Department of Orthopaedic Surgery, UC San Francisco, CA, USA
| | - Jeffrey Lotz
- Department of Radiation Oncology, UC San Francisco, CA USA.,Department of Orthopaedic Surgery, UC San Francisco, CA, USA
| | - Chris Diederich
- UC Berkeley - UC San Francisco Graduate Program in Bioengineering, CA, USA.,Department of Radiation Oncology, UC San Francisco, CA USA
| |
Collapse
|
26
|
Shen J, Xu S, Zhou H, Liu H, Jiang W, Hao J, Hu Z. IL-1β induces apoptosis and autophagy via mitochondria pathway in human degenerative nucleus pulposus cells. Sci Rep 2017; 7:41067. [PMID: 28120948 PMCID: PMC5264394 DOI: 10.1038/srep41067] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/14/2016] [Indexed: 12/13/2022] Open
Abstract
IL-1β has been reported highly expressed in degenerative intervertebral disc, and our previous study indicated IL-1β facilitates apoptosis of human degenerative nucleus pulposus (NP) cell. However, the underlying molecular mechanism remains unclear. We here demonstrate that IL-1β played a significantly pro-apoptotic effect under serum deprivation. IL-1β decreased Bcl-2/Bax ratio and enhanced cytochrome C released from mitochondria to cytosol, which proved mitochondria-meidated apoptosis was induced. Subsequently, mitochondria damage was detected under IL-1β stimualtion. In addition, IL-1β-mediated injuried mitochondria contributes to activate autophagy. However, pretreatment with the autophagy inhibitor 3-methyladenine showed the potential in further elevating the apoptosis rate induced by IL-1β in NP cells. Our results indicated that the mitochondrial pathway was involved in IL-1β-induced apoptosis of NP cells. Meanwhile, the damaged mitochondria-induced autophagy played a protective role against apoptosis, suggesting a postive feedback mechanism under inflammatory stress.
Collapse
Affiliation(s)
- Jieliang Shen
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengxi Xu
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Zhou
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huzhe Liu
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Jiang
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Hao
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhenming Hu
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
27
|
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: 35] [Impact Index Per Article: 4.4] [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
|