1
|
Wang X, Niu X, Wang Y, Liu Y, Yang C, Chen X, Qi Z. C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 pathway as a therapeutic target and regulatory mechanism for spinal cord injury. Neural Regen Res 2025; 20:2231-2244. [PMID: 39104168 DOI: 10.4103/nrr.nrr-d-24-00119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/07/2024] [Indexed: 08/07/2024] Open
Abstract
Spinal cord injury involves non-reversible damage to the central nervous system that is characterized by limited regenerative capacity and secondary inflammatory damage. The expression of the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis exhibits significant differences before and after injury. Recent studies have revealed that the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis is closely associated with secondary inflammatory responses and the recruitment of immune cells following spinal cord injury, suggesting that this axis is a novel target and regulatory control point for treatment. This review comprehensively examines the therapeutic strategies targeting the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis, along with the regenerative and repair mechanisms linking the axis to spinal cord injury. Additionally, we summarize the upstream and downstream inflammatory signaling pathways associated with spinal cord injury and the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis. This review primarily elaborates on therapeutic strategies that target the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis and the latest progress of research on antagonistic drugs, along with the approaches used to exploit new therapeutic targets within the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis and the development of targeted drugs. Nevertheless, there are presently no clinical studies relating to spinal cord injury that are focusing on the C-C motif chemokine ligand 2/C-C motif chemokine receptor 2 axis. This review aims to provide new ideas and therapeutic strategies for the future treatment of spinal cord injury.
Collapse
Affiliation(s)
- Xiangzi Wang
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xiaofei Niu
- Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingkai Wang
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yang Liu
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Cheng Yang
- Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Xuyi Chen
- Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Zhongquan Qi
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
- Fujian Maternity and Child Health Hospital, Fuzhou, Fujian Province, China
| |
Collapse
|
2
|
Liu Y, Guo F, Han Z, Yin Y, Chen G, Zhang Y, Tang Q, Chen L. Neutrophils inhibit bone formation by directly contacting osteoblasts and suppressing osteogenic differentiation. Bone 2024; 190:117310. [PMID: 39477179 DOI: 10.1016/j.bone.2024.117310] [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: 09/12/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/08/2024]
Abstract
Neutrophils have been extensively studied for their critical roles in supporting immune defense mechanisms, initiating bone regeneration, and promoting angiogenesis. Nonetheless, the influence of neutrophils on physiological conditions, particularly in the context of bone development, remains incompletely understood. In this study, we examined the effects of non-inflammatory neutrophils on bone physiology by depleting Ly6G+ neutrophils and inducing neutropenia through myelosuppression. Our results demonstrated a notable increase in bone mass and a decrease in the bone marrow cavity upon depletion of the neutrophils. These effects were attributed to the direct interaction between neutrophils and osteoblasts, independent of reduced secretion of typical inflammatory cytokines or diminished osteoclast differentiation. This observation suggests a non-inflammatory function of neutrophils within the endosteal microenvironment, where they regulate osteogenic differentiation to preserve optimal bone mass, shape healthy three-dimensional bone trabecular structures, and create ample space for hematopoietic niche development.
Collapse
Affiliation(s)
- Yijun Liu
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Fengyuan Guo
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Zhenshuo Han
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Ying Yin
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Guangjin Chen
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yifan Zhang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Qingming Tang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China.
| | - Lili Chen
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China.
| |
Collapse
|
3
|
Valdez CN, Sánchez-Zuno GA, Bucala R, Tran TT. Macrophage Migration Inhibitory Factor (MIF) and D-Dopachrome Tautomerase (DDT): Pathways to Tumorigenesis and Therapeutic Opportunities. Int J Mol Sci 2024; 25:4849. [PMID: 38732068 PMCID: PMC11084905 DOI: 10.3390/ijms25094849] [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: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Discovered as inflammatory cytokines, MIF and DDT exhibit widespread expression and have emerged as critical mediators in the response to infection, inflammation, and more recently, in cancer. In this comprehensive review, we provide details on their structures, binding partners, regulatory mechanisms, and roles in cancer. We also elaborate on their significant impact in driving tumorigenesis across various cancer types, supported by extensive in vitro, in vivo, bioinformatic, and clinical studies. To date, only a limited number of clinical trials have explored MIF as a therapeutic target in cancer patients, and DDT has not been evaluated. The ongoing pursuit of optimal strategies for targeting MIF and DDT highlights their potential as promising antitumor candidates. Dual inhibition of MIF and DDT may allow for the most effective suppression of canonical and non-canonical signaling pathways, warranting further investigations and clinical exploration.
Collapse
Affiliation(s)
- Caroline Naomi Valdez
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
| | - Gabriela Athziri Sánchez-Zuno
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
| | - Richard Bucala
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| | - Thuy T. Tran
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
- Section of Medical Oncology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| |
Collapse
|
4
|
Fang T, Liu L, Song D, Huang D. The role of MIF in periodontitis: A potential pathogenic driver, biomarker, and therapeutic target. Oral Dis 2024; 30:921-937. [PMID: 36883414 DOI: 10.1111/odi.14558] [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: 01/03/2023] [Revised: 02/08/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVE Periodontitis is an inflammatory disease that involves an imbalance in the oral microbiota, activation of inflammatory and immune responses, and alveolar bone destruction. Macrophage migration inhibitory factor (MIF) is a versatile cytokine involved in several pathological reactions, including inflammatory processes and bone destruction, both of which are characteristics of periodontitis. While the roles of MIF in cancer and other immune diseases have been extensively characterized, its role in periodontitis remains inconclusive. RESULTS In this review, we describe a comprehensive analysis of the potential roles of MIF in periodontitis from the perspective of immune response and bone regulation at the cellular and molecular levels. Moreover, we discuss its potential reliability as a novel diagnostic and therapeutic target for periodontitis. CONCLUSION This review can aid dental researchers and clinicians in understanding the current state of MIF-related pathogenesis, diagnosis, and treatment of periodontitis.
Collapse
Affiliation(s)
- Tongfeng Fang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liu Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dongzhe Song
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dingming Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Zhang Y, Zheng L, Fang J, Ni K, Hu X, Ye L, Lai H, Yang T, Chen Z, He D. Macrophage migration inhibitory factor (MIF) promotes intervertebral disc degeneration through the NF-κB pathway, and the MIF inhibitor CPSI-1306 alleviates intervertebral disc degeneration in a mouse model. FASEB J 2023; 37:e23303. [PMID: 37983963 DOI: 10.1096/fj.202301441r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 10/07/2023] [Accepted: 10/26/2023] [Indexed: 11/22/2023]
Abstract
Lumbar intervertebral disc degeneration(IDD) is a prevalent inflammatory disease caused by many proinflammatory factors, such as TNF and IL-1β. Migration inhibitory factor (MIF) is an upstream inflammatory factor widely expressed in vivo that is associated with a variety of inflammatory diseases or malignant tumors and has potential therapeutic value in many diseases. We explored the role of MIF in intervertebral disc degeneration by regulating the content of exogenous MIF or the expression of MIF in cells. Upon inducing degeneration of nucleus pulposus (NP) cells with IL-1β, we found that the increase in intracellular and exogenous MIF promoted the catabolism induced by proinflammatory factors in NP cells, while silencing of the MIF gene alleviated the degeneration to some extent. In a mouse model, the intervertebral disc degeneration of MIF-KO mice was significantly less than that of wild-type mice. To explore the treatment of intervertebral disc degeneration, we selected the small-molecular MIF inhibitor CPSI-1306. CPSI-1306 had a therapeutic effect on intervertebral disc degeneration in the mouse model. In summary, we believe that MIF plays an important role in intervertebral disc degeneration and is a potential therapeutic target for the treatment of intervertebral disc degeneration.
Collapse
Affiliation(s)
- Yejin Zhang
- Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Lin Zheng
- Department of Orthopaedics, The Second Affiliated Hospital of Wenzhou Medical College, Wenzhou, China
| | - Jiawei Fang
- Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Kainan Ni
- Department of Orthopaedics, The First People's Hospital of Fuyang, Hangzhou, China
| | - Xingyu Hu
- Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Lin Ye
- Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Hehuan Lai
- Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Tao Yang
- Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Zhenzhong Chen
- Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Dengwei He
- Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| |
Collapse
|
6
|
He J, Zheng L, Li X, Huang F, Hu S, Chen L, Jiang M, Lin X, Jiang H, Zeng Y, Ye T, Lin D, Liu Q, Xu J, Chen K. Obacunone targets macrophage migration inhibitory factor (MIF) to impede osteoclastogenesis and alleviate ovariectomy-induced bone loss. J Adv Res 2023; 53:235-248. [PMID: 36657717 PMCID: PMC10658311 DOI: 10.1016/j.jare.2023.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/21/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION Osteoporosis is the most common bone disorder where the hyperactive osteoclasts represent the leading role during the pathogenesis. Targeting hyperactive osteoclasts is currently the primary therapeutic strategy. However, concerns about the long-term efficacy and side effects of current frontline treatments persist. Alternative therapeutic agents are still needed. OBJECTIVES Obacunone (OB) is a small molecule with a broad spectrum of biological activities, particularly antioxidant and anti-inflammatory effects. This study aims to examine OB's therapeutic potential on osteoporosis and explore the rudimentary mechanisms. METHODS Osteoclast formation and osteoclastic resorption assays were carried out to examine OB's inhibitory effects in vitro, followed by the in-vivo studies of OB's therapeutic effects on ovariectomy-induced osteoporotic preclinical model. To further study the underlying mechanisms, mRNA sequencing and analysis were used to investigate the changes of downstream pathways. The molecular targets of OB were predicted, and in-silico docking analysis was performed. Ligand-target binding was verified by surface plasmon resonance (SPR) assay and Western Blotting assay. RESULTS The results indicated that OB suppressed the formation of osteoclast and its resorptive function in vitro. Mechanistically, OB interacts with macrophage migration inhibitory factor (MIF) which attenuates receptor activator of nuclear factor kappa B (NF-κB) ligand (RANKL)-induced signaling pathways, including reactive oxygen species (ROS), NF-κB pathway, and mitogen-activated protein kinases (MAPKs). These effects eventually caused the diminished expression level of the master transcriptional factor of osteoclastogenesis, nuclear factor of activated T cells 1 (NFATc1), and its downstream osteoclast-specific proteins. Furthermore, our data revealed that OB alleviated estrogen deficiency-induced osteoporosis by targeting MIF and thus inhibiting hyperactive osteoclasts in vivo. CONCLUSION These results together implicated that OB may represent as a therapeutic candidate for bone disorders caused by osteoclasts, such as osteoporosis.
Collapse
Affiliation(s)
- Jianbo He
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China; School of Biomedical Sciences, The University of Western Australia, Perth 6009, Australia
| | - Lin Zheng
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou 310000, China
| | - Xiaojuan Li
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Furong Huang
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Sitao Hu
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Lei Chen
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Manya Jiang
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou 510632, China
| | - Xianfeng Lin
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou 310000, China
| | - Haibo Jiang
- School of Molecular Sciences, The University of Western Australia, Perth 6009, Australia
| | - Yifan Zeng
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Tianshen Ye
- Department of Acupuncture, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Dingkun Lin
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, China
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth 6009, Australia.
| | - Kai Chen
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; School of Molecular Sciences, The University of Western Australia, Perth 6009, Australia.
| |
Collapse
|
7
|
Chai L, Wang Q, Wang Y, Li D, Zhang Q, Chen Y, Liu J, Chen H, Qiu Y, Shen N, Wang J, Xie X, Li M. Downregulation of PDCD4 through STAT3/ATF6/autophagy mediates MIF-induced PASMCs proliferation/migration and vascular remodeling. Eur J Pharmacol 2023; 956:175968. [PMID: 37549728 DOI: 10.1016/j.ejphar.2023.175968] [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: 02/12/2023] [Revised: 07/08/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
To address the molecular mechanisms underlying macrophage migration inhibitory factor (MIF) induced pulmonary artery smooth muscle cells (PASMCs) proliferation, migration and vascular remodeling in pulmonary hypertension (PH), primary cultured rat PASMCs and monocrotaline (MCT)-induced rats with PH were applied in the present study. The results showed that MIF increased signal transducer and activator of transcription 3 (STAT3) phosphorylation, and then stimulated activating transcription factor 6 (ATF6) activation, subsequently triggered autophagy activation, which further led to programmed cell death factor 4 (PDCD4) lysosomal degradation, and eventually promoted PASMCs proliferation/migration. In lung tissues of MCT rats, MIF protein expression was elevated, phosphorylation of STAT3 and activation of ATF6 were increased, activation of autophagy was evident, and reduction of PDCD4 was observed. Intervention with MIF inhibitor 4-Iodo-6-phenylpyrimidine (4-IPP), ATF6 blocker melatonin or autophagy inhibitor chloroquine, confirmed the in vitro interaction among MIF, STAT3, ATF6, autophagy and PDCD4 in MCT induced rats with PH. Targeting MIF/STAT3/ATF6/autophagy/PDCD4 axis effectively prevented the development of PH by suppressing PASMCs proliferation and vascular remodeling. In conclusions, we demonstrate that MIF activates the STAT3/ATF6/autophagy cascade and then degrades PDCD4 leading to PASMCs proliferation/migration and pulmonary vascular remodeling, suggesting that intervention this axis might have potential value in management of PH.
Collapse
Affiliation(s)
- Limin Chai
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Qingting Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yan Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Danyang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Qianqian Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yuqian Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Jin Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Huan Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yuanjie Qiu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Nirui Shen
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Jian Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Xinming Xie
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Manxiang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China.
| |
Collapse
|
8
|
Liu J, Chen Y, Chen H, Wang Y, Li D, Zhang Q, Chai L, Qiu Y, Zhang J, Shen N, Wang Q, Wang J, Li M. Macrophage migration inhibitory factor exacerbates asthmatic airway remodeling via dynamin-related protein 1-mediated autophagy activation. Respir Res 2023; 24:216. [PMID: 37674165 PMCID: PMC10481618 DOI: 10.1186/s12931-023-02526-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: 06/07/2023] [Accepted: 08/31/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) and GTPase dynamin-related protein 1 (Drp1)-dependent aberrant mitochondrial fission are closely linked to the pathogenesis of asthma. However, it is unclear whether Drp1-mediated mitochondrial fission and its downstream targets mediate MIF-induced proliferation of airway smooth muscle cells (ASMCs) in vitro and airway remodeling in chronic asthma models. The present study aims to clarify these issues. METHODS In this study, primary cultured ASMCs and ovalbumin (OVA)-induced asthmatic rats were applied. Cell proliferation was detected by CCK-8 and EdU assays. Western blotting was used to detect extracellular signal-regulated kinase (ERK) 1/2, Drp1, autophagy-related markers and E-cadherin protein phosphorylation and expression. Inflammatory cytokines production, airway reactivity test, histological staining and immunohistochemical staining were conducted to evaluate the development of asthma. Transmission electron microscopy was used to observe the mitochondrial ultrastructure. RESULTS In primary cultured ASMCs, MIF increased the phosphorylation level of Drp1 at the Ser616 site through activation of the ERK1/2 signaling pathway, which further activated autophagy and reduced E-cadherin expression, ultimately leading to ASMCs proliferation. In OVA-induced asthmatic rats, MIF inhibitor 4-iodo-6-phenylpyrimidine (4-IPP) treatment, suppression of mitochondrial fission by Mdivi-1 or inhibiting autophagy with chloroquine phosphate (CQ) all attenuated the development of airway remodeling. CONCLUSIONS The present study provides novel insights that MIF promotes airway remodeling in asthma by activating autophagy and degradation of E-cadherin via ERK/Drp1 signaling pathway, suggesting that targeting MIF/ERK/Drp1 might have potential therapeutic value for the prevention and treatment of asthma.
Collapse
Affiliation(s)
- Jin Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yuqian Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Huan Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Danyang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yuanjie Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Jia Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Nirui Shen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China.
| |
Collapse
|
9
|
Sun W, Ma J, Chen M, Zhang W, Xu C, Nan Y, Wu W, Mao X, Cheng X, Cai H, Zhang J, Xu H, Wang Y. 4-Iodo-6-phenylpyrimidine (4-IPP) suppresses fibroblast-like synoviocyte- mediated inflammation and joint destruction associated with rheumatoid arthritis. Int Immunopharmacol 2023; 115:109714. [PMID: 36657337 DOI: 10.1016/j.intimp.2023.109714] [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: 09/05/2022] [Revised: 12/20/2022] [Accepted: 01/06/2023] [Indexed: 01/19/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic immune-mediated inflammatory disease that significantly impacts patients' quality of life. Fibroblast-like synovial cells (FLSs) within the synovial intima exhibit "tumor-like" properties such as increased proliferation, migration, and invasion. Activation of FLSs and secretion of pro-inflammation factors result in pannus formation and cartilage destruction. As an inhibitor of the cytokine, macrophage migration inhibitory factor (MIF), 4-Iodo-6-phenylpyrimidine (4-IPP) has been shown to reduce cell proliferation, migration, invasion, and the secretion of pro-inflammatory mediators in a variety of diseases. However, the usefulness of 4-IPP for RA treatment has not been assessed and was the purpose of this study. In vitro, 4-IPP was demonstrated to inhibit proliferation, migration, and invasion of RA FLSs, as well as the expression of pro-inflammatory cytokines. 4-IPP was also shown to inhibit MIF-induced phosphorylation of ERK, JNK, and p38, as well as reduce expression of COX2 and PGE2. In order to efficiently deliver 4-IPP to anatomical RA sites, we developed lactic-co-glycolic acid (PLGA) nanospheres, which not only protected 4-IPP from degradation but also controlled the release of 4-IPP. 4-IPP/PLGA nanospheres had potent anti-inflammatory activity and a high degree of biosafety. Results showed that local 4-IPP concentration was increased by nanosphere delivery, effectively reducing the inflammatory microenvironment as well as synovial inflammation, joint swelling, and cartilage destruction in a collagen-induced rheumatoid arthritis (CIA) rat model. Therefore, 4-IPP nanospheres are a sustained-release delivery system that may be an effective therapeutic strategy for RA treatment.
Collapse
Affiliation(s)
- Weiwei Sun
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Jinquan Ma
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Minhao Chen
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Weidong Zhang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Chunxiang Xu
- Department of Nursing, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yunyi Nan
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Weijie Wu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Xingxing Mao
- Department of Orthopaedics, The Sixth People's Hospital of Nantong, Nantong, Jiangsu 226001, China
| | - Xi Cheng
- Department of Gynecology and Obstetrics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Hao Cai
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Jianhua Zhang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Hua Xu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Youhua Wang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
10
|
Shi X, Wu Y, Ni H, Li M, Qi B, Xu Y. Macrophage migration inhibitory factor (MIF) inhibitor iSO-1 promotes staphylococcal protein A-induced osteogenic differentiation by inhibiting NF-κB signaling pathway. Int Immunopharmacol 2023; 115:109600. [PMID: 36577150 DOI: 10.1016/j.intimp.2022.109600] [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: 10/04/2022] [Revised: 12/02/2022] [Accepted: 12/12/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Osteomyelitis is among the most difficult to treat diseases in the field of orthopedics, and there is a lack of effective treatment modalities. Exploring the mechanisms of its development is beneficial for finding molecular targets for treatment. Increasing evidence suggests that macrophage migration inhibitory factor (MIF), as a proinflammatory mediator, is not only involved in various pathophysiological processes of inflammation but also plays an important role in osteogenic differentiation, while its specific regulatory mechanism in osteomyelitis remains unclear. METHODS In the present study, staphylococcal protein A (SPA)-treated rat bone marrow mesenchymal stem cells (rBMSCs) were used to construct cell models of osteomyelitis. Rat and cell models of osteomyelitis were used to validate the expression levels of MIF, and to further explore the regulatory mechanisms of the MIF inhibitor methyl ester of (S, R)-3-(4-hydroxyphenyl)-4,5-dihydro-5-isoxazole acetic acid (iSO-1) and MIF knockdown on cell model of osteomyelitis toward osteogenic differentiation. RESULTS We found that the expression level of MIF was upregulated in rat and cell models of osteomyelitis and subsequently demonstrated by the GSE30119 dataset that the expression level of MIF was also significantly upregulated in patients with osteomyelitis. Furthermore, SPA promotes MIF expression in rBMSCs while inhibiting the expression of osteogenic-related genes such as Runt-related transcription factor 2 (RUNX2), osteocalcin (OCN), osteopontin (OPN) and collagen type-1 (COL-1) through activation of the nuclear factor kappa-B (NF-κB) pathway. In vivo, we further demonstrated that local injection of iSO-1 significantly increased the osteogenic activity in rat model of osteomyelitis. Importantly, we also demonstrated that MIF knockdown and the MIF inhibitor iSO-1 reversed the SPA-mediated inhibition of osteogenic differentiation of rBMSCs by inhibiting the activation of the NF-κB pathway, as evidenced by the upregulation of osteogenic-related gene expression and enhanced bone mineralization. CONCLUSION ISO-1 and MIF knockdown can reverse the SPA-mediated inhibition of osteogenic differentiation in the rBMSCs model of osteomyelitis by inhibiting the NF-κB signaling pathway, providing a potential target for the treatment of osteomyelitis.
Collapse
Affiliation(s)
| | - Yipeng Wu
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Haonan Ni
- Kunming Medical University, Kunming, China
| | - Mingjun Li
- Kunming Medical University, Kunming, China
| | | | - Yongqing Xu
- Department of Orthopedic Surgery, 920th Hospital of Joint Logistics Support Force, Kunming, China.
| |
Collapse
|
11
|
Osthole Inhibits M1 Macrophage Polarization and Attenuates Osteolysis in a Mouse Skull Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:2975193. [PMID: 36686380 PMCID: PMC9851800 DOI: 10.1155/2023/2975193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 01/15/2023]
Abstract
Excessive bone resorption due to increased inflammatory factors is a common feature of inflammatory lytic bone diseases. This group of diseases is effectively treated with drugs. In recent years, many studies have reported that traditional Chinese medicine herbs have substantial effects on inflammation, osteoclast differentiation and maturation, and bone destruction. Herein, we investigated the effects of osthole (OST) on lipopolysaccharide- (LPS-) induced macrophage polarization, inflammatory responses, and osteolysis. In vitro, we used immunofluorescence and quantitative real-time polymerase chain reaction assays to confirm whether bone marrow-derived macrophages showed an increased expression of inflammatory factors, such as interleukin-6, iNOS, CCR7, and CD86, in the presence of LPS. However, we found that such expression was suppressed and that the M2 macrophage expression increased in the presence of OST. OST reduced LPS- and RANKL-induced intracellular reactive oxygen species production in the bone marrow-derived macrophages. Further, it potently suppressed osteoclast differentiation and osteoclast-specific gene expression by suppressing the P38/MAPK and NF-κB pathways. Consistent with the in vitro observations, OST greatly ameliorated LPS-induced bone resorption and modulated the ratio of macrophages at the site of osteolysis. Taken together, OST has great potential for use in the management of osteolytic diseases.
Collapse
|
12
|
Zhang Z, Zhou X, Guo J, Zhang F, Qian Y, Wang G, Duan M, Wang Y, Zhao H, Yang Z, Liu Z, Jiang X. TA-MSCs, TA-MSCs-EVs, MIF: their crosstalk in immunosuppressive tumor microenvironment. J Transl Med 2022; 20:320. [PMID: 35842634 PMCID: PMC9287873 DOI: 10.1186/s12967-022-03528-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/08/2022] [Indexed: 11/24/2022] Open
Abstract
As an important component of the immunosuppressive tumor microenvironment (TME), it has been established that mesenchymal stem cells (MSCs) promote the progression of tumor cells. MSCs can directly promote the proliferation, migration, and invasion of tumor cells via cytokines and chemokines, as well as promote tumor progression by regulating the functions of anti-tumor immune and immunosuppressive cells. MSCs-derived extracellular vesicles (MSCs-EVs) contain part of the plasma membrane and signaling factors from MSCs; therefore, they display similar effects on tumors in the immunosuppressive TME. The tumor-promoting role of macrophage migration inhibitory factor (MIF) in the immunosuppressive TME has also been revealed. Interestingly, MIF exerts similar effects to those of MSCs in the immunosuppressive TME. In this review, we summarized the main effects and related mechanisms of tumor-associated MSCs (TA-MSCs), TA-MSCs-EVs, and MIF on tumors, and described their relationships. On this basis, we hypothesized that TA-MSCs-EVs, the MIF axis, and TA-MSCs form a positive feedback loop with tumor cells, influencing the occurrence and development of tumors. The functions of these three factors in the TME may undergo dynamic changes with tumor growth and continuously affect tumor development. This provides a new idea for the targeted treatment of tumors with EVs carrying MIF inhibitors.
Collapse
Affiliation(s)
- Zhenghou Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiangyu Zhou
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jinshuai Guo
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fusheng Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yiping Qian
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guang Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Meiqi Duan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yutian Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Haiying Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhi Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zunpeng Liu
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| | - Xiaofeng Jiang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
13
|
Wu B, Nakamura A. Deep Insight into the Role of MIF in Spondyloarthritis. Curr Rheumatol Rep 2022; 24:269-278. [PMID: 35809213 DOI: 10.1007/s11926-022-01081-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW Pathological roles of macrophage migration inhibitory factor (MIF) have recently been demonstrated in spondyloarthritis (SpA) preclinical models, identifying MIF as a new treatment target for SpA. However, the specific contribution of MIF and therapeutic potential of MIF-targeted therapies to various tissue types affected by SpA are not well delineated. RECENT FINDINGS MIF and its cognate receptor CD74 are extensively involved in the pathogenesis of SpA including inflammation in the spine, joint, eyes, skin, and gut. The majority of the current evidence has consistently shown that MIF drives the inflammation in these distinct anatomical sites. In preclinical models, genetic deletion or blockade of MIF reduces the severity of inflammation. Although MIF is generally an upstream cytokine which regulates downstream effector cytokines, MIF also intensifies type 3 immunity by promoting helper T 17 (Th17) plasticity. MIF- or CD74-targeted therapies have also reported to be well tolerated in clinical trials for other diseases. Recent findings suggest that MIF-CD74 axis is a new therapeutic target for SpA to improve various clinical features. Clinical trials for MIF- or CD74-targeted therapies for SpA patients are warranted.
Collapse
Affiliation(s)
- Brian Wu
- Schroeder Arthritis Institute, University Health Network, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Laboratory Medicine and Pathology, University of Toronto, Toronto, ON, Canada
| | - Akihiro Nakamura
- Schroeder Arthritis Institute, University Health Network, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada. .,Krembil Research Institute, University Health Network, Toronto, ON, Canada. .,Division of Rheumatology, Toronto Western Hospital, University Health Network, Toronto, ON, Canada. .,Institute of Medical Science, Temerty Faculty of Medicine of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
14
|
Macrophage Migration Inhibitory Factor (MIF) as a Stress Molecule in Renal Inflammation. Int J Mol Sci 2022; 23:ijms23094908. [PMID: 35563296 PMCID: PMC9102975 DOI: 10.3390/ijms23094908] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
Renal inflammation is an initial pathological process during progressive renal injury regardless of the initial cause. Macrophage migration inhibitory factor (MIF) is a truly proinflammatory stress mediator that is highly expressed in a variety of both inflammatory cells and intrinsic kidney cells. MIF is released from the diseased kidney immediately upon stimulation to trigger renal inflammation by activating macrophages and T cells, and promoting the production of proinflammatory cytokines, chemokines, and stress molecules via signaling pathways involving the CD74/CD44 and chemokine receptors CXCR2, CXCR4, and CXCR7 signaling. In addition, MIF can function as a stress molecule to counter-regulate the immunosuppressive effect of glucocorticoid in renal inflammation. Given the critical position of MIF in the upstream inflammatory cascade, this review focuses on the regulatory role and molecular mechanisms of MIF in kidney diseases. The therapeutic potential of targeting MIF signaling to treat kidney diseases is also discussed.
Collapse
|
15
|
Cao J, Ma X, Liu L, Zhang G, Wu Y, Fu Y, Gong A, Yang Z, Zhao Y, Zhang L, Li Y. Cortistatin attenuates titanium particle-induced osteolysis through regulation of TNFR1-ROS-caspase-3 signaling in osteoblasts. Ann N Y Acad Sci 2022; 1513:140-152. [PMID: 35419858 DOI: 10.1111/nyas.14774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aseptic loosening is a major complication of prosthetic joint surgery and is associated with impaired osteoblast homeostasis. Cortistatin (CST) is a neuropeptide that protects against inflammatory conditions. In this study, we found that expression of CST was diminished in patients with prosthetic joint loosening and in titanium (Ti) particle-induced animal models. A Ti particle-induced calvarial osteolysis model was established in wild-type and CST gene knockout mice; CST deficiency enhanced, while exogenously added CST attenuated, the severity of Ti particle-mediated osteolysis. CST protected against inflammation as well as apoptosis and maintained the osteogenic function of MC3T3-E1 osteoblasts upon stimulation with Ti particles. Furthermore, CST antagonized reactive oxygen species production and suppressed caspase-3-associated apoptosis mediated by Ti particles in osteoblasts. Additionally, CST protects against Ti particle-induced osteolysis through tumor necrosis factor receptor 1. Taken together, CST might provide a therapeutic strategy for wear debris-induced inflammatory osteolysis.
Collapse
Affiliation(s)
- Jiankang Cao
- Department of Pain, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P. R. China
| | - Xiaojie Ma
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Long Liu
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P. R. China
| | - Gaorui Zhang
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P. R. China
| | - Yawei Wu
- Caoxian People's Hospital, Heze, P. R. China
| | - Yu Fu
- The First Affiliated Hospital of Shandong First Medical University, Jinan, P. R. China
| | - Ao Gong
- Second Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Zhongbo Yang
- Shandong Yellow River Hospital, Yellow River Shandong Bureau, Jinan, P. R. China
| | - Yunpeng Zhao
- Department of Orthopaedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P. R. China
| | - Lei Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Shandong First Medical University, Jinan, P. R. China.,Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, P. R. China
| | - Yuhua Li
- Department of Orthopaedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P. R. China
| |
Collapse
|
16
|
Huang L, Chen W, Wei L, Su Y, Liang J, Lian H, Wang H, Long F, Yang F, Gao S, Tan Z, Xu J, Zhao J, Liu Q. Lonafarnib Inhibits Farnesyltransferase via Suppressing ERK Signaling Pathway to Prevent Osteoclastogenesis in Titanium Particle-Induced Osteolysis. Front Pharmacol 2022; 13:848152. [PMID: 35300293 PMCID: PMC8921770 DOI: 10.3389/fphar.2022.848152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/10/2022] [Indexed: 11/30/2022] Open
Abstract
Wear debris after total joint arthroplasty can attract the recruitment of macrophages, which release pro-inflammatory substances, triggering the activation of osteoclasts, thereby leading to periprosthetic osteolysis (PPOL) and aseptic loosening. However, the development of pharmacological strategies targeting osteoclasts to prevent periprosthetic osteolysis has not been fruitful. In this study, we worked toward researching the effects and mechanisms of a farnesyltransferase (FTase) inhibitor Lonafarnib (Lon) on receptor activator of nuclear factor κB (NF-κB) ligand (RANKL)-induced osteoclastogenesis and bone resorption, as well as the impacts of Lon on titanium particle-induced osteolysis. To investigate the impacts of Lon on bone resorption and osteoclastogenesis in vitro, bone marrow macrophages were incubated and stimulated with RANKL and macrophage colony-stimulating factor (M-CSF). The influence of Lon on osteolysis prevention in vivo was examined utilizing a titanium particle-induced mouse calvarial osteolysis model. The osteoclast-relevant genes expression was explored by real-time quantitative PCR. Immunofluorescence was used to detect intracellular localization of nuclear factor of activated T cells 1 (NFATc1). SiRNA silence assay was applied to examine the influence of FTase on osteoclasts activation. Related signaling pathways, including NFATc1 signaling, NF-κB, mitogen-activated protein kinases pathways were identified by western blot assay. Lon was illustrated to suppress bone resorptive function and osteoclastogenesis in vitro, and it also reduced the production of pro-inflammatory substances and prevented titanium particle-induced osteolysis in vivo. Lon decreased the expression of osteoclast-relevant genes and suppressed NFATc1 nuclear translocation and auto-amplification. Mechanistically, Lon dampened FTase, and inhibition of FTase reduced osteoclast formation by suppressing ERK signaling. Lon is a promising treatment option for osteoclast-related osteolysis diseases including periprosthetic osteolysis by targeted inhibition of FTase through suppressing ERK signaling.
Collapse
Affiliation(s)
- Linke Huang
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Orthopaedics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Weiwei Chen
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Linhua Wei
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China.,The Affiliated Nanning Infectious Disease Hospital of Guangxi Medical University, The Fourth People's Hospital of Nanning, Nanning, China
| | - Yuangang Su
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Jiamin Liang
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Haoyu Lian
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Hui Wang
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Feng Long
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Fan Yang
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Shiyao Gao
- Department of Orthopaedics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhen Tan
- Department of Orthopaedics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jinmin Zhao
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Qian Liu
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
17
|
Xu X, Li Y, Shi L, He K, Sun Y, Ding Y, Meng B, Zhang J, Xiang L, Dong J, Liu M, Zhang J, Xiang L, Xiang G. Myeloid-derived growth factor (MYDGF) protects bone mass through inhibiting osteoclastogenesis and promoting osteoblast differentiation. EMBO Rep 2022; 23:e53509. [PMID: 35068044 PMCID: PMC8892248 DOI: 10.15252/embr.202153509] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Whether bone marrow regulates bone metabolism through endocrine and paracrine mechanism remains largely unknown. Here, we found that (i) myeloid cell-specific myeloid-derived growth factor (MYDGF) deficiency decreased bone mass and bone strength in young and aged mice; (ii) myeloid cell-specific MYDGF restoration prevented decreases in bone mass and bone strength in MYDGF knockout mice; moreover, myeloid cell-derived MYDGF improved the progress of bone defects healing, prevented ovariectomy (OVX)-induced bone loss and age-related osteoporosis; (iii) MYDGF inhibited osteoclastogenesis and promoted osteoblast differentiation in vivo and in vitro; and (iv) PKCβ-NF-κB and MAPK1/3-STAT3 pathways were involved in the regulation of MYDGF on bone metabolism. Thus, we concluded that myeloid cell-derived MYDGF is a positive regulator of bone homeostasis by inhibiting bone resorption and promoting bone formation. MYDGF may become a potential novel therapeutic drug for osteoporosis, and bone marrow may become a potential therapeutic target for bone metabolic disorders.
Collapse
Affiliation(s)
- Xiaoli Xu
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Yixiang Li
- Department of Hematology and Medical OncologySchool of MedicineEmory UniversityAtlantaGAUSA
| | - Lingfeng Shi
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Kaiyue He
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Ying Sun
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Yan Ding
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Biying Meng
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Jiajia Zhang
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Lin Xiang
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Jing Dong
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Min Liu
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina
| | - Junxia Zhang
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Lingwei Xiang
- Centers for Surgery and Public HealthBrigham and Women's HospitalBostonMAUSA
| | - Guangda Xiang
- Department of EndocrinologyGeneral Hospital of Central Theater CommandWuhanChina,The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
18
|
Schwann Cells Accelerate Osteogenesis via the Mif/CD74/FOXO1 Signaling Pathway In Vitro. Stem Cells Int 2022; 2022:4363632. [PMID: 35069747 PMCID: PMC8776480 DOI: 10.1155/2022/4363632] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/13/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022] Open
Abstract
Schwann cells have been found to promote osteogenesis by an unclear molecular mechanism. To better understand how Schwann cells accelerate osteogenesis, RNA-Seq and LC-MS/MS were utilized to explore the transcriptomic and metabolic response of MC3T3-E1 to Schwann cells. Osteogenic differentiation was determined by ALP staining. Lentiviruses were constructed to alter the expression of Mif (macrophage migration inhibitory factor) in Schwann cells. Western blot (WB) analysis was employed to detect the protein expression. The results of this study show that Mif is essential for Schwann cells to promote osteogenesis, and its downstream CD74/FOXO1 is also involved in the promotion of Schwann cells on osteogenesis. Further, Schwann cells regulate amino acid metabolism and lipid metabolism in preosteoblasts. These findings unveil the mechanism for Schwann cells to promote osteogenesis where Mif is a key factor.
Collapse
|
19
|
Zheng L, Feng Z, Tao S, Gao J, Lin Y, Wei X, Zheng B, Huang B, Zheng Z, Zhang X, Liu J, Shan Z, Chen Y, Chen J, Zhao F. Destabilization of macrophage migration inhibitory factor by 4-IPP reduces NF-κB/P-TEFb complex-mediated c-Myb transcription to suppress osteosarcoma tumourigenesis. Clin Transl Med 2022; 12:e652. [PMID: 35060345 PMCID: PMC8777168 DOI: 10.1002/ctm2.652] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND As an inflammatory factor and oncogenic driver protein, the pleiotropic cytokine macrophage migration inhibitory factor (MIF) plays a crucial role in the osteosarcoma microenvironment. Although 4-iodo-6-phenylpyrimidine (4-IPP) can inactivate MIF biological functions, its anti-osteosarcoma effect and molecular mechanisms have not been investigated. In this study, we identified the MIF inhibitor 4-IPP as a specific double-effector drug for osteosarcoma with both anti-tumour and anti-osteoclastogenic functions. METHODS The anti-cancer effects of 4-IPP were evaluated by wound healing assay, cell cycle analysis, colony formation assay, CCK-8 assay, apoptosis analysis, and Transwell migration/invasion assays. Through the application of a luciferase reporter, chromatin immunoprecipitation assays, and immunofluorescence and coimmunoprecipitation analyses, the transcriptional regulation of the NF-κB/P-TEFb complex on c-Myb- and STUB1-mediated proteasome-dependent MIF protein degradation was confirmed. The effect of 4-IPP on tumour growth and metastasis was assessed using an HOS-derived tail vein metastasis model and subcutaneous and orthotopic xenograft tumour models. RESULTS In vitro, 4-IPP significantly reduced the proliferation and metastasis of osteosarcoma cells by suppressing the NF-κB pathway. 4-IPP hindered the binding between MIF and CD74 as well as p65. Moreover, 4-IPP inhibited MIF to interrupt the formation of downstream NF-κB/P-TEFb complexes, leading to the down-regulation of c-Myb transcription. Interestingly, the implementation of 4-IPP can mediate small molecule-induced MIF protein proteasomal degradation via the STUB1 E3 ligand. However, 4-IPP still interrupted MIF-mediated communication between osteosarcoma cells and osteoclasts, thus promoting osteoclastogenesis. Remarkably, 4-IPP strongly reduced HOS-derived xenograft osteosarcoma tumourigenesis and metastasis in an in vivo mouse model. CONCLUSIONS Our findings demonstrate that the small molecule 4-IPP targeting the MIF protein exerts an anti-osteosarcoma effect by simultaneously inactivating the biological functions of MIF and promoting its proteasomal degradation. Direct destabilization of the MIF protein with 4-IPP may be a promising therapeutic strategy for treating osteosarcoma.
Collapse
Affiliation(s)
- Lin Zheng
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Zhenhua Feng
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Siyue Tao
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Jiawei Gao
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Ye Lin
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Xiaoan Wei
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Bingjie Zheng
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Bao Huang
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Zeyu Zheng
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Xuyang Zhang
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Junhui Liu
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Zhi Shan
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Yilei Chen
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Jian Chen
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| | - Fengdong Zhao
- Department of Orthopaedic SurgerySir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province3 East Qingchun RoadHangzhouZhejiang Province310016China
| |
Collapse
|
20
|
Sun X, Zhu K, Feng C, Zhu J, Chen S, Tang W, Wang Z, Xiao L, Li H, Geng D, Wang Z. Paeoniflorin Ameliorates Hyperprolactinemia-Induced Inhibition of Osteoblastogenesis by Suppressing the NF- κB Signaling Pathway. Int J Endocrinol 2022; 2022:4572033. [PMID: 35465073 PMCID: PMC9033376 DOI: 10.1155/2022/4572033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 02/16/2022] [Accepted: 03/15/2022] [Indexed: 11/17/2022] Open
Abstract
Hyperprolactinemia is a common endocrine disease in women of reproductive age. Research has shown that patients with hyperprolactinemia often have decreased bone mineral density and an increased risk of fractures. However, there is still a lack of effective treatments. Paeoniflorin, one of the primary bioactive components in peony, is widely used in traditional Chinese medicine. Research has shown that paeoniflorin promotes osteoblast differentiation. However, whether paeoniflorin plays a role in hyperprolactinemia-induced osteoblastogenesis inhibition is not yet clear. In this study, we investigated the effect of paeoniflorin on prolactin (PRL)-mediated inhibition of osteoblast function. Our results showed that prolactin significantly reduced the expression of alkaline phosphatase (ALP), Osterix, and runt-related transcription factor 2 (RUNX2) in MC3T3-E1 cells cultured in an osteoblast differentiation medium, suggesting that prolactin inhibited osteoblast function. After treatment with paeoniflorin (PF), the expression of these osteoblast markers was upregulated. In addition, our findings proved that paeoniflorin increased the absorbance values of ALP-positive cells and the areas of alizarin red S (ARS) deposition compared to those in the prolactin group, suggesting that paeoniflorin reversed the PRL-induced reduction in osteoblast differentiation. The PRL-induced activation of nuclear factor kappa B (NF-κB) was significantly reversed by paeoniflorin, indicating that paeoniflorin promoted osteoblast function by inhibiting the NF-κB signaling pathway. In summary, these results showed that paeoniflorin alleviated the inhibitory effect of prolactin on osteoblastogenesis by suppressing the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiaohong Sun
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
- Department of Endocrinology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Keda Zhu
- Department of Endocrinology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Chengcheng Feng
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
- Department of Endocrinology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Jie Zhu
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
- Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Shuangshuang Chen
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
- Department of Endocrinology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Wenkai Tang
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
- Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Zhifang Wang
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Long Xiao
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
- Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Hong Li
- Department of Endocrinology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Zhirong Wang
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
- Department of Orthopedics, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, China
| |
Collapse
|
21
|
Pan B, Zheng L, Fang J, Lin Y, Lai H, Gao J, Pan W, Zhang Y, Ni K, Lou C, He D. Azilsartan Suppresses Osteoclastogenesis and Ameliorates Ovariectomy-Induced Osteoporosis by Inhibiting Reactive Oxygen Species Production and Activating Nrf2 Signaling. Front Pharmacol 2021; 12:774709. [PMID: 34899338 PMCID: PMC8662525 DOI: 10.3389/fphar.2021.774709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
Osteoporosis is characterized by a decrease in bone mass and destruction of the bone microarchitecture, and it commonly occurs in postmenopausal women and the elderly. Overactivation of osteoclasts caused by the inflammatory response or oxidative stress leads to osteoporosis. An increasing number of studies have suggested that intracellular reactive oxygen species (ROS) are strongly associated with osteoclastogenesis. As a novel angiotensin (Ang) II receptor blocker (ARB), azilsartan was reported to be associated with the inhibition of intracellular oxidative stress processes. However, the relationship between azilsartan and osteoclastogenesis is still unknown. In this study, we explored the effect of azilsartan on ovariectomy-induced osteoporosis in mice. Azilsartan significantly inhibited the receptor activator of nuclear factor-κB ligand (RANKL)-mediated osteoclastogenesis and downregulated the expression of osteoclast-associated markers (Nfatc1, c-Fos, and Ctsk) in vitro. Furthermore, azilsartan reduced RANKL-induced ROS production by increasing the expression of nuclear factor erythroid 2-related factor 2 (Nrf2). Mechanistically, azilsartan inhibited the activation of MAPK/NF-κB signaling pathways, while Nrf2 silencing reversed the inhibitory effect of azilsartan on MAPK/NF-κB signaling pathways. Consistent with the in vitro data, azilsartan administration ameliorated ovariectomy (OVX)-induced osteoporosis, and decreased ROS levels in vivo. In conclusion, azilsartan inhibited oxidative stress and may be a novel treatment strategy for osteoporosis caused by osteoclast overactivation.
Collapse
Affiliation(s)
- Bin Pan
- Department of Orthopedics, Lishui hospital, Zhejiang University School of Medicine, Lishui, China.,Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research of Zhejiang Province, Lishui hospital, Lishui, China
| | - Lin Zheng
- Department of Orthopedics, Lishui hospital, Zhejiang University School of Medicine, Lishui, China.,Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiawei Fang
- Department of Orthopedics, Lishui hospital, Zhejiang University School of Medicine, Lishui, China.,Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research of Zhejiang Province, Lishui hospital, Lishui, China
| | - Ye Lin
- Department of Orthopedics, Lishui hospital, Zhejiang University School of Medicine, Lishui, China.,Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research of Zhejiang Province, Lishui hospital, Lishui, China
| | - Hehuan Lai
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research of Zhejiang Province, Lishui hospital, Lishui, China
| | - Jiawei Gao
- Department of Orthopedics, Lishui hospital, Zhejiang University School of Medicine, Lishui, China.,Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research of Zhejiang Province, Lishui hospital, Lishui, China
| | - Wenzheng Pan
- Department of Orthopedics, Lishui hospital, Zhejiang University School of Medicine, Lishui, China.,Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research of Zhejiang Province, Lishui hospital, Lishui, China
| | - Yejin Zhang
- Department of Orthopedics, Lishui hospital, Zhejiang University School of Medicine, Lishui, China.,Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research of Zhejiang Province, Lishui hospital, Lishui, China
| | - Kainan Ni
- Department of Orthopedics, Lishui hospital, Zhejiang University School of Medicine, Lishui, China.,Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research of Zhejiang Province, Lishui hospital, Lishui, China
| | - Chao Lou
- Department of Orthopedics, Lishui hospital, Zhejiang University School of Medicine, Lishui, China.,Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research of Zhejiang Province, Lishui hospital, Lishui, China
| | - Dengwei He
- Department of Orthopedics, Lishui hospital, Zhejiang University School of Medicine, Lishui, China.,Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research of Zhejiang Province, Lishui hospital, Lishui, China
| |
Collapse
|
22
|
Xiao Z, Chen D, Mulder F, Song S, van der Wouden PE, Cool RH, Melgert BN, Poelarends GJ, Dekker FJ. 4-Iodopyrimidine Labeling Reveals Nuclear Translocation and Nuclease Activity for Both MIF and MIF2*. Chemistry 2021; 28:e202103030. [PMID: 34724273 PMCID: PMC9299485 DOI: 10.1002/chem.202103030] [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: 08/18/2021] [Indexed: 11/18/2022]
Abstract
Macrophage migration inhibitory factor (MIF) and its homolog MIF2 (also known as D‐dopachrome tautomerase or DDT) play key roles in cell growth and immune responses. MIF and MIF2 expression is dysregulated in cancers and neurodegenerative diseases. Accurate and convenient detection of MIF and MIF2 will facilitate research on their roles in cancer and other diseases. Herein, we report the development and application of a 4‐iodopyrimidine based probe 8 for the selective labeling of MIF and MIF2. Probe 8 incorporates a fluorophore that allows in situ imaging of these two proteins. This enabled visualization of the translocation of MIF2 from the cytoplasm to the nucleus upon methylnitronitrosoguanidine stimulation of HeLa cells. This observation, combined with literature on nuclease activity for MIF, enabled the identification of nuclease activity for MIF2 on human genomic DNA.
Collapse
Affiliation(s)
- Zhangping Xiao
- Department Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Deng Chen
- Department Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Fabian Mulder
- Department Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Shanshan Song
- Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Petra E van der Wouden
- Department Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Robbert H Cool
- Department Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Barbro N Melgert
- Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.,University Medical Center Groningen, Groningen Research Institute of Asthma and COPD, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Gerrit J Poelarends
- Department Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Frank J Dekker
- Department Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| |
Collapse
|
23
|
Deng M, Tan J, Dai Q, Luo F, Xu J. Macrophage-Mediated Bone Formation in Scaffolds Modified With MSC-Derived Extracellular Matrix Is Dependent on the Migration Inhibitory Factor Signaling Pathway. Front Cell Dev Biol 2021; 9:714011. [PMID: 34621738 PMCID: PMC8490662 DOI: 10.3389/fcell.2021.714011] [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: 05/24/2021] [Accepted: 08/09/2021] [Indexed: 12/25/2022] Open
Abstract
The positive role of macrophages in the osteogenesis of mesenchymal stem cells (MSCs) has been a recent research focus. On the other hand, MSCs could carefully regulate the paracrine molecules derived from macrophages. Human umbilical cord mesenchymal stem cells (hucMSCs) can reduce the secretion of inflammatory factors from macrophages to improve injury healing. hucMSC-derived extracellular matrix (hucMSC-ECM) has the similar effect to hucMSCs, which could combat the inflammatory response of macrophages. Additionally, MSC-derived extracellular matrix also enhanced bone regeneration by inhibiting osteoclastic differentiation of monocyte/macrophage lineage. However, whether hucMSC-ECM could improve bone formation by guiding macrophage-induced osteogenic differentiation of MSCs is unknown. Here, we present decalcified bone scaffolds modified by hucMSC-derived extracellular matrix (DBM-ECM), which maintained multiple soluble cytokines from hucMSCs, including macrophage migration inhibitory factor (MIF). Compared with DBM, the DBM-ECM scaffolds induced bone formation in an improved heterotopic ossification model of severe combined immunodeficiency (SCID) mice in a macrophage-dependent manner. Macrophages cocultured with DBM-ECM expressed four osteoinductive cytokines (BMP2, FGF2, TGFβ3 and OSM), which were screened out by RNA sequencing and measured by qPCR and western blot. The conditioned medium from macrophages cocultured with DBM-ECM improved the osteogenic differentiation of hBMSCs. Furthermore, DBM-ECM activated CD74/CD44 (the typical MIF receptors) signal transduction in macrophages, including phosphorylation of P38 and dephosphorylation of c-jun. On the other side, the inhibitory effects of the DBM-ECM scaffolds with a deficient of MIF on osteogenesis in vitro and in vivo revealed that macrophage-mediated osteogenesis depended on MIF/CD74 signal transduction. The results of this study indicate that the coordinated crosstalk of macrophages and MSCs plays a key role on bone regeneration, with an emphasis on hucMSC-ECM constructing a macrophage-derived osteoinductive microenvironment.
Collapse
Affiliation(s)
- Moyuan Deng
- Department of Orthopaedics, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jiulin Tan
- Department of Orthopaedics, Southwest Hospital, Army Medical University, Chongqing, China
| | - Qijie Dai
- Department of Orthopaedics, Southwest Hospital, Army Medical University, Chongqing, China
| | - Fei Luo
- Department of Orthopaedics, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jianzhong Xu
- Department of Orthopaedics, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
24
|
Han Y, Wang J, Li S, Li Y, Zhang Y, Zhang R, Zhang Y, Fan H, Shi H, Pan J, Song G, Ge L, Wang L. Isopsoralen ameliorates rheumatoid arthritis by targeting MIF. Arthritis Res Ther 2021; 23:243. [PMID: 34535196 PMCID: PMC8447788 DOI: 10.1186/s13075-021-02619-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 08/27/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Isopsoralen (IPRN), one of the active ingredients of Psoralea corylifolia Linn, has anti-inflammatory properties. We attempted to investigate the inhibitory effects of IPRN on rheumatoid arthritis (RA) and characterize its potential mechanism. METHODS RA fibroblast-like synoviocytes (FLSs) and mice with collagen-induced arthritis (CIA) were used as in vitro and in vivo models to analyze the antiarthritic effect of IPRN. Histological analysis of the inflamed joints from mice with CIA was performed using microcomputed tomography (micro-CT) and hematoxylin-eosin (HE) staining. RNA sequencing (RNA-Seq), network pharmacology analysis, molecular docking, drug affinity responsive target stability (DARTS) assay, and cellular thermal shift assay (CETSA) were performed to evaluate the targets of IPRN. RESULTS IPRN ameliorated the inflammatory phenotype of RA FLSs by inhibiting their cytokine production, migration, invasion, and proangiogenic ability. IPRN also significantly reduced the severity of CIA in mice by decreasing paw thickness, arthritis score, bone damage, and serum inflammatory cytokine levels. A mechanistic study demonstrated that macrophage migration inhibitory factor (MIF), a key protein in the inflammatory process, was the specific target by which IPRN exerted its anti-inflammatory effects in RA FLSs. CONCLUSION Our study demonstrates the antiarthritic effect of IPRN, which suggests the therapeutic potential of IPRN in RA.
Collapse
Affiliation(s)
- Yi Han
- China Academy of Chinese Medical Sciences, Guang'anmen Hospital, Beijing, China
| | - Jinguang Wang
- Department of Orthopedics, Dezhou People's Hospital, Dezhou, Shandong, China
| | - Shufeng Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Yi Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yongli Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Ruojia Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuang Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China
| | - Huancai Fan
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Haojun Shi
- The Second Clinical Medical College, Henan University of Chinese Medicine, Jinan, China
| | - Jihong Pan
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China
| | - Guanhua Song
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Luna Ge
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China.
| | - Lin Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China.
| |
Collapse
|
25
|
Jin K, Zheng L, Ye L, Xie Z, Gao J, Lou C, Pan W, Pan B, Liu S, Chen Z, He D. Chicago sky blue 6B (CSB6B), an allosteric inhibitor of macrophage migration inhibitory factor (MIF), suppresses osteoclastogenesis and promotes osteogenesis through the inhibition of the NF-κB signaling pathway. Biochem Pharmacol 2021; 192:114734. [PMID: 34411569 DOI: 10.1016/j.bcp.2021.114734] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 10/20/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic pro-inflammatory mediator involved in various pathophysiological and inflammatory states. Accumulating line of evidence suggests a role for MIF in regulating bone metabolism and therefore a prime candidate for therapeutic targeting. In this study, we showed that Chicago sky blue 6B (CSB6B) suppresses RANKL-induced osteoclast and bone resorption in vitro via the inhibition of NF-κB signaling activation and promoting proteasome-mediated degradation of MIF. Consequently, the induction of NFATc1 was impaired resulting in downregulation of NFATc1-responsive osteoclast genes. We also demonstrated that CSB6B treatment enhanced primary calvarial osteoblast differentiation and bone mineralization in vitro via the suppression of NF-κB activation and upregulation of Runx expression. Using two murine models of osteolytic bone disorders, we further showed that administration of CSB6B protected mice against pathological inflammatoryc calvarial bone destruction induced by titanium particles mice as well as estrogen-deficiency induced bone loss as a result of ovariectomy. Together, as an MIF inhibitor, CSB6B can inhibit osteoclast differentiation and bone resorption function and enhance the mineralization of osteoblasts through the inhibition of NF-κB pathway. MIF is a prime target for therapeutic targeting for the treatment of osteolytic bone disorders and the MIF inhibitor CSB6B could be potential anti-osteoporosis drug.
Collapse
Affiliation(s)
- Kangtao Jin
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/Lishui Central Hospital, Lishui, China
| | - Lin Zheng
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/Lishui Central Hospital, Lishui, China; Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Lin Ye
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/Lishui Central Hospital, Lishui, China
| | - Ziang Xie
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jiawei Gao
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/Lishui Central Hospital, Lishui, China
| | - Chao Lou
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/Lishui Central Hospital, Lishui, China
| | - Wenzheng Pan
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/Lishui Central Hospital, Lishui, China
| | - Bin Pan
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/Lishui Central Hospital, Lishui, China
| | - Shijie Liu
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/Lishui Central Hospital, Lishui, China
| | - Zhenzhong Chen
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/Lishui Central Hospital, Lishui, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Dengwei He
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/Lishui Central Hospital, Lishui, China.
| |
Collapse
|
26
|
Zheng H, Feng H, Zhang W, Han Y, Zhao W. Targeting autophagy by natural product Ursolic acid for prevention and treatment of osteoporosis. Toxicol Appl Pharmacol 2020; 409:115271. [PMID: 33065153 DOI: 10.1016/j.taap.2020.115271] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
With the growth of the aging population, osteoporosis is becoming a global health problem. Ursolic acid (UA) is an active ingredient existed in a variety of foods and nature plants and owns plenty of pharmacological effects especially in treating metabolic disease. Our predication from network pharmacology hinted that UA has potential for ameliorating osteoporosis. Firstly through in vivo experiment, we confirmed that UA administration obviously protected against ovariectomy (OVX)-induced osteoporosis in rats by improving microarchitectural deterioration of trabecular bone (P < 0.001), decreasing numbers of TRAP positive osteoclast in vertebra (P < 0.001), as well as decreasing serum osteoclast-specific cytokines release (P < 0.001). Besides, UA ameliorated kidney damage secondary to OVX-induced osteoporosis by ameliorating glomerular atrophy, decreasing BUN and creatinine levels in OVX rats. In vitro, UA noticeably decreased osteoclastic-special marker proteins c-Fos and NFATc1 expressions (P < 0.001) in response to RANKL stimulation in macrophagy. Importantly, autophagy pathway was activated in the process of osteoclast differentiation and blocked by UA pretreatment. Furthermore, autophagy inhibitors suppressed osteoclast differentiation (P < 0.001). Collectively, UA may ameliorate osteoporosis by suppressing osteoclast differentiation mediated by autophagy. Our research provides scientific support for UA treating osteoporosis and offers an optimal dose for daily intake of UA safely to prevent bone diseases.
Collapse
Affiliation(s)
- Haoyi Zheng
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China
| | - Haitao Feng
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Wenzhong Zhang
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Yantao Han
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China
| | - Wenwen Zhao
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China.
| |
Collapse
|
27
|
MIF inhibitor, ISO-1, attenuates human pancreatic cancer cell proliferation, migration and invasion in vitro, and suppresses xenograft tumour growth in vivo. Sci Rep 2020; 10:6741. [PMID: 32317702 PMCID: PMC7174354 DOI: 10.1038/s41598-020-63778-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 04/06/2020] [Indexed: 02/07/2023] Open
Abstract
This study sought to investigate the biological effects of specific MIF inhibitor, ISO-1, on the proliferation, migration and invasion of PANC-1 human pancreatic cells in vitro, and on tumour growth in a xenograft tumour model in vivo. The effect of ISO-1 on PANC-1 cell proliferation was examined using CCK-8 cell proliferation assay. The effect of ISO-1 on collective cell migration and recolonization of PANC-1 cells was evaluated using the cell-wound closure migration assay. The effect of ISO-1 on the migration and invasion of individual PANC-1 cells in a 3-dimensional environment in response to a chemo-attractant was investigated through the use of Transwell migration/invasion assays. Quantitative real time PCR and western blot analyses were employed to investigate the effects of ISO-1 on MIF, NF-κB p65 and TNF-α mRNA and protein expression respectively. Finally, a xenograft tumor model in BALB/c nude mice were used to assess the in vivo effects of ISO-1 on PANC-1-induced tumor growth. We found high expression of MIF in pancreatic cancer tissues. We demonstrated that ISO-1 exerts anti-cancer effects on PANC-1 cell proliferation, migration and invasion in vitro, and inhibited PANC-1 cell-induced tumour growth in xenograft mice in vivo. Our data suggests that ISO-1 and its derivative may have potential therapeutic applications in pancreatic cancer.
Collapse
|
28
|
Ye C, Hou W, Chen M, Lu J, Chen E, Tang L, Hang K, Ding Q, Li Y, Zhang W, He R. IGFBP7 acts as a negative regulator of RANKL-induced osteoclastogenesis and oestrogen deficiency-induced bone loss. Cell Prolif 2019; 53:e12752. [PMID: 31889368 PMCID: PMC7046308 DOI: 10.1111/cpr.12752] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/04/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Insulin-like growth factor-binding protein 7 (IGFBP7) is a low-affinity insulin growth factor (IGF) binder that may play an important role in bone metabolism. We previously reported that IGFBP7 enhanced osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) via the Wnt/β-catenin signalling pathway. In this study, we tried to reveal its function in osteoclast differentiation and osteoporosis. METHODS We used both in vitro and in vivo studies to investigate the effects of IGFBP7 on RANKL-induced osteoclastogenesis and osteoporosis, together with the underlying molecular mechanisms of these processes. RESULTS We show that IGFBP7 inhibited receptor activation of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclastogenesis, F-actin ring formation and bone resorption, which was confirmed by using recombinant IGFBP7 protein, lentivirus and siRNA. The NF-κB signalling pathway was inhibited during this process. Moreover, in a mouse ovariectomy-induced osteoporosis model, IGFBP7 treatment attenuated osteoporotic bone loss by inhibiting osteoclast activity. CONCLUSIONS Taken together, these findings show that IGFBP7 suppressed osteoclastogenesis in vitro and in vivo and suggest that IGFBP7 is a negative regulator of osteoclastogenesis and plays a protective role in osteoporosis. These novel insights into IGFBP7 may facilitate the development of potential treatment strategies for oestrogen deficiency-induced osteoporosis and other osteoclast-related disorders.
Collapse
Affiliation(s)
- Chenyi Ye
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Weiduo Hou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Mo Chen
- Department of Rheumatology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jinwei Lu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Erman Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Lan Tang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Kai Hang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Qianhai Ding
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Yan Li
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Wei Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Rongxin He
- Department of Orthopedic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Zhang Y, Zhu W, He H, Fan B, Deng R, Hong Y, Liang X, Zhao H, Li X, Zhang F. Macrophage migration inhibitory factor rejuvenates aged human mesenchymal stem cells and improves myocardial repair. Aging (Albany NY) 2019; 11:12641-12660. [PMID: 31881006 PMCID: PMC6949107 DOI: 10.18632/aging.102592] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022]
Abstract
The beneficial functions of mesenchymal stem cells (MSCs) decline with age, limiting their therapeutic efficacy for myocardial infarction (MI). Macrophage migration inhibitory factor (MIF) promotes cell proliferation and survival. We investigated whether MIF overexpression could rejuvenate aged MSCs and increase their therapeutic efficacy in MI. Young and aged MSCs were isolated from the bone marrow of young and aged donors. Young MSCs, aged MSCs, and MIF-overexpressing aged MSCs were transplanted into the peri-infarct region in a rat MI model. Aged MSCs exhibited a lower proliferative capacity, lower MIF level, greater cell size, greater senescence-associated-β-galactosidase activity, and weaker paracrine effects than young MSCs. Knocking down MIF in young MSCs induced cellular senescence, whereas overexpressing MIF in aged MSCs reduced cellular senescence. MIF rejuvenated aged MSCs by activating autophagy, an effect largely reversed by the autophagy inhibitor 3-methyladenine. MIF-overexpressing aged MSCs induced angiogenesis and prevented cardiomyocyte apoptosis to a greater extent than aged MSCs, and had improved heart function and cell survival more effectively than aged MSCs four weeks after MI. Thus, MIF rejuvenated aged MSCs by activating autophagy and enhanced their therapeutic efficacy in MI, suggesting a novel MSC-based therapeutic strategy for cardiovascular diseases in the aged population.
Collapse
Affiliation(s)
- Yuelin Zhang
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wenwu Zhu
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haiwei He
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Baohan Fan
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Deng
- Department of General Surgery, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yimei Hong
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaoting Liang
- Clinical Translational Medical Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hongyan Zhao
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Li
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fengxiang Zhang
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|