1
|
Sun K, Li H, Dong Y, Cao L, Li D, Li J, Zhang M, Yan D, Yang B. The Use of Identified Hypoxia-related Genes to Generate Models for Predicting the Prognosis of Cerebral Ischemia‒reperfusion Injury and Developing Treatment Strategies. Mol Neurobiol 2025; 62:3098-3124. [PMID: 39230867 PMCID: PMC11790705 DOI: 10.1007/s12035-024-04433-9] [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: 10/07/2023] [Accepted: 08/08/2024] [Indexed: 09/05/2024]
Abstract
Cerebral ischemia‒reperfusion injury (CIRI) is a type of secondary brain damage caused by reperfusion after ischemic stroke due to vascular obstruction. In this study, a CIRI diagnostic model was established by identifying hypoxia-related differentially expressed genes (HRDEGs) in patients with CIRI. The ischemia‒reperfusion injury (IRI)-related datasets were downloaded from the Gene Expression Omnibus (GEO) database ( http://www.ncbi.nlm.nih.gov/geo ), and hypoxia-related genes in the Gene Cards database were identified. After the datasets were combined, hypoxia-related differentially expressed genes (HRDEGs) expressed in CIRI patients were identified. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of the HRDEGs were performed using online tools. Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) were performed with the combined gene dataset. CIRI diagnostic models based on HRDEGs were constructed via least absolute shrinkage and selection operator (LASSO) regression analysis and a support vector machine (SVM) algorithm. The efficacy of the 9 identified hub genes for CIRI diagnosis was evaluated via mRNA‒microRNA (miRNA) interaction, mRNA-RNA-binding protein (RBP) network interaction, immune cell infiltration, and receiver operating characteristic (ROC) curve analyses. We then performed logistic regression analysis and constructed logistic regression models based on the expression of the 9 HRDEGs. We next established a nomogram and calibrated the prediction data. Finally, the clinical utility of the constructed logistic regression model was evaluated via decision curve analysis (DCA). This study revealed 9 critical genes with high diagnostic value, offering new insights into the diagnosis and selection of therapeutic targets for patients with CIRI. : Not applicable.
Collapse
Affiliation(s)
- Kaiwen Sun
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Hongwei Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yang Dong
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Lei Cao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dongpeng Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jinghong Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Manxia Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dongming Yan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Bo Yang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
2
|
Xu K, Huang RQ, Wen RM, Yao TT, Cao Y, Chang B, Cheng Y, Yi XJ. Annexin A family: A new perspective on the regulation of bone metabolism. Biomed Pharmacother 2024; 178:117271. [PMID: 39121589 DOI: 10.1016/j.biopha.2024.117271] [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: 05/31/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
Osteoblast-mediated bone formation and osteoclast-mediated bone resorption are critical processes in bone metabolism. Annexin A, a calcium-phospholipid binding protein, regulates the proliferation and differentiation of bone cells, including bone marrow mesenchymal stem cells, osteoblasts, and osteoclasts, and has gradually become a marker gene for the diagnosis of osteoporosis. As calcium channel proteins, the annexin A family members are closely associated with mechanical stress, which can target annexins A1, A5, and A6 to promote bone cell differentiation. Despite the significant clinical potential of annexin A family members in bone metabolism, few studies have reported on these mechanisms. Therefore, based on a review of relevant literature, this article elaborates on the specific functions and possible mechanisms of annexin A family members in bone metabolism to provide new ideas for their application in the prevention and treatment of bone diseases, such as osteoporosis.
Collapse
Affiliation(s)
- Ke Xu
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning 110102, China.
| | - Rui-Qi Huang
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning 110102, China.
| | - Rui-Ming Wen
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning 110102, China.
| | - Ting-Ting Yao
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning 110102, China.
| | - Yang Cao
- Graduate School, Anhui University of Traditional Chinese Medicine, Heifei, Anhui 230012, China.
| | - Bo Chang
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning 110102, China.
| | - Yang Cheng
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning 110102, China.
| | - Xue-Jie Yi
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning 110102, China.
| |
Collapse
|
3
|
Liu Q, Lu Z, Ren H, Fu L, Wang Y, Bu H, Ma M, Ma L, Huang C, Wang J, Zang W, Cao J, Fan X. Cav3.2 T-Type calcium channels downregulation attenuates bone cancer pain induced by inhibiting IGF-1/HIF-1α signaling pathway in the rat spinal cord. J Bone Oncol 2023; 42:100495. [PMID: 37583441 PMCID: PMC10423893 DOI: 10.1016/j.jbo.2023.100495] [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: 03/21/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/17/2023] Open
Abstract
Background Bone cancer pain (BCP) is one of the most ubiquitous and refractory symptoms of cancer patients that needs to be urgently addressed. Substantial studies have revealed the pivotal role of Cav3.2 T-type calcium channels in chronic pain, however, its involvement in BCP and the specific molecular mechanism have not been fully elucidated. Methods The expression levels of Cav3.2, insulin-like growth factor 1(IGF-1), IGF-1 receptor (IGF-1R) and hypoxia-inducible factor-1α (HIF-1α) were detected by Western blot in tissues and cells. X-ray and Micro CT used to detect bone destruction in rats. Immunofluorescence was used to detect protein expression and spatial location in the spinal dorsal horn. Electrophoretic mobility shift assay used to verify the interaction between HIF-1α and Cav3.2. Results The results showed that the expression of Cav3.2 channel was upregulated and blockade of this channel alleviated mechanical allodynia and thermal hyperalgesia in BCP rats. Additionally, inhibition of IGF-1/IGF-1R signaling not only reversed the BCP-induced upregulation of Cav3.2 and HIF-1α, but also decreased nociceptive hypersensitivity in BCP rats. Inhibition of IGF-1 increased Cav3.2 expression levels, which were abolished by pretreatment with HIF-1α siRNA in PC12 cells. Furthermore, nuclear HIF-1α bound to the promoter of Cav3.2 to regulate the Cav3.2 transcription level, and knockdown of HIF-1α suppresses the IGF-1-induced upregulation of Cav3.2 and pain behaviors in rats with BCP. Conclusion These findings suggest that spinal Cav3.2 T-type calcium channels play a central role during the development of bone cancer pain in rats via regulation of the IGF-1/IGF-1R/HIF-1α pathway.
Collapse
Affiliation(s)
- Qingying Liu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhongyuan Lu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huan Ren
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Lijun Fu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yueliang Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huilian Bu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Minyu Ma
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Letian Ma
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Chen Huang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Weidong Zang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Jing Cao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
4
|
Yang L, Fu Q, Yang L, Zhang Y. HIF-1α/MMP-9 promotes spinal cord central sensitization in rats with bone cancer pain. Eur J Pharmacol 2023; 954:175858. [PMID: 37356787 DOI: 10.1016/j.ejphar.2023.175858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/27/2023]
Abstract
Bone cancer pain (BCP) is one of the most prevalent and serious symptoms of patients with cancer. Currently, the medical interventions used for the treatment of BCP do not act with optimal safety and efficacy. In this study, we appraised whether the hypoxia-inducible factor 1α (HIF-1α)/metalloproteinase-9 (MMP9) axis activates the PI3K/AKT pathway, resulting in elevated spinal cord central sensitization and aggravated BCP. BCP rats were established by tibial injection of Walker 256 cells, followed by different interventions in rats using HIF-1ɑ inhibitor LW6 or antibody treatments. After treatment with LW6 or antibody against HIF-1α, central sensitization in the spinal cord tissues of rats was inhibited, and pain perception in rats was reduced. Moreover, the activation of glial cells in the spinal cord tissues was ameliorated. The expression of MMP9 was remarkably suppressed in spinal cord tissues after inhibition of HIF-1ɑ activity, and the activity of the PI3K/AKT signaling pathway was inhibited. Further activation of MMP9 expression suppressed the alleviating effect of HIF-1ɑ inhibitor LW6 or antibody on pain perception in rats inoculated with tumors. Taken together, our studies suggest a HIF-1α/MMP9-mediated activation of PI3K/AKT in the spinal cord tissues, resulting in increased pain perception in a rat model with BCP.
Collapse
Affiliation(s)
- Liyu Yang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110003, Liaoning, PR China
| | - Qin Fu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110003, Liaoning, PR China
| | - Liqing Yang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110003, Liaoning, PR China
| | - Yiqi Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110003, Liaoning, PR China.
| |
Collapse
|
5
|
Yang J, Pei T, Su G, Duan P, Liu X. AnnexinA6: a potential therapeutic target gene for extracellular matrix mineralization. Front Cell Dev Biol 2023; 11:1201200. [PMID: 37727505 PMCID: PMC10506415 DOI: 10.3389/fcell.2023.1201200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/10/2023] [Indexed: 09/21/2023] Open
Abstract
The mineralization of the extracellular matrix (ECM) is an essential and crucial process for physiological bone formation and pathological calcification. The abnormal function of ECM mineralization contributes to the worldwide risk of developing mineralization-related diseases; for instance, vascular calcification is attributed to the hyperfunction of ECM mineralization, while osteoporosis is due to hypofunction. AnnexinA6 (AnxA6), a Ca2+-dependent phospholipid-binding protein, has been extensively reported as an essential target in mineralization-related diseases such as osteoporosis, osteoarthritis, atherosclerosis, osteosarcoma, and calcific aortic valve disease. To date, AnxA6, as the largest member of the Annexin family, has attracted much attention due to its significant contribution to matrix vesicles (MVs) production and release, MVs-ECM interaction, cytoplasmic Ca2+ influx, and maturation of hydroxyapatite, making it an essential target in ECM mineralization. In this review, we outlined the recent advancements in the role of AnxA6 in mineralization-related diseases and the potential mechanisms of AnxA6 under normal and mineralization-related pathological conditions. AnxA6 could promote ECM mineralization for bone regeneration in the manner described previously. Therefore, AnxA6 may be a potential osteogenic target for ECM mineralization.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Liu X, Zhao S, Zhao Q, Chen Y, Jia S, Xiang R, Zhang J, Sun J, Xu Y, Zhao M. Butein, a potential drug for the treatment of bone cancer pain through bioinformatic and network pharmacology. Toxicol Appl Pharmacol 2023; 472:116570. [PMID: 37268026 DOI: 10.1016/j.taap.2023.116570] [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: 11/04/2022] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/04/2023]
Abstract
Bone cancer pain is a difficult-to-treat pathologic condition that impairs the patient's quality of life. The effective therapy options for BCP are restricted due to the unknown pathophysiology. Transcriptome data were obtained from the Gene Expression Omnibus database and differentially expressed gene extraction was performed. DEGs integrated with pathological targets found 68 genes in the study. Butein was discovered as a possible medication for BCP after the 68 genes were submitted to the Connectivity Map 2.0 database for drug prediction. Moreover, butein has good drug-likeness properties. To collect the butein targets, we used the CTD, SEA, TargetNet, and Super-PRED databases. Furthermore, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses revealed butein's pharmacological effects, indicating that butein may aid in treating BCP by altering the hypoxia-inducible factor, NF-kappa B, angiogenesis, and sphingolipid signaling pathways. Moreover, the pathological targets integrated with drug targets were obtained as the shared gene set A, which was analyzed by ClueGO and MCODE. Biological process analysis and MCODE algorithm further analyzed that BCP related targets were mainly involved in signal transduction process and ion channel-related pathways. Next, we integrated targets related to network topology parameters and targets of core pathways, identified PTGS2, EGFR, JUN, ESR1, TRPV1, AKT1 and VEGFA as butein regulated hub genes by molecular docking, which play a critical role in its analgesic effect. This study lays the scientific groundwork for elucidating the mechanism underlying butein's success in the treatment of BCP.
Collapse
Affiliation(s)
- Xin Liu
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Shangfeng Zhao
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Qianqian Zhao
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yiwei Chen
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Shubing Jia
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Rongwu Xiang
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jinghai Zhang
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jianfang Sun
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yijia Xu
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Mingyi Zhao
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
7
|
Ding Z, Liang X, Wang J, Song Z, Guo Q, Schäfer MKE, Huang C. Inhibition of spinal ferroptosis-like cell death alleviates hyperalgesia and spontaneous pain in a mouse model of bone cancer pain. Redox Biol 2023; 62:102700. [PMID: 37084690 PMCID: PMC10141498 DOI: 10.1016/j.redox.2023.102700] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/29/2023] [Accepted: 04/11/2023] [Indexed: 04/23/2023] Open
Abstract
Bone cancer pain (BCP) impairs patients' quality of life. However, the underlying mechanisms are still unclear. This study investigated the role of spinal interneuron death using a pharmacological inhibitor of ferroptosis in a mouse model of BCP. Lewis lung carcinoma cells were inoculated into the femur, resulting in hyperalgesia and spontaneous pain. Biochemical analysis revealed that spinal levels of reactive oxygen species and malondialdehyde were increased, while those of superoxide dismutase were decreased. Histological analysis showed the loss of spinal GAD65+ interneurons and provided ultrastructural evidence of mitochondrial shrinkage. Pharmacologic inhibition of ferroptosis using ferrostatin-1 (FER-1, 10 mg/kg, intraperitoneal for 20 consecutive days) attenuated ferroptosis-associated iron accumulation and lipid peroxidation and alleviated BCP. Furthermore, FER-1 inhibited the pain-associated activation of ERK1/2 and COX-2 expression and prevented the loss of GABAergic interneurons. Moreover, FER-1 improved analgesia by the COX-2 inhibitor Parecoxib. Taken together, this study shows that pharmacological inhibition of ferroptosis-like cell death of spinal interneurons alleviates BCP in mice. The results suggest that ferroptosis is a potential therapeutic target in patients suffering on BCP and possibly other types of pain.
Collapse
Affiliation(s)
- Zhuofeng Ding
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Xiaoshen Liang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Jian Wang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Zongbin Song
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Germany; Focus Program Translational Neurosciences (FTN) and Research Center of Immunotherapy of the Johannes Gutenberg-University Mainz, Germany
| | - Changsheng Huang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, 410008, China.
| |
Collapse
|
8
|
Wu J, Shi Y, Xing M, Deng M, Cao W, Guo Q, Zou W. CircRalgapa1 facilitates morphine tolerance via miR-873a-5p/A20 axis in mice. Neuropharmacology 2023; 224:109353. [PMID: 36455645 DOI: 10.1016/j.neuropharm.2022.109353] [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: 10/04/2022] [Revised: 11/08/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022]
Abstract
Morphine tolerance (MT) caused by long-term use of morphine is a major medical problem. The underlying molecular mechanisms of morphine tolerance remain unclear. Here, we establish the morphine tolerance model in mice and verify whether a novel circRNA, circRalgapa1 is involved in morphine tolerance and its specific molecular mechanism. We show that the expression of circRalgapa1 in the spinal cord is significantly down-expressed in the spinal cord of morphine-tolerant mice. CircRalgapa1 is mainly located in the neuronal cytoplasm and co-localizes with miR-873a-5p. Mechanically, circRalgapa1 acts as competing endogenous RNAs (ceRNAs) to regulate the inhibitory of miR-873a-5p on A20 (also known as tumor necrosis factor α-induced protein 3, TNFAIP3). Functionally, overexpression of circRalgapa1 by intrathecal injection of adeno-associated virus (AAV- circRalgapa1) attenuated the formation of morphine tolerance and partially reversed the development of morphine tolerance. Moreover, overexpression of miR-873a-5p blocked the effect of AAV-circRalgapa1 on alleviating morphine tolerance in mice. In conclusion, chronic morphine administration-mediated down-regulation of circRalgapa1 in the spinal cord contributes to morphine tolerance via miR-873a-5p/A20 axis in mice. Overexpression of circRalgapa1 may be a promising RNA-based therapy for morphine tolerance.
Collapse
Affiliation(s)
- Jing Wu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yufei Shi
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Manyu Xing
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Meiling Deng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Cao
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
9
|
Bui BP, Nguyen PL, Lee K, Cho J. Hypoxia-Inducible Factor-1: A Novel Therapeutic Target for the Management of Cancer, Drug Resistance, and Cancer-Related Pain. Cancers (Basel) 2022; 14:cancers14246054. [PMID: 36551540 PMCID: PMC9775408 DOI: 10.3390/cancers14246054] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a key transcription factor that regulates the transcription of many genes that are responsible for the adaptation and survival of tumor cells in hypoxic environments. Over the past few decades, tremendous efforts have been made to comprehensively understand the role of HIF-1 in tumor progression. Based on the pivotal roles of HIF-1 in tumor biology, many HIF-1 inhibitors interrupting expression, stabilization, DNA binding properties, or transcriptional activity have been identified as potential therapeutic agents for various cancers, yet none of these inhibitors have yet been successfully translated into clinically available cancer treatments. In this review, we briefly introduce the regulation of the HIF-1 pathway and summarize its roles in tumor cell proliferation, angiogenesis, and metastasis. In addition, we explore the implications of HIF-1 in the development of drug resistance and cancer-related pain: the most commonly encountered obstacles during conventional anticancer therapies. Finally, the current status of HIF-1 inhibitors in clinical trials and their perspectives are highlighted, along with their modes of action. This review provides new insights into novel anticancer drug development targeting HIF-1. HIF-1 inhibitors may be promising combinational therapeutic interventions to improve the efficacy of current cancer treatments and reduce drug resistance and cancer-related pain.
Collapse
|
10
|
Peng Z, Yang F, Huang S, Tang Y, Wan L. Targeting Vascular endothelial growth factor A with soluble vascular endothelial growth factor receptor 1 ameliorates nerve injury-induced neuropathic pain. Mol Pain 2022; 18:17448069221094528. [PMID: 35354377 PMCID: PMC9706061 DOI: 10.1177/17448069221094528] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Neuropathic pain is a distressing medical condition with few effective treatments. The role of Vascular endothelial growth factor A (VEGFA) in inflammation pain has been confirmed in many researches. However, the mechanism of VEGFA affects neuropathic pain remains unclear. In this study, we demonstrated that VEGFA plays an important role in spare nerve injury (SNI)-induced neuropathic pain, which is mediated by enhanced expression and colocalized of VEGFA, p-AKT and TRPV1 in SNI-induced neuropathic pain model. Soluble VEGFR1 (sFlt1) not only relieved mechanical hyperalgesia and the expression of inflammatory markers, but ameliorated the expression of VEGFA, VEGFR2, p-AKT, and TRPV1 in spinal cord. However, these effects of sFlt1 can be blocked by rpVEGFA and by 740 Y-P. Therefore, our study indication that targeting VEGFA with sFlt1 reduces neuropathic pain development via the AKT/TRPV1 pathway in SNI-induced nerve injury. This study elucidates a new therapeutic target for neuropathic pain.
Collapse
Affiliation(s)
- Zhe Peng
- Department of Pain Medicine, The
State Key Clinical Specialty in Pain Medicine, The Second Affiliated Hospital,
Guangzhou
Medical University, Guangzhou, P.R.
China,Stem Cell Translational Medicine
Center, The Second Affiliated Hospital, Guangzhou Medical
University, Guangzhou, P. R. of China
| | - Fan Yang
- Department of Pain Medicine, The
State Key Clinical Specialty in Pain Medicine, The Second Affiliated Hospital,
Guangzhou
Medical University, Guangzhou, P.R.
China,Stem Cell Translational Medicine
Center, The Second Affiliated Hospital, Guangzhou Medical
University, Guangzhou, P. R. of China
| | - Siting Huang
- Department of Pain Medicine, The
State Key Clinical Specialty in Pain Medicine, The Second Affiliated Hospital,
Guangzhou
Medical University, Guangzhou, P.R.
China
| | - Yang Tang
- Department of Pain Medicine, The
State Key Clinical Specialty in Pain Medicine, The Second Affiliated Hospital,
Guangzhou
Medical University, Guangzhou, P.R.
China,Stem Cell Translational Medicine
Center, The Second Affiliated Hospital, Guangzhou Medical
University, Guangzhou, P. R. of China
| | - Li Wan
- Department of Pain Medicine, The
State Key Clinical Specialty in Pain Medicine, The Second Affiliated Hospital,
Guangzhou
Medical University, Guangzhou, P.R.
China,Stem Cell Translational Medicine
Center, The Second Affiliated Hospital, Guangzhou Medical
University, Guangzhou, P. R. of China,Li Wan, Department of Pain management, The
Second Affiliated Hospital, Guangzhou Medical University, 250 Changgang Dong Lu,
Guangzhou 510260, P.R. China.
| |
Collapse
|
11
|
Peck AB, Ambrus JL. A Temporal Comparative RNA Transcriptome Profile of the Annexin Gene Family in the Salivary versus Lacrimal Glands of the Sjögren's Syndrome-Susceptible C57BL/6.NOD- Aec1Aec2 Mouse. Int J Mol Sci 2022; 23:11709. [PMID: 36233010 PMCID: PMC9570365 DOI: 10.3390/ijms231911709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2022] Open
Abstract
A generally accepted hypothesis for the initial activation of an immune or autoimmune response argues that alarmins are released from injured, dying and/or activated immune cells, and these products complex with receptors that activate signal transduction pathways and recruit immune cells to the site of injury where the recruited cells are stimulated to initiate immune and/or cellular repair responses. While there are multiple diverse families of alarmins such as interleukins (IL), heat-shock proteins (HSP), Toll-like receptors (TLR), plus individual molecular entities such as Galectin-3, Calreticulin, Thymosin, alpha-Defensin-1, RAGE, and Interferon-1, one phylogenetically conserved family are the Annexin proteins known to promote an extensive range of biomolecular and cellular products that can directly and indirectly regulate inflammation and immune activities. For the present report, we examined the temporal expression profiles of the 12 mammalian annexin genes (Anxa1-11 and Anxa13), applying our temporal genome-wide transcriptome analyses of ex vivo salivary and lacrimal glands from our C57BL/6.NOD-Aec1Aec2 mouse model of Sjögren's Syndrome (SS), a human autoimmune disease characterized primarily by severe dry mouth and dry eye symptoms. Results indicate that annexin genes Anax1-7 and -11 exhibited upregulated expressions and the initial timing for these upregulations occurred as early as 8 weeks of age and prior to any covert signs of a SS-like disease. While the profiles of the two glands were similar, they were not identical, suggesting the possibility that the SS-like disease may not be uniform in the two glands. Nevertheless, this early pre-clinical and concomitant upregulated expression of this specific set of alarmins within the immune-targeted organs represents a potential target for identifying the pre-clinical stage in human SS as well, a fact that would clearly impact future interventions and therapeutic strategies.
Collapse
Affiliation(s)
- Ammon B Peck
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, P.O. Box 100125, Gainesville, FL 32610, USA
| | - Julian L Ambrus
- Division of Allergy, Immunology and Rheumatology, SUNY Buffalo School of Medicine, 875 Ellicott Street, Buffalo, NY 14203, USA
| |
Collapse
|
12
|
Sodium aescinate alleviates bone cancer pain in rats by suppressing microglial activation via p38 MAPK/c-Fos signaling. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00234-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Iafrate L, Benedetti MC, Donsante S, Rosa A, Corsi A, Oreffo ROC, Riminucci M, Ruocco G, Scognamiglio C, Cidonio G. Modelling skeletal pain harnessing tissue engineering. IN VITRO MODELS 2022; 1:289-307. [PMID: 36567849 PMCID: PMC9766883 DOI: 10.1007/s44164-022-00028-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/27/2022]
Abstract
Bone pain typically occurs immediately following skeletal damage with mechanical distortion or rupture of nociceptive fibres. The pain mechanism is also associated with chronic pain conditions where the healing process is impaired. Any load impacting on the area of the fractured bone will stimulate the nociceptive response, necessitating rapid clinical intervention to relieve pain associated with the bone damage and appropriate mitigation of any processes involved with the loss of bone mass, muscle, and mobility and to prevent death. The following review has examined the mechanisms of pain associated with trauma or cancer-related skeletal damage focusing on new approaches for the development of innovative therapeutic interventions. In particular, the review highlights tissue engineering approaches that offer considerable promise in the application of functional biomimetic fabrication of bone and nerve tissues. The strategic combination of bone and nerve tissue engineered models provides significant potential to develop a new class of in vitro platforms, capable of replacing in vivo models and testing the safety and efficacy of novel drug treatments aimed at the resolution of bone-associated pain. To date, the field of bone pain research has centred on animal models, with a paucity of data correlating to the human physiological response. This review explores the evident gap in pain drug development research and suggests a step change in approach to harness tissue engineering technologies to recapitulate the complex pathophysiological environment of the damaged bone tissue enabling evaluation of the associated pain-mimicking mechanism with significant therapeutic potential therein for improved patient quality of life. Graphical abstract Rationale underlying novel drug testing platform development. Pain detected by the central nervous system and following bone fracture cannot be treated or exclusively alleviated using standardised methods. The pain mechanism and specificity/efficacy of pain reduction drugs remain poorly understood. In vivo and ex vivo models are not yet able to recapitulate the various pain events associated with skeletal damage. In vitro models are currently limited by their inability to fully mimic the complex physiological mechanisms at play between nervous and skeletal tissue and any disruption in pathological states. Robust innovative tissue engineering models are needed to better understand pain events and to investigate therapeutic regimes.
Collapse
Affiliation(s)
- Lucia Iafrate
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
| | - Maria Cristina Benedetti
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandro Rosa
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Richard O. C. Oreffo
- Bone and Joint Research Group, Stem Cells and Regeneration, Institute of Developmental Sciences, Centre for Human Development, University of Southampton, Southampton, UK
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
| | - Chiara Scognamiglio
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
| | - Gianluca Cidonio
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
- Bone and Joint Research Group, Stem Cells and Regeneration, Institute of Developmental Sciences, Centre for Human Development, University of Southampton, Southampton, UK
| |
Collapse
|
14
|
Mechanisms of bone pain: Progress in research from bench to bedside. Bone Res 2022; 10:44. [PMID: 35668080 PMCID: PMC9170780 DOI: 10.1038/s41413-022-00217-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 12/27/2022] Open
Abstract
AbstractThe field of research on pain originating from various bone diseases is expanding rapidly, with new mechanisms and targets asserting both peripheral and central sites of action. The scope of research is broadening from bone biology to neuroscience, neuroendocrinology, and immunology. In particular, the roles of primary sensory neurons and non-neuronal cells in the peripheral tissues as important targets for bone pain treatment are under extensive investigation in both pre-clinical and clinical settings. An understanding of the peripheral mechanisms underlying pain conditions associated with various bone diseases will aid in the appropriate application and development of optimal strategies for not only managing bone pain symptoms but also improving bone repairing and remodeling, which potentially cures the underlying etiology for long-term functional recovery. In this review, we focus on advances in important preclinical studies of significant bone pain conditions in the past 5 years that indicated new peripheral neuronal and non-neuronal mechanisms, novel targets for potential clinical interventions, and future directions of research.
Collapse
|
15
|
Deng M, Zhang Z, Xing M, Liang X, Li Z, Wu J, Jiang S, Weng Y, Guo Q, Zou W. LncRNA MRAK159688 facilitates morphine tolerance by promoting REST-mediated inhibition of mu opioid receptor in rats. Neuropharmacology 2022; 206:108938. [PMID: 34982972 DOI: 10.1016/j.neuropharm.2021.108938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 12/11/2022]
Abstract
Morphine tolerance (MT) caused by the long-term use of morphine is a major medical problem. The molecular mechanism of morphine tolerance remains elusive. Here, we established a morphine tolerance model in rats and verified whether the long noncoding RNA (lncRNA) MRAK159688 is involved in morphine tolerance and its specific molecular mechanism. We show the significant upregulation of MRAK159688 expression in the spinal cord of morphine-tolerant rats. Overexpression of MRAK159688 by a lentivirus reduces the analgesic efficacy of morphine and induces pain behavior. Downregulation of MRAK159688 using a small interfering RNA (siRNA) attenuates the formation of morphine tolerance, partially reverses the development of morphine tolerance and alleviates morphine-induced hyperalgesia. MRAK159688 is located in the nucleus and cytoplasm of neurons, and it colocalizes with repressor element-1 silencing transcription factor (REST) in the nucleus. MRAK159688 potentiates the expression and function of REST, thereby inhibiting the expression of mu opioid receptor (MOR) and subsequently inducing morphine tolerance. Moreover, REST overexpression blocks the effects of MRAK159688 siRNA on relieving morphine tolerance. In general, chronic morphine administration-mediated upregulation of MRAK159688 in the spinal cord contributes to morphine tolerance and hyperalgesia by promoting REST-mediated inhibition of MOR. MRAK159688 downregulation may represent a novel RNA-based therapy for morphine tolerance.
Collapse
Affiliation(s)
- Meiling Deng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zengli Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Department of Anesthesiology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, 300000, China
| | - Manyu Xing
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xia Liang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Zhengyiqi Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jing Wu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shasha Jiang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yingqi Weng
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
16
|
Ai-Tong-An-Gao-Ji and Fisetin Inhibit Tumor Cell Growth in Rat CIBP Models by Inhibiting the AKT/HIF-1 α Signaling Pathway. JOURNAL OF ONCOLOGY 2022; 2022:1459636. [PMID: 35222641 PMCID: PMC8866002 DOI: 10.1155/2022/1459636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/29/2021] [Accepted: 01/12/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Ai-Tong-An-Gao-Ji (ATAGJ) has been extensively applied for acute bone cancer pain treatment with a satisfactory efficacy, while the specific mechanisms remain unclear and require further investigation. METHODS Overlapped genes of ATAGJ and CIBP obtained from SwissTargetPrediction website and GeneCards database were presented as a Venn diagram. A network diagram of drug-component-target was further established using the Cytoscape 3.6.0 software. The effect of fisetin on Walker 256 cell proliferation was observed by clone formation assay and EDU assay, and the interaction between fisetin and AKT was revealed using the immunoprecipitation assay. Effects of fisetin on AKT/HIF-1α signaling pathway in Walker 256 cells were ultimately detected using Western blot and qPCR assays. RESULTS The key component fisetin and core target gene AKT were sorted out using the drug-component-target network with a binding energy between fisetin and AKT less than -5 kcal/mol. Clone formation assay and EDU assay showed that fisetin substantially suppressed the proliferation of Walker 256 cells. Immunoprecipitation assay results revealed that the combination of fisetin and AKT decreased the level of AKT/HIF-1α signaling pathway of Walker 256 cells. CONCLUSIONS The fisetin of ATAGJ can markedly suppress Walker 256 cells, and the mechanisms may be intimately associated with the combination of fisetin and AKT. Furthermore, fisetin decreased the level of p-AKT and inhibited the expression of the AKT/HIF-1α signaling pathway.
Collapse
|
17
|
Zhou YS, Cui Y, Zheng JX, Quan YQ, Wu SX, Xu H, Han Y. Luteolin relieves lung cancer-induced bone pain by inhibiting NLRP3 inflammasomes and glial activation in the spinal dorsal horn in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 96:153910. [PMID: 35026502 DOI: 10.1016/j.phymed.2021.153910] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Bone cancer pain (BCP) is one of the most severe complications in cancer patients. However, the pharmacological therapeutic approaches are limited. Luteolin, a major component of flavones, is widely distributed in plants and plays a critical role in the antinociceptive effects, but whether luteolin could alleviate cancer pain and its underlying mechanisms are not known. HYPOTHESIS/PURPOSE This study investigated the molecular mechanisms by which luteolin reduced BCP. METHODS Behavioral, pharmacological, immunohistochemical, and biochemical approaches were used to investigate the effect of luteolin on BCP. RESULTS Luteolin treatment ameliorated Lewis lung cancer (LLC)-induced bone pain in mice in a dose-dependent manner. Luteolin treatment could inhibit the activation of neurons, glial cells, and NOD-like receptor protein 3 (NLRP3) inflammasomes in the dorsal spinal cord in the BCP mouse model. Furthermore, phosphorylated p-38 mitogen-activated protein kinase (MAPK) in the spinal dorsal horn (SDH) was suppressed by luteolin treatment that could influence the analgesic and glial inhibition effects of luteolin. CONCLUSION Our results demonstrated that luteolin inhibited neuroinflammation by obstructing glial cell and NLRP3 inflammasome activation via modulating p38 MAPK activity in SDH, ultimately improving LLC-induced BCP.
Collapse
Affiliation(s)
- Yong-Sheng Zhou
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China; Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yue Cui
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China; College of Life Sciences and Research Center for Resource Peptide Drugs, Shaanxi Engineering and Technological Research Center for Conversation and Utilization of Regional Biological Resources, Yanan University, Yanan, 716099, China
| | - Jia-Xin Zheng
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ya-Qi Quan
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Sheng-Xi Wu
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Xu
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yong Han
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China; Department of Thoracic Surgery, Air Force Medical Center, PLA, Beijing, 100142, China.
| |
Collapse
|
18
|
Fu Q, Huang X, Li W, Wan S, Li Y, Li X, Su S, Xu X, Wu Y. P-Rex2 mediation of synaptic plasticity contributes to bone cancer pain. Mol Pain 2022; 18:17448069221076460. [PMID: 35083941 PMCID: PMC8891909 DOI: 10.1177/17448069221076460] [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] [Indexed: 11/17/2022] Open
Abstract
Bone cancer pain (BCP) seriously affects the quality of life; however, due to its complex
mechanism, the clinical treatment was unsatisfactory. Recent studies have showed several
Rac-specific guanine nucleotide exchange factors (GEFs) that affect development and
structure of neuronal processes play a vital role in the regulation of chronic pain.
P-Rex2 is one of GEFs that regulate spine density, and the present study was performed to
examine the effect of P-Rex2 on the development of BCP. Tumor cells implantation induced
the mechanical hyperalgesia, which was accompanied by an increase in spinal protein
P-Rex2, phosphorylated Rac1 (p-Rac1) and phosphorylated GluR1 (p-GluR1), and number of
spines. Intrathecal injection a P-Rex2-targeting RNAi lentivirus relieved BCP and reduced
the expression of P-Rex2, p-Rac1, p-GluR1, and number of spines in the BCP mice.
Meanwhile, P-Rex2 knockdown reversed BCP-enhanced AMPA receptor (AMPAR)-induced current in
dorsal horn neurons. In summary, this study suggested that P-Rex2 regulated
GluR1-containing AMPAR trafficking and spine morphology via Rac1/pGluR1 pathway is a
fundamental pathogenesis of BCP. Our findings provide a better understanding of the
function of P-Rex2 as a possible therapeutic target for relieving BCP.
Collapse
Affiliation(s)
- Qiaochu Fu
- Anesthesiology105738Beijing Tiantan Hospital
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Fan LJ, Kan HM, Chen XT, Sun YY, Chen LP, Shen W. Vascular endothelial growth factor-A/vascular endothelial growth factor2 signaling in spinal neurons contributes to bone cancer pain. Mol Pain 2022; 18:17448069221075891. [PMID: 35083936 PMCID: PMC8874205 DOI: 10.1177/17448069221075891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Tumor metastasis to bone is often accompanied by a severe pain syndrome (cancer-induced bone pain, CIBP) that is frequently unresponsive to analgesics, which markedly reduces patient quality of life and cancer treatment tolerance in patients. Prolonged pain can induce hypersensitivity via spinal plasticity, and several recent studies have implicated the involvement of vascular endothelial growth factor-A (VEGF-A) signaling in this process. Here, we speculated that CIBP is associated with VEGF-A/VEGFR2 signaling in the spinal cord. A mouse model of CIBP was established by intramedullary injection of Lewis lung carcinoma (LLC) cells in the mouse femur. Pain sensitization and potential amelioration via VEGF-A/VEGFR2 blockade were measured using paw withdrawal threshold to mechanical stimulation and paw withdrawal latency to thermal. Spinal VEGF-A/VEGFR2 signaling was blocked by intrathecal injection of the VEGF-A antibody or the specific VEGFR2 inhibitor ZM323881. Changes in the expression levels of VEGF-A, VEGFR2, and other pain-related signaling factors were measured using western blotting and immunofluorescence staining. Mice after LLC injection demonstrated mechanical allodynia and thermal hyperalgesia, both of which were suppressed via anti-VEGF-A antibody or ZM323881. Conversely, the intrathecal injection of exogenous VEGF-A was sufficient to cause pain hypersensitivity in naïve mice via the VEGFR2-mediated activation of protein kinase C. Moreover, the spinal blockade of VEGF-A or VEGFR2 also suppressed N-methyl-D-aspartate receptor (NMDAR) activation and downstream Ca2+-dependent signaling. Thus, spinal VEGF-A/VEGFR2/NMDAR signaling pathways may be critical mediators of CIBP.
Collapse
Affiliation(s)
- Li-Jun Fan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Hou-Ming Kan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Xue-Tai Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Yuan-Yuan Sun
- Department of Anesthesiology, Xuzhou First People’s Hospital, Xuzhou, China
| | - Li-ping Chen
- Department of Pain Management, Affiliated Hospital of Xuzhou, Xuzhou, China
| | - Wen Shen
- Department of Pain Management, Affiliated Hospital of Xuzhou, Xuzhou, China
| |
Collapse
|
20
|
Gadepalli A, Akhilesh, Uniyal A, Modi A, Chouhan D, Ummadisetty O, Khanna S, Solanki S, Allani M, Tiwari V. Multifarious Targets and Recent Developments in the Therapeutics for the Management of Bone Cancer Pain. ACS Chem Neurosci 2021; 12:4195-4208. [PMID: 34723483 DOI: 10.1021/acschemneuro.1c00414] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Bone cancer pain (BCP) is a distinct pain state showing characteristics of both neuropathic and inflammatory pain. On average, almost 46% of cancer patients exhibit BCP with numbers flaring up to as high as 76% for terminally ill patients. Patients suffering from BCP experience a compromised quality of life, and the unavailability of effective therapeutics makes this a more devastating condition. In every individual cancer patient, the pain is driven by different mechanisms at different sites. The mechanisms behind the manifestation of BCP are very complex and poorly understood, which creates a substantial barrier to drug development. Nevertheless, some of the key mechanisms involved have been identified and are being explored further to develop targeted molecules. Developing a multitarget approach might be beneficial in this case as the underlying mechanism is not fixed and usually a number of these pathways are simultaneously dysregulated. In this review, we have discussed the role of recently identified novel modulators and mechanisms involved in the development of BCP. They include ion channels and receptors involved in sensing alteration of temperature and acidic microenvironment, immune system activation, sodium channels, endothelins, protease-activated receptors, neurotrophins, motor proteins mediated trafficking of glutamate receptor, and some bone-specific mechanisms. Apart from this, we have also discussed some of the novel approaches under preclinical and clinical development for the treatment of bone cancer pain.
Collapse
Affiliation(s)
- Anagha Gadepalli
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Ankit Uniyal
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Ajay Modi
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Obulapathi Ummadisetty
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Shreya Khanna
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Shreya Solanki
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Meghana Allani
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh India
| |
Collapse
|
21
|
Xiao B, Wang G, Huo H, Li W. Identification of HIF-1α/VEGFA signaling pathway and transcription factors in Kashin-Beck disease by integrated bioinformatics analysis. Exp Ther Med 2021; 22:1115. [PMID: 34504569 PMCID: PMC8383754 DOI: 10.3892/etm.2021.10549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/16/2021] [Indexed: 12/29/2022] Open
Abstract
Kashin-Beck disease (KBD) is a chronic and endemic osteoarthropathy. The pathogenesis of KBD has yet to be fully elucidated, although previous studies have shown that its etiology may be associated with low selenium abundance and high exposure to mycotoxins, such as T-2 toxin. In the present study, the Comparative Toxicogenomics Database was used to identify key genes associated with KBD, T-2 toxin and selenium. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were used to identify the biological processes and pathways that key genes may be associated with. By searching the Search Tool for the Retrieval of Interacting Genes database and the Molecular Complex Detection plug-in with Cytoscape, it was possible to construct a KBD-associated protein-protein interaction (PPI) network, and screen the core modules and genes. Western blot analysis was subsequently used to verify the expression levels of hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor A (VEGFA), two components that are associated with the HIF-1 signaling pathway in KBD disease. Via this approach, a total of 301 key genes were identified that were associated with KBD, T-2 toxin and selenium. The results of the GO and KEGG enrichment analyses demonstrated that these key genes were mainly involved in the process of apoptosis. Previous studies have demonstrated that excessive apoptosis of chondrocytes plays a crucial role in the pathophysiology of KBD, and that HIF-1α has an important role in chondrocyte apoptosis; therefore, the present study was focused on the expression level of HIF-1α in KBD. By analyzing the PPI network constructed from the key genes, a total of 10 core genes were obtained that may be associated with KBD. The results of western blotting experiments revealed that, after treating chondrocytes with different concentrations of T-2 toxin, the expression levels of HIF-1α and VEGFA were markedly downregulated. The iRegulon plug-in for Cytoscape was used to predict the transcription factors that may regulate HIF-1α and VEGFA in the HIF-1 signaling pathway. Using this approach, 10 core genes and 15 transcription factors were obtained. These results may help to clarify the pathogenesis of KBD, thereby providing further avenues for the therapeutic treatment of KBD.
Collapse
Affiliation(s)
- Bin Xiao
- Department of Orthopedics, Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Guozhu Wang
- Department of Orthopedics, Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Hongliang Huo
- Department of Orthopedics, Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Weiwei Li
- Department of Orthopedics, Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| |
Collapse
|
22
|
Yang L, Lu P, Yang X, Li K, Qu S. Annexin A3, a Calcium-Dependent Phospholipid-Binding Protein: Implication in Cancer. Front Mol Biosci 2021; 8:716415. [PMID: 34355022 PMCID: PMC8329414 DOI: 10.3389/fmolb.2021.716415] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/08/2021] [Indexed: 12/20/2022] Open
Abstract
Annexin A3 (ANXA3), also known as lipocortin III and placental anticoagulant protein III, has been reported to be dysregulated in tumor tissues and cancer cell lines, and harbors pronounced diagnostic and prognostic value for certain malignancies, such as breast, prostate, colorectal, lung and liver cancer. Aberrant expression of ANXA3 promotes tumor cell proliferation, invasion, metastasis, angiogenesis, and therapy resistance to multiple chemotherapeutic drugs including platinum-based agents, fluoropyrimidines, cyclophosphamide, doxorubicin, and docetaxel. Genetic alterations on the ANXA3 gene have also been reported to be associated with the propensity to form certain inherited, familial tumors. These diverse functions of ANXA3 in tumors collectively indicate that ANXA3 may serve as an attractive target for novel anticancer therapies and a powerful diagnostic and prognostic biomarker for early tumor detection and population risk screening. In this review, we dissect the role of ANXA3 in cancer in detail.
Collapse
Affiliation(s)
- Liu Yang
- Key Laboratory of High-Incidence Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Pingan Lu
- Faculty of Medicine, Amsterdam Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Xiaohui Yang
- Key Laboratory of High-Incidence Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Kaiguo Li
- Key Laboratory of High-Incidence Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Song Qu
- Key Laboratory of High-Incidence Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
23
|
Annexin A3 as a Marker Protein for Microglia in the Central Nervous System of Rats. Neural Plast 2021; 2021:5575090. [PMID: 34221002 PMCID: PMC8211522 DOI: 10.1155/2021/5575090] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/21/2021] [Accepted: 06/01/2021] [Indexed: 01/08/2023] Open
Abstract
The parenchymal microglia possess different morphological characteristics in cerebral physiological and pathological conditions; thus, visualizing these cells is useful as a means of further investigating parenchymal microglial function. Annexin A3 (ANXA3) is expressed in microglia, but it is unknown whether it can be used as a marker protein for microglia and its physiological function. Here, we compared the distribution and morphology of parenchymal microglia labeled by ANXA3, cluster of differentiation 11b (CD11b), and ionized calcium-binding adaptor molecule 1 (Iba1) and measured the expression of ANXA3 in nonparenchymal macrophages (meningeal and perivascular macrophages). We also investigated the spatiotemporal expression of ANXA3, CD11b, and Iba1 in vivo and in vitro and the cellular function of ANXA3 in microglia. We demonstrated that ANXA3-positive cells were abundant and evenly distributed throughout the whole brain tissue and spinal cord of adult rats. The morphology and distribution of ANXA3-labeled microglia were quite similar to those labeled by the microglial-specific markers CD11b and Iba1 in the central nervous system (CNS). ANXA3 was expressed in the cytoplasm of microglia, and its expression was significantly increased in activated microglia. ANXA3 was almost undetectable in the nonparenchymal macrophages. Meanwhile, the protein and mRNA expression levels of ANXA3 in different regions of the CNS were different from those of CD11b and Iba1. Moreover, knockdown of ANXA3 inhibited the proliferation and migration of microglia, while overexpression of ANXA3 enhanced these activities. This study confirms that ANXA3 may be a novel marker for parenchymal microglia in the CNS of adult rats and enriches our understanding of ANXA3 from expression patterns to physiological function.
Collapse
|
24
|
Grewal T, Rentero C, Enrich C, Wahba M, Raabe CA, Rescher U. Annexin Animal Models-From Fundamental Principles to Translational Research. Int J Mol Sci 2021; 22:ijms22073439. [PMID: 33810523 PMCID: PMC8037771 DOI: 10.3390/ijms22073439] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Routine manipulation of the mouse genome has become a landmark in biomedical research. Traits that are only associated with advanced developmental stages can now be investigated within a living organism, and the in vivo analysis of corresponding phenotypes and functions advances the translation into the clinical setting. The annexins, a family of closely related calcium (Ca2+)- and lipid-binding proteins, are found at various intra- and extracellular locations, and interact with a broad range of membrane lipids and proteins. Their impacts on cellular functions has been extensively assessed in vitro, yet annexin-deficient mouse models generally develop normally and do not display obvious phenotypes. Only in recent years, studies examining genetically modified annexin mouse models which were exposed to stress conditions mimicking human disease often revealed striking phenotypes. This review is the first comprehensive overview of annexin-related research using animal models and their exciting future use for relevant issues in biology and experimental medicine.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Mohamed Wahba
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Carsten A. Raabe
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
| | - Ursula Rescher
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| |
Collapse
|
25
|
An Investigation of the Molecular Mechanisms Underlying the Analgesic Effect of Jakyak-Gamcho Decoction: A Network Pharmacology Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:6628641. [PMID: 33343676 PMCID: PMC7732394 DOI: 10.1155/2020/6628641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/05/2020] [Accepted: 11/24/2020] [Indexed: 12/20/2022]
Abstract
Herbal drugs have drawn substantial interest as effective analgesic agents; however, their therapeutic mechanisms remain to be fully understood. To address this question, we performed a network pharmacology study to explore the system-level mechanisms that underlie the analgesic activity of Jakyak-Gamcho decoction (JGd; Shaoyao-Gancao-Tang in Chinese and Shakuyaku-Kanzo-To in Japanese), an herbal prescription consisting of Paeonia lactiflora Pallas and Glycyrrhiza uralensis Fischer. Based on comprehensive information regarding the pharmacological and chemical properties of the herbal constituents of JGd, we identified 57 active chemical compounds and their 70 pain-associated targets. The JGd targets were determined to be involved in the regulation of diverse biological activities as follows: calcium- and cytokine-mediated signalings, calcium ion concentration and homeostasis, cellular behaviors of muscle and neuronal cells, inflammatory response, and response to chemical, cytokine, drug, and oxidative stress. The targets were further enriched in various pain-associated signalings, including the PI3K-Akt, estrogen, ErbB, neurotrophin, neuroactive ligand-receptor interaction, HIF-1, serotonergic synapse, JAK-STAT, and cAMP pathways. Thus, these data provide a systematic basis to understand the molecular mechanisms underlying the analgesic activity of herbal drugs.
Collapse
|