1
|
Abbasifard M, Bagherzadeh K, Khorramdelazad H. The story of clobenpropit and CXCR4: can be an effective drug in cancer and autoimmune diseases? Front Pharmacol 2024; 15:1410104. [PMID: 39070795 PMCID: PMC11272485 DOI: 10.3389/fphar.2024.1410104] [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: 03/31/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Clobenpropit is a histamine H3 receptor antagonist and has developed as a potential therapeutic drug due to its ability to inhibit CXCR4, a chemokine receptor involved in autoimmune diseases and cancer pathogenesis. The CXCL12/CXCR4 axis involves several biological phenomena, including cell proliferation, migration, angiogenesis, inflammation, and metastasis. Accordingly, inhibiting CXCR4 can have promising clinical outcomes in patients with malignancy or autoimmune disorders. Based on available knowledge, Clobenpropit can effectively regulate the release of monocyte-derived inflammatory cytokine in autoimmune diseases such as juvenile idiopathic arthritis (JIA), presenting a potential targeted target with possible advantages over current therapeutic approaches. This review summarizes the intricate interplay between Clobenpropit and CXCR4 and the molecular mechanisms underlying their interactions, comprehensively analyzing their impact on immune regulation. Furthermore, we discuss preclinical and clinical investigations highlighting the probable efficacy of Clobenpropit for managing autoimmune diseases and cancer. Through this study, we aim to clarify the immunomodulatory role of Clobenpropit and its advantages and disadvantages as a novel therapeutic opportunity.
Collapse
Affiliation(s)
- Mitra Abbasifard
- Department of Internal Medicine, School of Medicine, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Kowsar Bagherzadeh
- Eye Research Center, The Five Senses Health Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
2
|
Prince EW, Apps JR, Jeang J, Chee K, Medlin S, Jackson EM, Dudley R, Limbrick D, Naftel R, Johnston J, Feldstein N, Prolo LM, Ginn K, Niazi T, Smith A, Kilburn L, Chern J, Leonard J, Lam S, Hersh DS, Gonzalez-Meljem JM, Amani V, Donson AM, Mitra SS, Bandopadhayay P, Martinez-Barbera JP, Hankinson TC. Unraveling the complexity of the senescence-associated secretory phenotype in adamantinomatous craniopharyngioma using multimodal machine learning analysis. Neuro Oncol 2024; 26:1109-1123. [PMID: 38334125 PMCID: PMC11145462 DOI: 10.1093/neuonc/noae015] [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: 06/13/2023] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Cellular senescence can have positive and negative effects on the body, including aiding in damage repair and facilitating tumor growth. Adamantinomatous craniopharyngioma (ACP), the most common pediatric sellar/suprasellar brain tumor, poses significant treatment challenges. Recent studies suggest that senescent cells in ACP tumors may contribute to tumor growth and invasion by releasing a senesecence-associated secretory phenotype. However, a detailed analysis of these characteristics has yet to be completed. METHODS We analyzed primary tissue samples from ACP patients using single-cell, single-nuclei, and spatial RNA sequencing. We performed various analyses, including gene expression clustering, inferred senescence cells from gene expression, and conducted cytokine signaling inference. We utilized LASSO to select essential gene expression pathways associated with senescence. Finally, we validated our findings through immunostaining. RESULTS We observed significant diversity in gene expression and tissue structure. Key factors such as NFKB, RELA, and SP1 are essential in regulating gene expression, while senescence markers are present throughout the tissue. SPP1 is the most significant cytokine signaling network among ACP cells, while the Wnt signaling pathway predominantly occurs between epithelial and glial cells. Our research has identified links between senescence-associated features and pathways, such as PI3K/Akt/mTOR, MYC, FZD, and Hedgehog, with increased P53 expression associated with senescence in these cells. CONCLUSIONS A complex interplay between cellular senescence, cytokine signaling, and gene expression pathways underlies ACP development. Further research is crucial to understand how these elements interact to create novel therapeutic approaches for patients with ACP.
Collapse
Affiliation(s)
- Eric W Prince
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - John R Apps
- Oncology Department, Birmingham Women’s and Children’s NHS Foundation Trust, Birmingham, UK
| | - John Jeang
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
- Division of Pediatric Neurosurgery, Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Keanu Chee
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Stephen Medlin
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Eric M Jackson
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Roy Dudley
- Department of Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - David Limbrick
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Robert Naftel
- Department of Neurological Surgery, Vanderbilt University Medical Center, Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, Tennessee, USA
| | - James Johnston
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
- Division of Pediatric Neurosurgery, Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Neil Feldstein
- Department of Neurosurgery, Columbia University Medical Center, New York, New York, USA
| | - Laura M Prolo
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Lucile Packard Children’s Hospital, Stanford University School of Medicine, Palo Alto, California, USA
| | - Kevin Ginn
- The Division of Pediatric Hematology and Oncology, Department of Pediatrics, Children’s Mercy Hospital, Kansas City, Missouri, USA
| | - Toba Niazi
- Department of Pediatric Neurosurgery, Nicklaus Children’s Hospital, Miami, Florida, USA
| | - Amy Smith
- Department of Pediatric Hematology‐Oncology, Arnold Palmer Hospital, Orlando, Florida, USA
| | - Lindsay Kilburn
- Children’s National Health System, Center for Cancer and Blood Disorders, Washington, District of Columbia, USA
- Children’s National Health System, Brain Tumor Institute, Washington, District of Columbia, USA
| | - Joshua Chern
- Departments of Pediatrics and Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Pediatric Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Jeffrey Leonard
- Division of Pediatric Neurosurgery, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Sandi Lam
- Division of Neurosurgery, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - David S Hersh
- Division of Neurosurgery, Connecticut Children’s, Hartford, Connecticut, USA
| | | | - Vladimir Amani
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Andrew M Donson
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Siddhartha S Mitra
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Morgan Adams Foundation for Pediatric Brain Tumor Research Program, Aurora, Colorado, USA
| | - Pratiti Bandopadhayay
- Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer, Birth Defects Research Centre, GOS Institute of Child Health, University College London, London, UK
| | - Todd C Hankinson
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
3
|
Wang MJ, Xia Y, Gao QL. DNA Damage-driven Inflammatory Cytokines: Reprogramming of Tumor Immune Microenvironment and Application of Oncotherapy. Curr Med Sci 2024; 44:261-272. [PMID: 38561595 DOI: 10.1007/s11596-024-2859-1] [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: 12/15/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024]
Abstract
DNA damage occurs across tumorigenesis and tumor development. Tumor intrinsic DNA damage can not only increase the risk of mutations responsible for tumor generation but also initiate a cellular stress response to orchestrate the tumor immune microenvironment (TIME) and dominate tumor progression. Accumulating evidence documents that multiple signaling pathways, including cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) and ataxia telangiectasia-mutated protein/ataxia telangiectasia and Rad3-related protein (ATM/ATR), are activated downstream of DNA damage and they are associated with the secretion of diverse cytokines. These cytokines possess multifaced functions in the anti-tumor immune response. Thus, it is necessary to deeply interpret the complex TIME reshaped by damaged DNA and tumor-derived cytokines, critical for the development of effective tumor therapies. This manuscript comprehensively reviews the relationship between the DNA damage response and related cytokines in tumors and depicts the dual immunoregulatory roles of these cytokines. We also summarize clinical trials targeting signaling pathways and cytokines associated with DNA damage and provide future perspectives on emerging technologies.
Collapse
Affiliation(s)
- Meng-Jie Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Xia
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Qing-Lei Gao
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
4
|
Pang C, Li Y, Shi M, Fan Z, Gao X, Meng Y, Liu S, Gao C, Su P, Wang X, Zhan H. Expression and clinical value of CXCR4 in high grade gastroenteropancreatic neuroendocrine neoplasms. Front Endocrinol (Lausanne) 2024; 15:1281622. [PMID: 38524630 PMCID: PMC10960360 DOI: 10.3389/fendo.2024.1281622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 02/23/2024] [Indexed: 03/26/2024] Open
Abstract
Background CXC chemokine receptor 4 (CXCR4) is associated with the progression and metastasis of numerous malignant tumors. However, its relationship with Gastroenteropancreatic Neuroendocrine Neoplasms Grade 3 (GEP-NENs G3) is unclear. The aim of this study was to characterize the expression of CXCR4 in GEP-NENS and to explore the clinical and prognostic value of CXCR4. Methods This study retrospectively collected clinical and pathological data from patients with GEP-NENs who receiving surgery in Qilu Hospital of Shandong University from January 2013 to April 2021, and obtained the overall survival of the patients based on follow-up. Immunohistochemistry (IHC) was performed on pathological paraffin sections to observe CXCR4 staining. Groups were made according to pathological findings. Kaplan-Meier (K-M) curve was used to evaluate prognosis. SPSS 26.0 was used for statistical analysis. Results 100 GEP-NENs G3 patients were enrolled in this study. There was a significant difference in primary sites (P=0.002), Ki-67 index (P<0.001), and Carcinoembryonic Antigen (CEA) elevation (P=0.008) between neuroendocrine tumor (NET) G3 and neuroendocrine carcinoma (NEC). CXCR4 was highly expressed only in tumors, low or no expressed in adjacent tissues (P<0.001). The expression level of CXCR4 in NEC was significantly higher than that in NET G3 (P=0.038). The K-M curves showed that there was no significant difference in overall survival between patients with high CXCR4 expression and patients with low CXCR4 expression, either in GEP-NEN G3 or NEC (P=0.920, P=0.842. respectively). Conclusion Differential expression of CXCR4 was found between tumor and adjacent tissues and between NET G3 and NEC. Our results demonstrated that CXCR4 can be served as a new IHC diagnostic indicator in the diagnosis and differential diagnosis of GEP-NENs G3. Further studies with multi-center, large sample size and longer follow-up are needed to confirm the correlation between CXCR4 expression level and prognosis.
Collapse
Affiliation(s)
- Chaoyu Pang
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yongzheng Li
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ming Shi
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xin Gao
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yufan Meng
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Shujie Liu
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Changhao Gao
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Peng Su
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiao Wang
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
5
|
Bao S, Darvishi M, H Amin A, Al-Haideri MT, Patra I, Kashikova K, Ahmad I, Alsaikhan F, Al-Qaim ZH, Al-Gazally ME, Kiasari BA, Tavakoli-Far B, Sidikov AA, Mustafa YF, Akhavan-Sigari R. CXC chemokine receptor 4 (CXCR4) blockade in cancer treatment. J Cancer Res Clin Oncol 2023; 149:7945-7968. [PMID: 36905421 DOI: 10.1007/s00432-022-04444-w] [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: 05/25/2022] [Accepted: 10/19/2022] [Indexed: 03/12/2023]
Abstract
CXC chemokine receptor type 4 (CXCR4) is a member of the G protein-coupled receptors (GPCRs) superfamily and is specific for CXC chemokine ligand 12 (CXCL12, also known as SDF-1), which makes CXCL12/CXCR4 axis. CXCR4 interacts with its ligand, triggering downstream signaling pathways that influence cell proliferation chemotaxis, migration, and gene expression. The interaction also regulates physiological processes, including hematopoiesis, organogenesis, and tissue repair. Multiple evidence revealed that CXCL12/CXCR4 axis is implicated in several pathways involved in carcinogenesis and plays a key role in tumor growth, survival, angiogenesis, metastasis, and therapeutic resistance. Several CXCR4-targeting compounds have been discovered and used for preclinical and clinical cancer therapy, most of which have shown promising anti-tumor activity. In this review, we summarized the physiological signaling of the CXCL12/CXCR4 axis and described the role of this axis in tumor progression, and focused on the potential therapeutic options and strategies to block CXCR4.
Collapse
Affiliation(s)
- Shunshun Bao
- The First Clinical Medical College, Xuzhou Medical University, 221000, Xuzhou, China
| | - Mohammad Darvishi
- Infectious Diseases and Tropical Medicine Research Center (IDTMRC), Department of Aerospace and Subaquatic Medicine, AJA University of Medicinal Sciences, Tehran, Iran
| | - Ali H Amin
- Deanship of Scientific Research, Umm Al-Qura University, 21955, Makkah, Saudi Arabia
- Zoology Department, Faculty of Science, Mansoura University, 35516, Mansoura, Egypt
| | - Maysoon T Al-Haideri
- Department of Physiotherapy, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Indrajit Patra
- An Independent Researcher, National Institute of Technology Durgapur, Durgapur, West Bengal, India
| | | | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | | | | | - Bahman Abedi Kiasari
- Virology Department, Faculty of Veterinary Medicine, The University of Tehran, Tehran, Iran.
| | - Bahareh Tavakoli-Far
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran.
- Department of Physiology and Pharmacology, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| | - Akmal A Sidikov
- Rector, Ferghana Medical Institute of Public Health, Ferghana, Uzbekistan
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tübingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
6
|
Naimo GD, Paolì A, Giordano F, Forestiero M, Panno ML, Andò S, Mauro L. Unraveling the Role of Adiponectin Receptors in Obesity-Related Breast Cancer. Int J Mol Sci 2023; 24:ijms24108907. [PMID: 37240258 DOI: 10.3390/ijms24108907] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/28/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity has a noteworthy role in breast tumor initiation and progression. Among the mechanisms proposed, the most validated is the development of chronic low-grade inflammation, supported by immune cell infiltration along with dysfunction in adipose tissue biology, characterized by an imbalance in adipocytokines secretion and alteration of their receptors within the tumor microenvironment. Many of these receptors belong to the seven-transmembrane receptor family, which are involved in physiological features, such as immune responses and metabolism, as well as in the development and progression of several malignancies, including breast cancer. These receptors are classified as canonical (G protein-coupled receptors, GPCRs) and atypical receptors, which fail to interact and activate G proteins. Among the atypical receptors, adiponectin receptors (AdipoRs) mediate the effect of adiponectin, the most abundant adipocytes-derived hormone, on breast cancer cell proliferation, whose serum levels are reduced in obesity. The adiponectin/AdipoRs axis is becoming increasingly important regarding its role in breast tumorigenesis and as a therapeutic target for breast cancer treatment. The objectives of this review are as follows: to point out the structural and functional differences between GPCRs and AdipoRs, and to focus on the effect of AdipoRs activation in the development and progression of obesity-dependent breast cancer.
Collapse
Affiliation(s)
- Giuseppina Daniela Naimo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Alessandro Paolì
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Martina Forestiero
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Maria Luisa Panno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Loredana Mauro
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| |
Collapse
|
7
|
Integrin α6 Indicates a Poor Prognosis of Craniopharyngioma through Bioinformatic Analysis and Experimental Validation. JOURNAL OF ONCOLOGY 2022; 2022:6891655. [PMID: 36268277 PMCID: PMC9578790 DOI: 10.1155/2022/6891655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/17/2022] [Accepted: 09/24/2022] [Indexed: 11/18/2022]
Abstract
Background. Craniopharyngioma (CP) is a benign slow-growing tumor. It tends to affect children, and the number of patients is on rise. Considering the high morbidity and mortality of CP, it is urgent and pivotal to identify new biomarkers to uncover the etiology and pathogenesis of CP. Methods. The “limma” package was utilized to calculate the data from the Gene Expression Omnibus (GEO) database. Based on differentially expressed genes (DEGs), gene ontology and pathway analysis were deduced from the DAVID web tool. Further, we constructed a protein-protein interaction (PPI) network. Weighted correlation network analysis (WGCNA) was utilized to build a coexpression network. Finally, Western blotting and survival analysis were performed to examine the expression level of important metabolism-related genes. Results. Three hundred and eighty-four DEGs were identified between normal tissues and CPs from the GSE94349 and GSE26966 datasets. The Venn diagram for DEGs and hub genes in the ‘turquoise’ module revealed four key genes. Finally, the outcome of the survival analysis suggested that Integrin α6 (ITGA6) significantly affected the overall survival time of the patients with CP. Conclusion. IGTA6, as a metabolism-related molecule, was found to be substantially related to the overall survival of patients with CP.
Collapse
|
8
|
Zhu L, Yang F, Wang G, Li Q. CXC Motif Chemokine Receptor Type 4 Disrupts Blood-Brain Barrier and Promotes Brain Metastasis Through Activation of the PI3K/AKT Pathway in Lung Cancer. World Neurosurg 2022; 166:e369-e381. [PMID: 35817351 DOI: 10.1016/j.wneu.2022.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND CXC motif chemokine receptor type 4 (CXCR4) is an indispensable factor in the process of lung cancer brain metastasis (LCBM). The PI3K/AKT signal pathway is crucial in affecting cell invasion and metastasis and serves as a pivotal regulator in LCBM. However, the relationship between CXCR4 and the PI3K/AKT signal pathway is unclear. This study aimed to explore the underlying mechanisms of CXCR4 and PI3K/AKT in LCBM. METHODS Two lung cancer cells (A549 and H1299) and cells transfected with short hairpin RNA (shRNA)-CXCR4 were cocultured with normal human astrocyte cells and human brain endothelial (hCMEC/D3) cells to establish a blood-brain barrier model in vitro. The proliferation, migration, and invasion tight junction proteins (claudin-5, occludin, and ZO-1) were examined. Finally, results were verified in a nude mice model. RESULTS The abilities of cell proliferation, migration, and invasion were significantly reduced in A549 and H1299 cells transfected with shRNA-CXCR4 compared with the negative control group. The proteins phosphorylated PI3K and phosphorylated AKT were downregulated in lung cancer cells transfected with shRNA-CXCR4. The proteins claudin-5, occludin, and ZO-1 were upregulated in the A549 and H1299 cells transfected with shRNA-CXCR4. In vivo experiment results confirmed that the knockdown of CXCR4 played a protective role in the process of LCBM. CONCLUSIONS Our findings revealed that CXCR4 promotes LCBM by regulating the PI3K/Akt signal pathway. We also demonstrated that inhibiting CXCR4 could lead to prevention of LCBM. This study provides further rationale for clinical therapy that targets CXCR4/PI3K/AKT.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Thoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fugui Yang
- Department of Thoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guangxue Wang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qinchuan Li
- Department of Thoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
9
|
Katagiri T, Espinoza JL, Uemori M, Ikeda H, Hosokawa K, Ishiyama K, Yoroidaka T, Imi T, Takamatsu H, Ozawa T, Kishi H, Yamamoto Y, Elbadry MI, Yoshida Y, Chonabayashi K, Takenaka K, Akashi K, Nannya Y, Ogawa S, Nakao S. Hematopoietic stem progenitor cells with malignancy-related gene mutations in patients with acquired aplastic anemia are characterized by the increased expression of CXCR4. EJHAEM 2022; 3:669-680. [PMID: 36051022 PMCID: PMC9422028 DOI: 10.1002/jha2.515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 11/09/2022]
Abstract
The phenotypic changes in hematopoietic stem progenitor cells (HSPCs) with somatic mutations of malignancy-related genes in patients with acquired aplastic anemia (AA) are poorly understood. As our initial study showed increased CXCR4 expression on HLA allele-lacking (HLA[-]) HSPCs that solely support hematopoiesis in comparison to redundant HLA(+) HSPCs in AA patients, we screened the HSPCs of patients with various types of bone marrow (BM) failure to investigate their CXCR4 expression. In comparison to healthy individuals (n = 15, 12.3%-49.9%, median 43.2%), the median CXCR4+ cell percentages in the HSPCs of patients without somatic mutations were low: 29.3% (14.3%-37.3%) in the eight patients without HLA(-) granulocytes, 8.8% (4.1%-9.8%) in the five patients with HLA(-) cells accounting for >90% of granulocytes, and 7.8 (2.1%-8.7%) in the six patients with paroxysmal nocturnal hemoglobinuria. In contrast, the median percentage was much higher (78% [61.4%-88.7%]) in the five AA patients without HLA(-) granulocytes possessing somatic mutations (c-kit, t[8;21], monosomy 7 [one for each], ASXL1 [n = 2]), findings that were comparable to those (66.5%, 63.1%-88.9%) in the four patients with advanced myelodysplastic syndromes. The increased expression of CXCR4 may therefore reflect intrinsic abnormalities of HSPCs caused by somatic mutations that allow them to evade restriction by BM stromal cells.
Collapse
Affiliation(s)
- Takamasa Katagiri
- Department of Clinical Laboratory ScienceGraduate School of Medical ScienceInstitute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaIshikawaJapan
| | - Jorge Luis Espinoza
- Department of Occupational TherapyGraduate School of Medical ScienceInstitute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaIshikawaJapan
| | - Mizuho Uemori
- Department of Clinical Laboratory ScienceGraduate School of Medical ScienceInstitute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaIshikawaJapan
| | - Honoka Ikeda
- Department of Clinical Laboratory ScienceGraduate School of Medical ScienceInstitute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaIshikawaJapan
| | - Kohei Hosokawa
- Department of HematologyFaculty of MedicineInstitute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaIshikawaJapan
| | - Ken Ishiyama
- Department of HematologyFaculty of MedicineInstitute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaIshikawaJapan
| | - Takeshi Yoroidaka
- Department of HematologyFaculty of MedicineInstitute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaIshikawaJapan
| | - Tatsuya Imi
- Department of HematologyFaculty of MedicineInstitute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaIshikawaJapan
| | - Hiroyuki Takamatsu
- Department of HematologyFaculty of MedicineInstitute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaIshikawaJapan
| | - Tatsuhiko Ozawa
- Department of ImmunologyFaculty of MedicineAcademic AssemblyUniversity of ToyamaToyama CityToyamaJapan
| | - Hiroyuki Kishi
- Department of ImmunologyFaculty of MedicineAcademic AssemblyUniversity of ToyamaToyama CityToyamaJapan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular BiologyKanazawa University Graduate School of Medical SciencesKanazawaIshikawaJapan
| | - Mahmoud Ibrahim Elbadry
- Division of HematologyDepartment of Internal MedicineFaculty of MedicineSohag UniversitySohagEgypt
| | - Yoshinori Yoshida
- Center for iPS Cell Research and ApplicationKyoto UniversitySakyo‐kuKyotoJapan
| | - Kazuhisa Chonabayashi
- Center for iPS Cell Research and ApplicationKyoto UniversitySakyo‐kuKyotoJapan
- Department of Hematology and OncologyGraduate School of MedicineKyoto UniversitySakyo‐kuKyotoJapan
| | - Katsuto Takenaka
- Department of HematologyClinical Immunology and Infectious DiseasesEhime University Graduate School of MedicineToonEhimeJapan
| | - Koichi Akashi
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuoka CityFukuokaJapan
| | - Yasuhito Nannya
- Division of Hematopoietic Disease ControlInstitute of Medical ScienceUniversity of TokyoMinato‐kuTokyoJapan
- Department of Pathology and Tumor BiologyKyoto UniversityYoshida‐Konoe‐choSakyo‐kuKyotoJapan
| | - Seishi Ogawa
- Department of Pathology and Tumor BiologyKyoto UniversityYoshida‐Konoe‐choSakyo‐kuKyotoJapan
- Institute for the Advanced Study of Human Biology (WPI‐ASHBi)Kyoto UniversitySakyo‐kuKyotoJapan
- Department of MedicineCentre for Hematology and Regenerative MedicineKarolinska InstituteStockholmSweden
| | - Shinji Nakao
- Department of HematologyFaculty of MedicineInstitute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaIshikawaJapan
| |
Collapse
|
10
|
Wu Y, Hu Y, Tang L, Yin S, Lv L, Zhou P. Targeting CXCR4 to suppress glioma-initiating cells and chemoresistance in glioma. Cell Biol Int 2022; 46:1519-1529. [PMID: 35731168 DOI: 10.1002/cbin.11836] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 04/09/2022] [Accepted: 05/07/2022] [Indexed: 02/05/2023]
Abstract
Glioma initiating cells (GICs), also known as glioma stem cells, display the capacity to recapitulate the functional diversity within the tumor. Despite the great progress achieved over the last decades, defining the key molecular regulators of GICs has represented a major obstacle in this field. In our study, data from The Cancer Genome Atlas database illustrated a relationship between C-X-C motif chemokine receptor 4 (CXCR4) expression and the survival of glioma patients. Mechanistically, we further indicated that CXCR4 mediated the upregulation of Kruppel like factor 5 (KLF5), a zinc-finger-containing transcription factor, to facilitate the proliferation of GICs. What's more, CXCR4 also enhanced the chemoresistance through KLF5/Bcl2-like 12 (BCl2L12) in glioma. The elevated expression of KLF5 and BCL2L12 induced by CXCR4 was dependent on phosphoinositide 3-kinases (PI3K)/serine/threonine kinase (AKT) signaling. Importantly, combined application of temozolomide and a CXCR4 inhibitor efficiently reversed CXCR4 mediated drugs resistance and improved anticancer effects in vivo. Collectively, our findings confirmed that CXCR4 promoted GICs proliferation via the KLF5/BCL2L12 dependent pathway, which may enrich the understanding of GICs and help drive the design of efficacious therapeutic strategies.
Collapse
Affiliation(s)
- Yao Wu
- Department of Neurosurgery, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yu Hu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Lingli Tang
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Senlin Yin
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Liang Lv
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Peizhi Zhou
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
11
|
LncRNA PTAR activates the progression of bladder cancer by modulating miR-299-3p/CD164 axis. Pathol Res Pract 2022; 237:153994. [DOI: 10.1016/j.prp.2022.153994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/01/2022] [Accepted: 06/21/2022] [Indexed: 12/24/2022]
|
12
|
Yan M, Bai R, Zhang H, Yan W. Stromal Cell-Derived Factor-1 α (SDF-1 α) Promotes Growth and Migration of Bone Marrow Stromal Cells (BMSCs) and Gastric Cancer Cells Through Phosphatidylinositol 3-Kinase/AKT (PI3K/Akt) Pathway. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
SDF-1α activity is closely related to information transmission and cell migration when contributing to lymphatic metastasis in various tumors. Herein, we explored the interaction among SDF-1α, CXCR4 and PI3K/Akt signaling pathway in gastric cancer (GC) and
their roles in this disorder. Human GC cells KATO-III and BMSCs were co-cultured without contact. GC cells were transfected with SDF-1α, CXCR4 inhibitor, and PI3K inhibitor. After examining the efficiency of transfection, cell migration was evaluated using Transwell chamber, and
expression SDF-1α, CD133, and CXCR4 was determined by RT-qPCR. With transfection rate of 98%, the number of migrated cells reduced upon inhibition of CXCR4 and PI3K. Luciferase activity in 565 nm are high than CXCR4 inhibition group. (p < 0.05). Likewise, up-regulation
of SDF-1α increased the expression of SDF-1 (0.825±0.061), CD133 (0.875±0.058), CXCR4 (0.801±0.052), and Akt (0.852±0.062), compared to the blank group, CXCR4 inhibition group and PI3K inhibition group (p < 0.05). Down-regulation of CXCR4
and PI3K, however, decreased the expression insignificantly (p > 0.05). Collectively, up-regulation of SDF-1α activates CXCR4 signaling pathway of BMSCs and stimulates its downstream PI3K/Akt signaling pathway and and increases the expression of CD133, thereby promoting
malignant behaviors of GC cells.
Collapse
Affiliation(s)
- Ming Yan
- Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, Fengtai District, Beijing, 100070, China
| | - Ringxing Bai
- Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, Fengtai District, Beijing, 100070, China
| | - Hongyi Zhang
- Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, Fengtai District, Beijing, 100070, China
| | - Wenmao Yan
- Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, Fengtai District, Beijing, 100070, China
| |
Collapse
|
13
|
Yang KD, Wang Y, Zhang F, Luo BH, Feng DY, Zeng ZJ. CircN4BP2L2 promotes colorectal cancer growth and metastasis through regulation of the miR-340-5p/CXCR4 axis. J Transl Med 2022; 102:38-47. [PMID: 34326457 DOI: 10.1038/s41374-021-00632-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 11/09/2022] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related death worldwide. Dysregulation of circular RNAs (circRNAs) appears to be a critical factor in CRC progression. However, mechanistic studies delineating the role of circRNAs in CRC remain limited. In this study, qRT-PCR and western blot assays were used to measure the expression of genes and proteins. Migration, invasion, proliferation, and apoptosis were examined by wound-healing, transwell, CCK-8, colony formation, and flow cytometry assays, respectively. Molecular interactions were validated by a dual-luciferase report system. A xenograft animal model was established to examine in vivo tumor growth and lung metastasis. Our data indicated that circN4BP2L2 expression was increased in CRC tissues and cell lines. Notably, inhibition of circN4BP2L2 effectively inhibited proliferation, migration, and invasion of LoVo cells, and inhibited tumor growth and metastasis in vivo, whereas the forced expression of circN4BP2L2 facilitated the proliferation, migration, and invasion of HT-29 cells. Mechanistic studies revealed that circN4BP2L2 acted as a molecular sponge of miR-340-5p to competitively promote CXCR4 expression. Furthermore, inhibition of miR-340-5p reversed the anti-cancer effects of circN4BP2L2 or CXCR4 silencing. Our data indicated an oncogenic role of circN4BP2L2 in CRC via regulation of the miR-340-5p/CXCR4 axis, which may be a promising biomarker and target for CRC treatment.
Collapse
Affiliation(s)
- Ke-Da Yang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
| | - Ying Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
| | - Fan Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
| | - Bai-Hua Luo
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
| | - De-Yun Feng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
| | - Zhi-Jun Zeng
- Department of Geriatric Surgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China.
| |
Collapse
|
14
|
Si M, Ma Z, Zhang J, Li X, Li R, Wang C, Jia H, Luo S. Qingluoyin granules protect against adjuvant-induced arthritis in rats via downregulating the CXCL12/CXCR4-NF-κB signalling pathway. PHARMACEUTICAL BIOLOGY 2021; 59:1441-1451. [PMID: 34693865 PMCID: PMC8547818 DOI: 10.1080/13880209.2021.1991386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/02/2021] [Accepted: 10/05/2021] [Indexed: 06/13/2023]
Abstract
CONTEXT Qingluoyin (QLY) is a traditional Chinese medicine (TCM) formula which has been used in treating human rheumatoid arthritis (RA) for years in China. OBJECTIVE This study investigates the effect of QLY granules on adjuvant arthritis (AA) and the possible mechanism. MATERIALS AND METHODS Sprague-Dawley (SD) rats were injected with Complete Freund's adjuvant (CFA) to induce the AA model. After the onset of arthritis, rats received intragastric administrations of the QLY granules (1.35, 2.70, and 5.40 g/kg) or Tripterygium glycosides (TG) tablets (positive drug, 10 mg/kg) for 14 d. After 28 d immunization, the symptoms, inflammatory parameters and molecular mechanisms were investigated. RESULTS In the QLY granule (1.35, 2.70, and 5.40 g/kg) therapy groups, the arthritis index decreased to 6.30 ± 2.06, 5.80 ± 1.55, 5.30 ± 1.16 compared with the model (9.00 ± 3.01), paw swelling decreased to 1.56 ± 0.40, 1.28 ± 0.38, 1.12 ± 0.41 mL compared with the model (2.22 ± 0.73 mL). QLY granules (1.35, 2.70 and 5.40 g/kg) significantly reduced the thymus and the spleen indexes, inhibited the production of pro-inflammatory cytokines, and alleviated the pathological changes of joints compared with the model group. Furthermore, the treatment of QLY granules (2.70 and 5.40 g/kg) markedly inhibited CXCL12, CXCR4 (in spleen and synovium) and p-NF-κB p65 (in synovium) protein expression of AA rats. CONCLUSIONS QLY granules have obvious therapeutic effects on AA rats, which may be associated with downregulating the CXCL12/CXCR4-NF-κB signalling pathway. QLY granules can be used as a candidate for the treatment of RA, which deserves further study.
Collapse
Affiliation(s)
- Min Si
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Zheng Ma
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Jie Zhang
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Xinwei Li
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Rui Li
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Chao Wang
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Huiyu Jia
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| | - Shengyong Luo
- Anhui Academy of Medical Sciences, Hefei, Anhui, China
| |
Collapse
|
15
|
Li M, Zhou L, Li S, Fang L, Yang L, Wu X, Yang C, Bao Y, Lan S, Tong Z, Zheng S, Tang B, Zeng E, Xie S, Chen C, Hong T. MMP12 is a potential therapeutic target for Adamantinomatous craniopharyngioma: Conclusions from bioinformatics analysis and in vitro experiments. Oncol Lett 2021; 22:536. [PMID: 34084216 PMCID: PMC8161407 DOI: 10.3892/ol.2021.12797] [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: 09/22/2020] [Accepted: 04/20/2021] [Indexed: 12/24/2022] Open
Abstract
Adamantinomatous craniopharyngioma (ACP) is considered a benign intracranial tumor, but it can also exhibit aggressive characteristics. Due to its unique location in the suprasellar, which brings it close to important nerves and vascular structures, ACP can often lead to significant neuroendocrine diseases. The current treatments primarily include surgical intervention, radiation therapy or a combination of the two, but these can lead to serious complications and adversely affect the quality of life of patients. Thus, it is important to identify effective and safe alternatives. Recently, studies have focused on the tumor genome, transcriptome and proteome in an attempt to identify potential therapeutic targets for clinical use. However, studies on this region of the CP are limited; thus, the present study focused on this region. The GSE94349 and GSE68015 datasets were downloaded from the Gene Expression Omnibus database and analyzed. In the in vitro studies, the effect of the matrix metalloproteinase (MMP)12 inhibitor, MMP408, on cell proliferation and protein expression was assessed. The results demonstrated that MMP408 effectively inhibited cell proliferation and migration of ACP cells, and decreased the expression levels of the related proteins. Thus, MMP12 may be used as a potential therapeutic target for the treatment of ACP.
Collapse
Affiliation(s)
- Minde Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lin Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shaoyang Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Linchun Fang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Le Yang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiao Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Chenxing Yang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Youyuan Bao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Sihai Lan
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhigao Tong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Suyue Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bin Tang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Erming Zeng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shenhao Xie
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Cheng Chen
- Department of Rehabilitation Medicine, Lushan Sanatorium, Jiujiang, Jiangxi 332000, P.R. China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
16
|
Wang J, Wang Y, Zuo Y, Duan J, Pan A, Li JM, Yan XX, Liu F. MFGE8 mitigates brain injury in a rat model of SAH by maintaining vascular endothelial integrity via TIGβ5/PI3K/CXCL12 signaling. Exp Brain Res 2021; 239:2193-2205. [PMID: 33991211 DOI: 10.1007/s00221-021-06111-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 04/08/2021] [Indexed: 12/25/2022]
Abstract
Leaked blood components, injured endothelial cells, local inflammatory response and vasospasm may converge to promote microthrombosis following subarachnoid hemorrhage (SAH). Previously, we showed that the milk fat globule-epidermal growth factor 8 (MFGE8) can mitigate SAH-induced microthrombosis. This present study was aimed to explore the molecular pathway participated in MFGE8-dependent protection on vascular endothelium. Immunofluorescence, immunoblot and behavioral tests were used to determine the molecular partner and signaling pathway mediating the effect of MFGE8 in vascular endothelium in rats with experimental SAH and controls, together with the applications of RNA silencing and pharmacological intervention methods. Relative to control, recombinant human MFGE8 (rhMFGE8) treatment increased 5-bromo-2'-deoxyuridine (BrdU) labeled new endothelial cells, reduced TUNUL-positive endothelial cells and elevated the expression of phosphatidylinositol 3-kinase (PI3K) and chemokine (C-X-C motif) ligand 12 (CXCL12), in the brains of SAH rats. These effects were reversed by MFGE8 RNA silencing, as well as following cilengitide and wortmannin intervention. These results suggest that MFGE8 promotes endothelial regeneration and mitigates endothelial DNA damage through the activation of the TIGβ5/PI3K/CXCL12 signaling pathway.
Collapse
Affiliation(s)
- Jikai Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No. 52 Meihuadong Road, Zhuhai, 519000, Guangdong, China
| | - Yiping Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No. 52 Meihuadong Road, Zhuhai, 519000, Guangdong, China
| | - Yuchun Zuo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jiajia Duan
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Jian-Ming Li
- Department of Anatomy, School of Basic Sciences, Changsha Medical University, Changsha, 410219, Hunan, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Fei Liu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, No. 52 Meihuadong Road, Zhuhai, 519000, Guangdong, China.
| |
Collapse
|
17
|
Hou Y, Chen K, Liao R, Li Y, Yang H, Gong J. LINC01419-mediated epigenetic silencing of ZIC1 promotes metastasis in hepatocellular carcinoma through the PI3K/Akt signaling pathway. J Transl Med 2021; 101:570-587. [PMID: 33772101 DOI: 10.1038/s41374-021-00539-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a rapidly growing tumor characterized by a high potential for vascular invasion and metastasis. The purpose of our study is to explore the regulation mechanism of long noncoding RNA (lncRNA) LINC01419 on cell-cycle distribution and metastasis in hepatocellular carcinoma (HCC) by regulating zinc finger of the cerebellum (ZIC1) through PI3K/Akt signaling pathway. Bioinformatics analysis and dual-luciferase reporter assay were used to analyze LINC01419 and related genes in HCC, and their expression in HCC tissues and adjacent normal tissues were determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot. Then, HCC cell lines were subjected to the construction of LINC01419/ZIC1 overexpression/knockdown cells utilizing lentiviral vectors. RIP and ChIP assays were applied to identify the LINC01419-binding protein. BSP and MSP assays were used to determine gene methylation. According to the results, LINC01419 was highly expressed in HCC tissues and cells, while ZIC1 was poorly expressed. LINC01419 targeted and downregulated ZIC1 expression. Furthermore, LINC01419 increased the methylation of ZIC1 promoter and repressed ZIC1 expression. PI3K/Akt signaling pathway was activated by LINC01419 overexpression and ZIC1 knockdown, under which conditions, the HCC cell self-renewal and proliferation were promoted while cell apoptosis was attenuated, accompanied by accelerated formation and metastasis of xenografted tumors in mice. In conclusion, LINC01419 enhances the methylation of ZIC1 promoter, inhibits ZIC1 expression, and activates the PI3K/Akt signaling pathway, thereby enhancing the malignant phenotypes of HCC cells in vitro as well as tumor formation and metastasis in vivo.
Collapse
Affiliation(s)
- Yifu Hou
- Organ Transplant Center and Third Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, PR China
| | - Kai Chen
- Organ Transplant Center and Third Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, PR China
| | - Rui Liao
- Department of Hepatobiliary, Southwest Medical University, Luzhou, PR China
| | - Youzan Li
- Department of Hepatobiliary, Southwest Medical University, Luzhou, PR China
| | - Hongji Yang
- Organ Transplant Center and Third Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China.
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, PR China.
| | - Jun Gong
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, PR China.
- Second Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China.
| |
Collapse
|
18
|
Tang K, Zhang H, Li Y, Sun Q, Jin H. Circular RNA as a Potential Biomarker for Melanoma: A Systematic Review. Front Cell Dev Biol 2021; 9:638548. [PMID: 33869186 PMCID: PMC8047128 DOI: 10.3389/fcell.2021.638548] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/08/2021] [Indexed: 12/22/2022] Open
Abstract
Circular RNAs (circRNAs) are newly discovered RNAs with covalently looped structures. Due to their resistance to RNAase degradation and tissue-specific expression, circRNAs are expected to be potential biomarkers in early diagnosis and target treatment of many diseases. However, the role of circRNAs in melanoma still needs to be systematically reviewed for better understanding and further research. Based on published articles in PubMed, Embase, Cochrane Library, and Web of Science database, we systematically reviewed the implications and recent advances of circRNAs in melanoma, focusing on function, mechanism, and correlation with melanoma progression. According to inclusion and exclusion criteria, a total of 19 articles were finally included in this systematic review. Of the 19 studies, 17 used human samples, including melanoma tissues (n = 16) and blood serum of patients with melanoma (n = 1). The sample size of the study group ranged from 20 to 105 based on the reported data. Several studies explored the association between circRNAs and clinicopathological characteristics. circRNA dysregulation was commonly observed in melanoma patients. circRNAs function in melanoma by miRNA sponging and interaction with RNA binding proteins (RBP), ultimately controlling several important signaling pathways and cancer-related cellular processes, including proliferation, migration, invasion, metastasis, apoptosis, and glucose metabolism. circRNA expression could be associated with prognostic factors and drug responses, consolidating the potential clinical value in melanoma. Herein, we clarified the functional, prognostic, and predictive roles of circRNAs in melanoma in this systematic review, providing future directions for studies on melanoma-associated circRNAs.
Collapse
Affiliation(s)
- Keyun Tang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hanlin Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yaqi Li
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Qiuning Sun
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hongzhong Jin
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
19
|
Wu X, Zhang H, Sui Z, Wang Y, Yu Z. The biological role of the CXCL12/CXCR4 axis in esophageal squamous cell carcinoma. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0140. [PMID: 33710803 PMCID: PMC8185864 DOI: 10.20892/j.issn.2095-3941.2020.0140] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022] Open
Abstract
Esophageal cancer is the eighth most common malignant tumor and the sixth leading cause of cancer-related death worldwide. Esophageal squamous cell carcinoma (ESCC) is the main histological type of esophageal cancer, and accounts for 90% of all cancer cases. Despite the progress made in surgery, chemotherapy, and radiotherapy, the mortality rate from esophageal cancer remains high, and the overall 5-year survival rate is less than 20%, even in developed countries. The C-X-C motif chemokine ligand 12 (CXCL12) is a member of the CXC chemokine subgroup, which is widely expressed in a variety of tissues and cells. CXCL12 participates in the regulation of many physiological and pathological processes by binding to its specific receptor, C-X-C motif chemokine receptor type 4 (CXCR4), where it causes embryonic development, immune response, and angiogenesis. In addition, increasing evidence indicates that the CXCL12/CXCR4 axis plays an important role in the biological processes of tumor cells. Studies have shown that CXCL12 and its receptor, CXCR4, are highly expressed in ESCC. This abnormal expression contributes to tumor proliferation, lymph node and distant metastases, and worsening prognosis. At present, antagonists and imaging agents against CXCL12 or CXCR4 have been developed to interfere with the malignant process and monitor metastasis of tumors. This article summarizes the structure, function, and regulatory mechanism of CXCL12/CXCR4 and its role in the malignancy of ESCC. Current results from preclinical research targeting CXCL12/CXCR4 are also summarized to provide a reference for the clinical diagnosis and treatment of ESCC.
Collapse
Affiliation(s)
- Xianxian Wu
- Departments of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Hongdian Zhang
- Departments of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhilin Sui
- Departments of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yang Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhentao Yu
- Departments of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
20
|
Zhang H, Luan S, Xiao X, Lin L, Zhao X, Liu X. Silenced microRNA-222 suppresses inflammatory response in gestational diabetes mellitus mice by promoting CXCR4. Life Sci 2020; 266:118850. [PMID: 33278386 DOI: 10.1016/j.lfs.2020.118850] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Gestational diabetes mellitus (GDM) is induced by multiple factors, and the microRNAs (miRNAs) are well-known to be implicated in GDM progression. We aimed to explore the functional mechanisms of miR-222 in the inflammatory response in GDM by mediating C-X-C chemokine receptor type 4 (CXCR4) and NLRP3 inflammasomes. METHODS GDM models were established by intraperitoneal injection of streptozocin, and the levels of miR-222 and CXCR4 in GDM patients' placenta tissues as well as GDM mice' placenta and pancreatic tissues were determined. The GDM mice were treated with miR-222 Antagomir/Agomir or overexpressed CXCR4 to evaluate the apoptosis and pathological changes in tissues, and the levels of blood glucose, insulin, biochemical indices, inflammatory factors and inflammasome-related proteins. Importantly, the target relation between miR-222 and CXCR4 was verified. RESULTS MiR-222 was increased while CXCR4 was decreased in GDM patients and mice. The down-regulated miR-222 and up-regulated CXCR4 could promote insulin sensitivity and insulin level, while inhibit apoptosis, inflammation and glucagon level in GDM mice. Moreover, CXCR4 was targeted by miR-222. CONCLUSION We demonstrated that the silenced miR-222 could suppress inflammatory response in GDM mice by promoting CXCR4 and inactivating NLRP3 inflammasomes, which may contribute to GDM treatment.
Collapse
Affiliation(s)
- Haiyu Zhang
- Department of Prenatal Diagnosis, Weifang People's Hospital, Weifang 261041, Shandong, China
| | - Shoujing Luan
- Department of Endocrinology and Metabolism, Weifang People's Hospital, Weifang 261041, Shandong, China
| | - Xiao Xiao
- Department of Endocrinology and Metabolism, Weifang People's Hospital, Weifang 261041, Shandong, China
| | - Lingyu Lin
- Clinical Laboratory, Weifang People's Hospital, Weifang 261041, Shandong, China
| | - Xiaowei Zhao
- Department of Prenatal Diagnosis, Weifang People's Hospital, Weifang 261041, Shandong, China
| | - Xueyao Liu
- Clinical Laboratory, Weifang People's Hospital, Weifang 261041, Shandong, China.
| |
Collapse
|
21
|
Xia M, Shao J, Qiao M, Luo Z, Deng X, Ke Q, Dong X, Shen L. Identification of LCA-binding Glycans as a Novel Biomarker for Esophageal Cancer Metastasis using a Lectin Array-based Strategy. J Cancer 2020; 11:4736-4745. [PMID: 32626520 PMCID: PMC7330695 DOI: 10.7150/jca.43806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/16/2020] [Indexed: 12/17/2022] Open
Abstract
Esophageal cancer (EC) is a unique and heterogeneous disease diagnosed mostly at advanced stages. Altered glycans presented on cell surfaces are involved in the occurrence and development of malignancy. However, the effects of glycans on EC progression are largely unexplored. Here, a lectin array was utilized to detect the glycan profiling of the normal esophageal mucosal epithelial cell line and two EC cell lines. The binding of Lens culinaris lectin (LCA) to EC cells was found to be stronger than that of the normal cells. Lectin immunohistochemical staining revealed that LCA-binding glycans were markedly elevated in EC tissues compared to adjacent non-cancerous tissues. LCA staining was significantly associated with lymph node metastasis, depth of invasion, TNM stage and poor overall survival of EC patients. Added LCA to block LCA recognized glycans could inhibit the migration and invasion of EC cells. Further analysis revealed that blocking the biosynthesis of LCA-binding glycans by tunicamycin attenuated cellular migratory and invasive abilities. Additionally, a membrane glycoprotein CD147 was recognized as a binder of LCA. There was a positive correlation between LCA-binding glycans and CD147 expression in clinical samples. Interestingly, CD147 inhibition also reduced cell migration and invasion. These findings indicated that LCA-binding glycans may function as a novel indicator to predict metastasis for patients with EC.
Collapse
Affiliation(s)
- Min Xia
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China.,Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Jun Shao
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Meimei Qiao
- Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Zhiguo Luo
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xinzhou Deng
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Qing Ke
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xiaoxia Dong
- Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Li Shen
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China.,Department of Biochemistry, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan,Hubei 442000, P.R. China
| |
Collapse
|
22
|
Wei CY, Zhu MX, Lu NH, Liu JQ, Yang YW, Zhang Y, Shi YD, Feng ZH, Li JX, Qi FZ, Gu JY. Circular RNA circ_0020710 drives tumor progression and immune evasion by regulating the miR-370-3p/CXCL12 axis in melanoma. Mol Cancer 2020; 19:84. [PMID: 32381016 PMCID: PMC7204052 DOI: 10.1186/s12943-020-01191-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/24/2020] [Indexed: 02/26/2023] Open
Abstract
Background Circular RNAs (circRNAs) have been reported to have critical regulatory roles in tumor biology. However, their contribution to melanoma remains largely unknown. Methods CircRNAs derived from oncogene CD151 were detected and verified by analyzing a large number of melanoma samples through quantitative real-time polymerase chain reaction (qRT-PCR). Melanoma cells were stably transfected with lentiviruses using circ_0020710 interference or overexpression plasmid, and then CCK-8, colony formation, wound healing, transwell invasion assays, and mouse xenograft models were employed to assess the potential role of circ_0020710. RNA immunoprecipitation, luciferase reporter assay and fluorescence in situ hybridization were used to evaluate the underlying mechanism of circ_0020710. Results Our findings indicated that circ_0020710 was generally overexpressed in melanoma tissues, and high level of circ_0020710 was positively correlated with malignant phenotype and poor prognosis of melanoma patients. Elevated circ_0020710 promoted melanoma cell proliferation, migration and invasion in vitro as well as tumor growth in vivo. Mechanistically, we found that high level of circ_0020710 could upregulate the CXCL12 expression via sponging miR-370-3p. CXCL12 downregulation could reverse the malignant behavior of melanoma cells conferred by circ_0020710 over expression. Moreover, we also found that elevated circ_0020710 was correlated with cytotoxic lymphocyte exhaustion, and a combination of AMD3100 (the CXCL12/CXCR4 axis inhibitor) and anti-PD-1 significantly attenuated tumor growth. Conclusions Elevated circ_0020710 drives tumor progression via the miR-370-3p/CXCL12 axis, and circ_0020710 is a potential target for melanoma treatment.
Collapse
Affiliation(s)
- Chuan-Yuan Wei
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Meng-Xuan Zhu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Nan-Hang Lu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Jia-Qi Liu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Yan-Wen Yang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Yong Zhang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Yue-Dong Shi
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Zi-Hao Feng
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Jia-Xia Li
- Department of Neurology, Hefei Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, P. R. China
| | - Fa-Zhi Qi
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.
| | - Jian-Ying Gu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.
| |
Collapse
|
23
|
Wang TX, Tan WL, Huang JC, Cui ZF, Liang RD, Li QC, Lu H. Identification of aberrantly methylated differentially expressed genes targeted by differentially expressed miRNA in osteosarcoma. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:373. [PMID: 32355817 PMCID: PMC7186728 DOI: 10.21037/atm.2020.02.74] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Osteosarcoma (OS) is the most common primary bone tumors diagnosed in children and adolescents. Recent studies have shown a prognostic role of DNA methylation in various cancers, including OS. The aim of this study was to identify the aberrantly methylated genes that are prognostically relevant in OS. Methods The differentially expressed mRNAs, miRNAs and methylated genes (DEGs, DEMs and DMGs respectively) were screened from various GEO databases, and the potential target genes of the DEMs were predicted by the RNA22 program. The protein-protein interaction (PPI) networks were constructed using the STRING database and visualized by Cytoscape software. The functional enrichment and survival analyses of the screened genes was performed using the R software. Results Forty-seven downregulated hypermethylated genes and three upregulated hypomethylated genes were identified that were enriched in cell activation, migration and proliferation functions, and were involved in cancer-related pathways like JAK-STAT and PI3K-AKT. Eight downregulated hypermethylated tumor suppressor genes (TSGs) were identified among the screened genes based on the TSGene database. These hub genes are likely involved in OS genesis, progression and metastasis, and are potential prognostic biomarkers and therapeutic targets. Conclusions TSGs including PYCARD, STAT5A, CXCL12 and CXCL14 were aberrantly methylated in OS, and are potential prognostic biomarkers and therapeutic targets. Our findings provide new insights into the role of methylation in OS progression.
Collapse
Affiliation(s)
- Ting-Xuan Wang
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Zhuhai 519000, China
| | - Wen-Le Tan
- Department of Orthopedics, Luoding People's Hospital, Luoding 527200, China
| | - Jin-Cheng Huang
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou 450003, China
| | - Zhi-Fei Cui
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Zhuhai 519000, China
| | - Ri-Dong Liang
- Department of Orthopedics, Southern Medical University Affiliated Nanhai Hospital, Southern Medical University, Foshan 523800, China
| | - Qing-Chu Li
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Academy of Orthopedics, Southern Medical University, Guangzhou 510000, China
| | - Hai Lu
- Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Zhuhai 519000, China
| |
Collapse
|
24
|
Chen L, Zhu M, Yu S, Hai L, Zhang L, Zhang C, Zhao P, Zhou H, Wang S, Yang X. Arg kinase mediates CXCL12/CXCR4-induced invadopodia formation and invasion of glioma cells. Exp Cell Res 2020; 389:111893. [PMID: 32035133 DOI: 10.1016/j.yexcr.2020.111893] [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] [Received: 10/17/2019] [Revised: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 01/01/2023]
Abstract
Compared with noninvasive tumor cells, glioma cells overexpress chemokine receptor type 4 (CXCR4), which exhibits significantly greater expression in invasive tumor cells than in noninvasive tumor cells. C-X-C motif chemokine ligand 12 (CXCL12, also known as stromal derived factor-1, SDF-1) and its cell surface receptor CXCR4 activate a signaling axis that induces the expression of membrane type-2 matrix metalloproteinase (MT2-MMP), which plays a pivotal role in the invasion and migration of various cancer cells; however, the specific mechanism involved in this is unclear. Recently, studies have shown that invadopodia can recruit and secrete related enzymes, such as matrix metalloproteinases (MMPs), to degrade the surrounding extracellular matrix (ECM), promoting the invasion and migration of tumor cells. Phosphorylated cortactin (pY421-cortactin) is required for the formation and maturation of invadopodia, but the upstream regulatory factors and kinases involved in phosphorylation have not been elucidated. In this study, we found that CXCL12/CXCR4 was capable of inducing glioma cell invadopodia formation, probably by regulating cortactin phosphorylation. The interaction of cortactin and Arg (also known as Abl-related nonreceptor tyrosine kinase, ABL2) in glioma cells was demonstrated. The silencing of Arg inhibited glioma cell invadopodia formation and invasion by blocking cortactin phosphorylation. Moreover, CXCL12 could not induce glioma cell invasion in Arg-knockdown glioma cells. Based on these results, it can be concluded that Arg mediates CXCL12/CXCR4-induced glioma cell invasion, and CXCL12/CXCR4 regulates invadopodia maturation through the Arg-cortactin pathway, which indicates that Arg could be a candidate therapeutic target to inhibit glioma cell invasion.
Collapse
Affiliation(s)
- Lei Chen
- Department of Neurosurgery, Tianjin First Central Hospital, Tianjin, 300192, PR China
| | - Meng Zhu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, PR China
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Long Hai
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Liang Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Chen Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Pengfei Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Hua Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Song Wang
- Department of Neurosurgery, Tianjin First Central Hospital, Tianjin, 300192, PR China
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, PR China.
| |
Collapse
|
25
|
Wang Y, Zhao L, Han X, Wang Y, Mi J, Wang C, Sun D, Fu Y, Zhao X, Guo H, Wang Q. Saikosaponin A Inhibits Triple-Negative Breast Cancer Growth and Metastasis Through Downregulation of CXCR4. Front Oncol 2020; 9:1487. [PMID: 32047724 PMCID: PMC6997291 DOI: 10.3389/fonc.2019.01487] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/11/2019] [Indexed: 12/21/2022] Open
Abstract
Purpose: Due to a lack of recognized molecular targets for therapy, patients with triple-negative breast cancer (TNBC), unlike other subtypes of breast cancers, generally have not benefited from the advances made with targeted agents. The CXCR4/SDF-1 axis is involved in tumor growth and metastasis of TNBC. Therefore, down-regulation of the expression of CXCR4 in cancer cells is a potential therapeutic strategy for inhibiting primary tumor growth and metastasis of TNBC. In order to identify bioactive compounds that inhibit the expression of CXCR4 in traditional Chinese medicines, we investigated the capacity of saikosaponin A (SSA), one of the active ingredients isolated from Radix bupleuri, to affect CXCR4 expression and function in TNBC cells. Methods: Analyses of cell growth, migration, invasion, and protein expression were performed. Knockdowns by small interfering RNA (siRNA) and non-invasive bioluminescence were also used. Results: SSA reduced proliferation and colony formation of SUM149 and MDA-MB-231 cells. SSA inhibited migration and invasion of TNBC cells. For mice, SSA inhibited primary tumor growth and reduced lung metastasis of highly metastatic, triple-negative 4T1-luc cells. SSA inhibited CXCR4 expression but did not regulate CXCR7 expression in vitro and in vivo. The inhibitory effects on the migration and invasion of TNBC cells were reversed by down-regulation of CXCR4 expression. In addition, SSA inactivated the Akt/mTOR signaling pathway and inhibited MMP-9 and MMP-2 expression. Conclusions: The results show that SSA exerts an anti-TNBC effect through the inhibition of CXCR4 expression and thus has the potential to be a candidate therapeutic agent for TNBC patients.
Collapse
Affiliation(s)
- Ying Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Zhao
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Xianghui Han
- Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yahui Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinxia Mi
- Science and Technology Center, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Yunfei Fu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaodong Zhao
- Department of Pathology, National Shanghai Center for New Drug Safety Evaluation and Research, Shanghai, China
| | - Haidong Guo
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiangli Wang
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Lu W, He Z, Shi J, Wang Z, Wu W, Liu J, Kang H, Li F, Liang S. AMD3100 Attenuates Post-Traumatic Osteoarthritis by Maintaining Transforming Growth Factor-β1-Induced Expression of Tissue Inhibitor of Metalloproteinase-3 via the Phosphatidylinositol 3-Kinase/Akt Pathway. Front Pharmacol 2020; 10:1554. [PMID: 32038242 PMCID: PMC6987846 DOI: 10.3389/fphar.2019.01554] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/02/2019] [Indexed: 12/20/2022] Open
Abstract
AMD3100 is a small-molecule inhibitor of the C-X-C motif chemokine ligand 12/C-X-C chemokine receptor type 4 (CXCL12/CXCR4) axis, while its role in aggrecan metabolism is unclear. We hypothesized that the AMD3100 modulates the transforming growth factor-β1 (TGF-β1)-induced expression of tissue inhibitor of metalloproteinase-3 (TIMP-3) in chondrocytes. We evaluated expression of CXCL12/CXCR4 and TIMP-3 in the knee joints of rats with and without osteoarthritis (OA) by immunohistochemistry, immunofluorescence, Western blotting, and enzyme-linked immunosorbent assay (ELISA). The rats were divided into sham control, destabilization of the medial meniscus/AMD3100-treated (DMM/AMD3100-treated), and DMM/phosphate-buffered saline (PBS)-treated groups. After 6 weeks, the rats were euthanized and subjected to histological and immunohistochemical analyses. Also, interleukin (IL)-1-pretreated primary chondrocytes were cultured in the presence of empty control (−, −), CXCL12a (+,−), CXCL12a + small interfering RNA (siRNA) CXCR4 (+,+), or CXCL12a + siNC (+NC), and the expression levels of target markers were evaluated by Western blotting and real-time reverse transcription PCR (RT-PCR). The CXCL12/CXCR4 levels were higher, and the expression of TIMP-3 was lower, in the OA rats compared to the healthy control rats. The rats in the DMM/AMD3100-treated group revealed a markedly decreased immunological response and mild pathology. Treatment with CXCL12a increased expression of aggrecan and disintegrin and metalloproteinase with thrombospondin motifs-5 (ADAMTS-5) and suppressed that of TIMP-3 in IL-1-pretreated primary chondrocytes. TGF-β1 increased expression of TIMP-3, and this increase was reversed by CXCL12a via the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. Moreover, these effects were inhibited by the CXCR4 antagonist AMD3100 and the PI3K inhibitor LY303511. In conclusion, inhibition of the CXCL12a/CXCR4 signaling axis maintained TIMP-3 expression via the PI3K/Akt pathway. Our findings provide insight into the mechanism by which AMD3100 prevents OA.
Collapse
Affiliation(s)
- Weiwei Lu
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyi He
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Shi
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenggang Wang
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wu
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Liu
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Kang
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Li
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Liang
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Mousavi A. CXCL12/CXCR4 signal transduction in diseases and its molecular approaches in targeted-therapy. Immunol Lett 2019; 217:91-115. [PMID: 31747563 DOI: 10.1016/j.imlet.2019.11.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/01/2019] [Accepted: 11/15/2019] [Indexed: 02/08/2023]
Abstract
Chemokines are small molecules called "chemotactic cytokines" and regulate many processes like leukocyte trafficking, homing of immune cells, maturation, cytoskeletal rearrangement, physiology, migration during development, and host immune responses. These proteins bind to their corresponding 7-membrane G-protein-coupled receptors. Chemokines and their receptors are anti-inflammatory factors in autoimmune conditions, so consider as potential targets for neutralization in such diseases. They also express by cancer cells and function as angiogenic factors, and/or survival/growth factors that enhance tumor angiogenesis and development. Among chemokines, the CXCL12/CXCR4 axis has significantly been studied in numerous cancers and autoimmune diseases. CXCL12 is a homeostatic chemokine, which is acts as an anti-inflammatory chemokine during autoimmune inflammatory responses. In cancer cells, CXCL12 acts as an angiogenic, proliferative agent and regulates tumor cell apoptosis as well. CXCR4 has a role in leukocyte chemotaxis in inflammatory situations in numerous autoimmune diseases, as well as the high levels of CXCR4, observed in different types of human cancers. These findings suggest CXCL12/CXCR4 as a potential therapeutic target for therapy of autoimmune diseases and open a new approach to targeted-therapy of cancers by neutralizing CXCL12 and CXCR4. In this paper, we reviewed the current understanding of the role of the CXCL12/CXCR4 axis in disease pathology and cancer biology, and discuss its therapeutic implications in cancer and diseases.
Collapse
|
28
|
García-Cuesta EM, Santiago CA, Vallejo-Díaz J, Juarranz Y, Rodríguez-Frade JM, Mellado M. The Role of the CXCL12/CXCR4/ACKR3 Axis in Autoimmune Diseases. Front Endocrinol (Lausanne) 2019; 10:585. [PMID: 31507535 PMCID: PMC6718456 DOI: 10.3389/fendo.2019.00585] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/09/2019] [Indexed: 12/19/2022] Open
Abstract
Chemokine receptors are members of the G protein-coupled receptor superfamily. These receptors are intimately involved in cell movement, and thus play a critical role in several physiological and pathological situations that require the precise regulation of cell positioning. CXCR4 is one of the most studied chemokine receptors and is involved in many functions beyond leukocyte recruitment. During embryogenesis, it plays essential roles in vascular development, hematopoiesis, cardiogenesis, and nervous system organization. It has been also implicated in tumor progression and autoimmune diseases and, together with CD4, is one of the co-receptors used by the HIV-1 virus to infect immune cells. In contrast to other chemokine receptors that are characterized by ligand promiscuity, CXCR4 has a unique ligand-stromal cell-derived factor-1 (SDF1, CXCL12). However, this ligand also binds ACKR3, an atypical chemokine receptor that modulates CXCR4 functions and is overexpressed in multiple cancer types. The CXCL12/CXCR4/ACKR3 axis constitutes a potential therapeutic target for a wide variety of inflammatory diseases, not only by interfering with cell migration but also by modulating immune responses. Thus far, only one antagonist directed against the ligand-binding site of CXCR4, AMD3100, has demonstrated clinical relevance. Here, we review the role of this ligand and its receptors in different autoimmune diseases.
Collapse
Affiliation(s)
- Eva M. García-Cuesta
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - César A. Santiago
- Macromolecular X-Ray Crystallography Unit, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Jesús Vallejo-Díaz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Yasmina Juarranz
- Department Cell Biology, Research Institute Hospital 12 de Octubre (i+12), Complutense University of Madrid, Madrid, Spain
| | | | - Mario Mellado
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
- *Correspondence: Mario Mellado
| |
Collapse
|