1
|
Masrour M, Moeinafshar A, Poopak A, Razi S, Rezaei N. The role of CXC chemokines and receptors in breast cancer. Clin Exp Med 2025; 25:128. [PMID: 40278951 PMCID: PMC12031896 DOI: 10.1007/s10238-025-01662-7] [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: 09/17/2024] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
CXC chemokines are a class of cytokines possessing chemotactic properties. Studies indicate that CXC chemokines exhibit dysregulation in miscellaneous cancer categories and are significantly associated with the advancement of tumors. Breast cancer is a commonly diagnosed and fatal cancer among the female population. Breast cancer pathogenesis and progression involve various mechanisms, including invasion, metastasis, angiogenesis, and inflammation. Chemokines and their receptors are involved in all of these processes. The CXC chemokine receptors (CXCRs) and their related ligands have attracted considerable attention due to their multifaceted functions in facilitating and controlling tumor proliferation. CXCRs are expressed by both cancer cells and immune cells, and they play a crucial role in regulating the tumor microenvironment and the immune response. This review aims to assess the potential of CXCRs and CXC chemokines as therapeutic targets or biomarkers for personalized therapy. Additionally, it provides an overview of the current understanding of the expression, function, and prognostic relevance of CXCRs in breast cancer. Furthermore, the challenges and potential prospects pertaining to CXCR investigation in breast cancer are deliberated.
Collapse
Affiliation(s)
- Mahdi Masrour
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Center for Orthopedic Trans-Disciplinary Applied Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Aysan Moeinafshar
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amirhossein Poopak
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific and Education Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
2
|
Yin Q, Yang Y, Qu Z, Ouchari M, Zeng L, Tang S, Zheng J, Zhang S, Ma H, Chen Y, Wang J, Shi L, Zheng X. Unraveling the Multifaceted Roles of Atypical Chemokine Receptors in Breast Cancer. J Interferon Cytokine Res 2025; 45:43-52. [PMID: 39526942 DOI: 10.1089/jir.2024.0186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Breast cancer (BC) remains one of the most prevalent and deadly malignancies among women globally. A deeper understanding of the molecular mechanisms driving BC progression and metastasis is essential for the development of effective therapeutic strategies. While traditional chemokine receptors are well known for their roles in immune cell migration and positioning, atypical chemokine receptors (ACKRs) have recently gained attention as key modulators in cancer-related processes. Unlike conventional receptors, ACKRs-comprising ACKR1, ACKR2, ACKR3, and ACKR4-primarily function by scavenging chemokines, regulating their availability, and modulating receptor signaling in a ligand-independent manner. This review aims to elucidate the roles of ACKRs in BC, focusing on their influence on the tumor microenvironment (TME), cancer cell proliferation, survival, metastasis, and angiogenesis. Additionally, we will explore the potential of ACKRs as diagnostic and prognostic markers and assess their viability as therapeutic targets. By synthesizing recent research findings and highlighting future research directions, this review seeks to provide a comprehensive understanding of the significance of ACKRs in BC and underscore the need for continued investigation into their therapeutic potential.
Collapse
Affiliation(s)
- Qinan Yin
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Yisha Yang
- Department of Finance and Management, Henan Vocational College of Agriculture at Luoyang, Luoyang, China
| | - Zhifeng Qu
- Radiology Department, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Mouna Ouchari
- Laboratory of Translational Redox Medicine (TRx Med), University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Li Zeng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Siya Tang
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Jiayu Zheng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Shunshun Zhang
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Haodi Ma
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Youyou Chen
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Jiayi Wang
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Linlin Shi
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment, Henan Key Laboratory of Cancer Epigenetics, College of Basic Medicine and Forensic Medicine, Cancer Hospital, The First Affiliated Hospital (College of Clinical Medicine) of Henan University of Science and Technology, Luoyang, China
| | - Xuewei Zheng
- Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
3
|
Zhao C, Taliento AE, Belkin EM, Fearns R, Lerou PH, Ai X, Bai Y. Infant RSV infection desensitizes β2-adrenergic receptor via CXCL11-CXCR7 signaling in airway smooth muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632772. [PMID: 39868223 PMCID: PMC11761401 DOI: 10.1101/2025.01.13.632772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Rationale Airflow obstruction refractory to β2 adrenergic receptor (β2AR) agonists is an important clinical feature of infant respiratory syncytial virus (RSV) bronchiolitis, with limited treatment options. This resistance is often linked to poor drug delivery and potential viral infection of airway smooth muscle cells (ASMCs). Whether RSV inflammation causes β2AR desensitization in infant ASMCs is unknown. Objectives To investigate the interaction of RSV inflammation with the β2AR signaling pathway in infant ASMCs. Methods Infant precision-cut lung slices (PCLSs) and mouse pup models of RSV infection were subjected to airway physiological assays. Virus-free, conditioned media from RSV-infected infant bronchial epithelial cells in air-liquid interface (ALI) culture and nasopharyngeal aspirates (NPA) from infants with severe RSV bronchiolitis were collected and applied to infant PCLSs and ASMCs. Cytokines in these samples were profiled and assessed for the effects on β2AR expression, cell surface distribution, and relaxant function in ASMCs. Measurements and Main Results Conditioned media and NPA induced similar resistance to β2AR agonists in ASMCs as RSV infection. Cytokine profiling identified CXCL11 as one of the most elevated signals following RSV infection. CXCL11 activated its receptor CXCR7 in a complex with β2AR in ASMCs to promote β2AR phosphorylation, internalization, and degradation. Blockade of CXCR7 partially restored airway relaxation in response to β2AR agonists in infant PCLSs and mouse pup models of RSV infection. Conclusions The CXCL11-CXCR7 pathway plays a critical role in β2AR desensitization in ASMCs during RSV infection and represents a potential therapeutic target in alleviating airflow obstruction in infant RSV bronchiolitis.
Collapse
|
4
|
Gupta KB, Gao J, Li X, Thangaraju M, Panda SS, Lokeshwar BL. Cytotoxic Autophagy: A Novel Treatment Paradigm against Breast Cancer Using Oleanolic Acid and Ursolic Acid. Cancers (Basel) 2024; 16:3367. [PMID: 39409987 PMCID: PMC11476055 DOI: 10.3390/cancers16193367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Oleanolic acid (OA) and Ursolic acid (UA) are bioactive triterpenoids. Reported activities vary with the dose used for testing their activities in vitro. Studies using doses of ≥20 µM showed apoptosis activities in cancer cells. However, reported drug levels in circulation achieved by oral administration of UA and OA are ≤2 µM, thus limiting their use for treatment or delivering a combination treatment. MATERIALS AND METHODS The present report demonstrates the efficacy of OA, UA, and OA + UA on tumor cell-specific cytotoxicity at low doses (5 µM to 10 µM) in breast cancer (BrCa) cell lines MCF7 and MDA-MB231. RESULTS The data show that both OA and UA killed BrCa cells at low doses, but were significantly less toxic to MCF-12A, a non-tumorigenic cell line. Moreover, OA + UA at ≤10 µM was lethal to BrCa cells. Mechanistic studies unraveled the significant absence of apoptosis, but their cytotoxicity was due to the induction of excessive autophagy at a OA + UA dose of 5 µM each. A link to drug-induced cytotoxic autophagy was established by demonstrating a lack of their cytotoxicity by silencing the autophagy-targeting genes (ATGs), which prevented OA-, UA-, or OA + UA-induced cell death. Further, UA or OA + UA treatment of BrCa cells caused an inhibition of PI3 kinase-mediated phosphorylation of Akt/mTOR, the key pathways that regulate cancer cell survival, metabolism, and proliferation. DISCUSSION Combinations of a PI3K inhibitor (LY294002) with OA, UA, or OA + UA synergistically inhibited BrCa cell survival. Therefore, the dominance of cytotoxic autophagy by inhibiting PI3K-mediated autophagy may be the primary mechanism of PTT-induced anticancer activity in BrCa cells. CONCLUSION These results suggest it would be worthwhile testing combined OA and UA in clinical settings.
Collapse
Affiliation(s)
- Kunj Bihari Gupta
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA; (K.B.G.); (J.G.); (X.L.)
| | - Jie Gao
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA; (K.B.G.); (J.G.); (X.L.)
- Department of Clinical and Diagnostic Science, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Xin Li
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA; (K.B.G.); (J.G.); (X.L.)
- The Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Muthusamy Thangaraju
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.T.); (S.S.P.)
| | - Siva S. Panda
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.T.); (S.S.P.)
- Department of Chemistry and Biochemistry, College of Science and Mathematics, Augusta University, Augusta, GA 30912, USA
| | - Bal L. Lokeshwar
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA; (K.B.G.); (J.G.); (X.L.)
| |
Collapse
|
5
|
Zhao D, Guo Y, Wei H, Jia X, Zhi Y, He G, Nie W, Huang L, Wang P, Laster KV, Liu Z, Wang J, Lee MH, Dong Z, Liu K. Multi-omics characterization of esophageal squamous cell carcinoma identifies molecular subtypes and therapeutic targets. JCI Insight 2024; 9:e171916. [PMID: 38652547 PMCID: PMC11141925 DOI: 10.1172/jci.insight.171916] [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: 05/02/2023] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the predominant form of esophageal cancer and is characterized by an unfavorable prognosis. To elucidate the distinct molecular alterations in ESCC and investigate therapeutic targets, we performed a comprehensive analysis of transcriptomics, proteomics, and phosphoproteomics data derived from 60 paired treatment-naive ESCC and adjacent nontumor tissue samples. Additionally, we conducted a correlation analysis to describe the regulatory relationship between transcriptomic and proteomic processes, revealing alterations in key metabolic pathways. Unsupervised clustering analysis of the proteomics data stratified patients with ESCC into 3 subtypes with different molecular characteristics and clinical outcomes. Notably, subtype III exhibited the worst prognosis and enrichment in proteins associated with malignant processes, including glycolysis and DNA repair pathways. Furthermore, translocase of inner mitochondrial membrane domain containing 1 (TIMMDC1) was validated as a potential prognostic molecule for ESCC. Moreover, integrated kinase-substrate network analysis using the phosphoproteome nominated candidate kinases as potential targets. In vitro and in vivo experiments further confirmed casein kinase II subunit α (CSNK2A1) as a potential kinase target for ESCC. These underlying data represent a valuable resource for researchers that may provide better insights into the biology and treatment of ESCC.
Collapse
Affiliation(s)
- Dengyun Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, China
| | - Yaping Guo
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Huifang Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Xuechao Jia
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Yafei Zhi
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Guiliang He
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Wenna Nie
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Limeng Huang
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Penglei Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | | | - Zhicai Liu
- Linzhou Cancer Hospital, Anyang, Henan, China
| | - Jinwu Wang
- Linzhou Cancer Hospital, Anyang, Henan, China
| | - Mee-Hyun Lee
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- College of Korean Medicine, Dongshin University, Naju, Jeonnam, Republic of Korea
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, Henan, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Naser IH, Hamza AA, Alhili A, Faisal AN, Ali MS, Kadhim NA, Suliman M, Alshahrani MY, Alawadi A. Atypical chemokine receptor 4 (ACKR4/CCX-CKR): A comprehensive exploration across physiological and pathological landscapes in contemporary research. Cell Biochem Funct 2024; 42:e4009. [PMID: 38597217 DOI: 10.1002/cbf.4009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/02/2024] [Accepted: 03/31/2024] [Indexed: 04/11/2024]
Abstract
Atypical chemokine receptor 4 (ACKR4), also known as CCX-CKR, is a member of the chemokine receptor family that lacks typical G protein signaling activity. Instead, ACKR4 functions as a scavenger receptor that can bind and internalize a wide range of chemokines, influencing their availability and activity in the body. ACKR4 is involved in various physiological processes, such as immune cell trafficking and the development of thymus, spleen, and lymph nodes. Moreover, ACKR4 has been implicated in several pathological conditions, including cancer, heart and lung diseases. In cancer, ACKR4 plays a complex role, acting as a tumor suppressor or promoter depending on the type of cancer and the stage of the disease. For instance, ACKR4 may inhibit the growth and metastasis of breast cancer, but it may also promote the progression of hepatocellular carcinoma and gastric cancer. In inflammatory situations, ACKR4 has been found to modulate the recruitment and activation of immune cells, contributing to the pathogenesis of diseases such as myocardial infraction and pulmonary sarcoidosis. The study of ACKR4 is still ongoing, and further research is needed to fully understand its role in different physiological and pathological contexts. Nonetheless, ACKR4 represents a promising target for the development of novel therapeutic strategies for various diseases.
Collapse
Affiliation(s)
- Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University College, Hillah, Babil, Iraq
| | - Asia Ali Hamza
- Department of Pharmaceutics, Faculty of pharmacy, University of Al-Ameed, Karbala, Iraq
| | - Ahmed Alhili
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | | | | | | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Ahmed Alawadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
7
|
Samus M, Rot A. Atypical chemokine receptors in cancer. Cytokine 2024; 176:156504. [PMID: 38266462 DOI: 10.1016/j.cyto.2024.156504] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/28/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Atypical chemokine receptors (ACKRs) are a group of seven-transmembrane spanning serpentine receptors that are structurally homologous to classical G-protein-coupled receptors and bind cognate chemokines with high affinities but do not signal via G-proteins or mediate cell migration. However, ACKRs efficiently modify the availability and function of chemokines in defined microanatomical environments, can signal via intracellular effectors other than G-proteins, and play complex roles in physiology and disease, including in cancer. In this review, we summarize the findings on the diverse contributions of individual ACKRs to cancer development, progression, and tumor-host interactions. We discuss how changes in ACKR expression within tumor affect cancer growth, tumor vascularization, leukocyte infiltration, and metastasis formation, ultimately resulting in differential disease outcomes. Across many studies, ACKR3 expression was shown to support tumor growth and dissemination, whereas ACKR1, ACKR2, and ACKR4 in tumors were more likely to contribute to tumor suppression. With few notable exceptions, the insights on molecular and cellular mechanisms of ACKRs activities in cancer remain sparse, and the intricacies of their involvement are not fully appreciated. This is particularly true for ACKR1, ACKR2 and ACKR4. A better understanding of how ACKR expression and functions impact cancer should pave the way for their future targeting by new and effective therapies.
Collapse
Affiliation(s)
- Maryna Samus
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Antal Rot
- Centre for Microvascular Research, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK; Institute for Cardiovascular Prevention, Ludwig-Maximilians University, Munich 80336, Germany.
| |
Collapse
|
8
|
Klaver D, Gander H, Frena B, Amato M, Thurnher M. Crosstalk between purinergic receptor P2Y 11 and chemokine receptor CXCR7 is regulated by CXCR4 in human macrophages. Cell Mol Life Sci 2024; 81:132. [PMID: 38472446 DOI: 10.1007/s00018-024-05158-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 03/14/2024]
Abstract
P2Y11 is a G protein-coupled ATP receptor that activates IL-1 receptor (IL-1R) in a cyclic AMP dependent manner. In human macrophages, P2Y11/IL-1R crosstalk with CCL20 as a prime target is controlled by phosphodiesterase 4 (PDE4), which mediates breakdown of cyclic AMP. Here, we used gene expression analysis to identify activation of CXCR4 and CXCR7 as a hallmark of P2Y11 signaling. We found that PDE4 inhibition with rolipram boosts P2Y11/IL-1R-induced upregulation of CXCR7 expression and CCL20 production in an epidermal growth factor receptor dependent manner. Using an astrocytoma cell line, naturally expressing CXCR7 but lacking CXCR4, P2Y11/IL-1R activation effectively induced and CXCR7 agonist TC14012 enhanced CCL20 production even in the absence of PDE4 inhibition. Moreover, CXCR7 depletion by RNA interference suppressed CCL20 production. In macrophages, the simultaneous activation of P2Y11 and CXCR7 by their respective agonists was sufficient to induce CCL20 production with no need of PDE4 inhibition, as CXCR7 activation increased its own and eliminated CXCR4 expression. Finally, analysis of multiple CCL chemokines in the macrophage secretome revealed that CXCR4 inactivation and CXCR7 activation selectively enhanced P2Y11/IL-1R-mediated secretion of CCL20. Altogether, our data establish CXCR7 as an integral component of the P2Y11/IL-1R-initiated signaling cascade and CXCR4-associated PDE4 as a regulatory checkpoint.
Collapse
Affiliation(s)
- Dominik Klaver
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innrain 66a, Innsbruck, 6020, Austria
| | - Hubert Gander
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innrain 66a, Innsbruck, 6020, Austria
| | - Beatrice Frena
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innrain 66a, Innsbruck, 6020, Austria
| | - Marco Amato
- Central Institute for Blood Transfusion & Department of Immunology (ZIB), Tirol Kliniken GmbH, Innsbruck, Austria
| | - Martin Thurnher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innrain 66a, Innsbruck, 6020, Austria.
| |
Collapse
|
9
|
Gritsina G, Fong KW, Lu X, Lin Z, Xie W, Agarwal S, Lin D, Schiltz GE, Beltran H, Corey E, Morrissey C, Wang Y, Zhao JC, Hussain M, Yu J. Chemokine receptor CXCR7 activates Aurora Kinase A and promotes neuroendocrine prostate cancer growth. J Clin Invest 2023; 133:e166248. [PMID: 37347559 PMCID: PMC10378179 DOI: 10.1172/jci166248] [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: 10/13/2022] [Accepted: 06/15/2023] [Indexed: 06/24/2023] Open
Abstract
CXCR7 is an atypical chemokine receptor that recruits β-arrestin (ARRB2) and internalizes into clathrin-coated intracellular vesicles where the complex acts as a scaffold for cytoplasmic kinase assembly and signal transduction. Here, we report that CXCR7 was elevated in the majority of prostate cancer (PCa) cases with neuroendocrine features (NEPC). CXCR7 markedly induced mitotic spindle and cell cycle gene expression. Mechanistically, we identified Aurora Kinase A (AURKA), a key regulator of mitosis, as a novel target that was bound and activated by the CXCR7-ARRB2 complex. CXCR7 interacted with proteins associated with microtubules and golgi, and, as such, the CXCR7-ARRB2-containing vesicles trafficked along the microtubules to the pericentrosomal golgi apparatus, where the complex interacted with AURKA. Accordingly, CXCR7 promoted PCa cell proliferation and tumor growth, which was mitigated by AURKA inhibition. In summary, our study reveals a critical role of CXCR7-ARRB2 in interacting and activating AURKA, which can be targeted by AURKA inhibitors to benefit a subset of patients with NEPC.
Collapse
Affiliation(s)
- Galina Gritsina
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ka-wing Fong
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Xiaodong Lu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Zhuoyuan Lin
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Urology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wanqing Xie
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shivani Agarwal
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Dong Lin
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gary E. Schiltz
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Chemistry, Northwestern University, Evanston, Illinois, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington, USA
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan C. Zhao
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Human Genetics and
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Maha Hussain
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jindan Yu
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Human Genetics and
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
10
|
Blakely B, Shin S, Jin K. Overview of the therapeutic strategies for ER positive breast cancer. Biochem Pharmacol 2023; 212:115552. [PMID: 37068524 PMCID: PMC10394654 DOI: 10.1016/j.bcp.2023.115552] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 04/19/2023]
Abstract
Estrogen Receptor is the driving transcription factor in about 75% of all breast cancers, which is the target of endocrine therapies, but drug resistance is a common clinical problem. ESR1 point mutations at the ligand binding domain are frequently identified in metastatic tumor and ctDNA (Circulating tumor DNA) derived from ER positive breast cancer patients with endocrine therapies. Although endocrine therapy and CDK4/6 inhibitor therapy have demonstrated preclinical and clinical benefits for breast cancer, the development of resistance remains a significant challenge and the detailed mechanisms, and potential therapeutic targets in advanced breast cancer yet to be revealed. Since a crosstalk between tumor and tumor microenvironment (TME) plays an important role to grow tumor and metastasis, this effect could serve as key regulators in the resistance of endocrine therapy and the transition of breast cancer cells to metastasis. In this article, we have reviewed recent progress in endocrine therapy and the contribution of TME to ER positive breast cancer.
Collapse
Affiliation(s)
- Brianna Blakely
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Science, Albany, NY, United States
| | - Seobum Shin
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Science, Albany, NY, United States
| | - Kideok Jin
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Science, Albany, NY, United States.
| |
Collapse
|
11
|
Zeitz C, Roger JE, Audo I, Michiels C, Sánchez-Farías N, Varin J, Frederiksen H, Wilmet B, Callebert J, Gimenez ML, Bouzidi N, Blond F, Guilllonneau X, Fouquet S, Léveillard T, Smirnov V, Vincent A, Héon E, Sahel JA, Kloeckener-Gruissem B, Sennlaub F, Morgans CW, Duvoisin RM, Tkatchenko AV, Picaud S. Shedding light on myopia by studying complete congenital stationary night blindness. Prog Retin Eye Res 2023; 93:101155. [PMID: 36669906 DOI: 10.1016/j.preteyeres.2022.101155] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/20/2023]
Abstract
Myopia is the most common eye disorder, caused by heterogeneous genetic and environmental factors. Rare progressive and stationary inherited retinal disorders are often associated with high myopia. Genes implicated in myopia encode proteins involved in a variety of biological processes including eye morphogenesis, extracellular matrix organization, visual perception, circadian rhythms, and retinal signaling. Differentially expressed genes (DEGs) identified in animal models mimicking myopia are helpful in suggesting candidate genes implicated in human myopia. Complete congenital stationary night blindness (cCSNB) in humans and animal models represents an ON-bipolar cell signal transmission defect and is also associated with high myopia. Thus, it represents also an interesting model to identify myopia-related genes, as well as disease mechanisms. While the origin of night blindness is molecularly well established, further research is needed to elucidate the mechanisms of myopia development in subjects with cCSNB. Using whole transcriptome analysis on three different mouse models of cCSNB (in Gpr179-/-, Lrit3-/- and Grm6-/-), we identified novel actors of the retinal signaling cascade, which are also novel candidate genes for myopia. Meta-analysis of our transcriptomic data with published transcriptomic databases and genome-wide association studies from myopia cases led us to propose new biological/cellular processes/mechanisms potentially at the origin of myopia in cCSNB subjects. The results provide a foundation to guide the development of pharmacological myopia therapies.
Collapse
Affiliation(s)
- Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.
| | - Jérome E Roger
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, Saclay, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France
| | | | | | - Juliette Varin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Helen Frederiksen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, APHP, Paris, France
| | | | - Nassima Bouzidi
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Frederic Blond
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Stéphane Fouquet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Vasily Smirnov
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Ajoy Vincent
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, Toronto, ON, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Elise Héon
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, Toronto, ON, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France; Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Catherine W Morgans
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Robert M Duvoisin
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Andrei V Tkatchenko
- Oujiang Laboratory, Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health, Wenzhou, China; Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University, New York, NY, USA
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
12
|
Vacchini A, Maffioli E, Di Silvestre D, Cancellieri C, Milanesi S, Nonnis S, Badanai S, Mauri P, Negri A, Locati M, Tedeschi G, Borroni EM. Phosphoproteomic mapping of CCR5 and ACKR2 signaling properties. Front Mol Biosci 2022; 9:1060555. [PMID: 36483536 PMCID: PMC9723398 DOI: 10.3389/fmolb.2022.1060555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 10/31/2022] [Indexed: 07/25/2024] Open
Abstract
ACKR2 is an atypical chemokine receptor which is structurally uncoupled from G proteins and is unable to activate signaling pathways used by conventional chemokine receptors to promote cell migration. Nonetheless, ACKR2 regulates inflammatory and immune responses by shaping chemokine gradients in tissues via scavenging inflammatory chemokines. To investigate the signaling pathways downstream to ACKR2, a quantitative SILAC-based phosphoproteomic analysis coupled with a systems biology approach with network analysis, was carried out on a HEK293 cell model expressing either ACKR2 or its conventional counterpart CCR5. The model was stimulated with the common agonist CCL3L1 for short (3 min) and long (30 min) durations. As expected, many of the identified proteins are known to participate in conventional signal transduction pathways and in the regulation of cytoskeleton dynamics. However, our analyses revealed unique phosphorylation and network signatures, suggesting roles for ACKR2 other than its scavenger activity. In conclusion, the mapping of phosphorylation events at a holistic level indicated that conventional and atypical chemokine receptors differ in signaling properties. This provides an unprecedented level of detail in chemokine receptor signaling and identifying potential targets for the regulation of ACKR2 and CCR5 function.
Collapse
Affiliation(s)
- Alessandro Vacchini
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Segrate, Italy
| | - Elisa Maffioli
- Department of Veterinary Medicine and Animal Science, University of Milan, Lodi, Italy
| | - Dario Di Silvestre
- Institute of Technologies in Biomedicine, National Research Council (ITB-CNR), Milan, Italy
| | | | - Samantha Milanesi
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Segrate, Italy
| | - Simona Nonnis
- Department of Veterinary Medicine and Animal Science, University of Milan, Lodi, Italy
| | | | | | - Armando Negri
- Department of Veterinary Medicine and Animal Science, University of Milan, Lodi, Italy
| | - Massimo Locati
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Segrate, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science, University of Milan, Lodi, Italy
- CIMAINA, Milan, Italy
| | - Elena Monica Borroni
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Segrate, Italy
| |
Collapse
|
13
|
A new obligate CXCL4-CXCL12 heterodimer for studying chemokine heterodimer activities and mechanisms. Sci Rep 2022; 12:17204. [PMID: 36229490 PMCID: PMC9561612 DOI: 10.1038/s41598-022-21651-0] [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: 05/24/2022] [Accepted: 09/29/2022] [Indexed: 01/06/2023] Open
Abstract
Chemokines form a family of proteins with critical roles in many biological processes in health and disease conditions, including cardiovascular, autoimmune diseases, infections, and cancer. Many chemokines engage in heterophilic interactions to form heterodimers, leading to synergistic activity enhancement or reduction dependent on the nature of heterodimer-forming chemokines. In mixtures, different chemokine species with diverse activities coexist in dynamic equilibrium, leading to the observation of their combined response in biological assays. To overcome this problem, we produced a non-dissociating CXCL4-CXCL12 chemokine heterodimer OHD4-12 as a new tool for studying the biological activities and mechanisms of chemokine heterodimers in biological environments. Using the OHD4-12, we show that the CXCL4-CXCL12 chemokine heterodimer inhibits the CXCL12-driven migration of triple-negative MDA-MB-231 breast cancer cells. We also show that the CXCL4-CXCL12 chemokine heterodimer binds and activates the CXCR4 receptor.
Collapse
|
14
|
Aamna B, Kumar Dan A, Sahu R, Behera SK, Parida S. Deciphering the signaling mechanisms of β-arrestin1 and β-arrestin2 in regulation of cancer cell cycle and metastasis. J Cell Physiol 2022; 237:3717-3733. [PMID: 35908197 DOI: 10.1002/jcp.30847] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/22/2022] [Accepted: 07/18/2022] [Indexed: 11/05/2022]
Abstract
β-Arrestins are ubiquitously expressed intracellular proteins with many functions which interact directly and indirectly with a wide number of cellular partners and mediate downstream signaling. Originally, β-arrestins were identified for their contribution to GPCR desensitization to agonist-mediated activation, followed by receptor endocytosis and ubiquitylation. However, current investigations have now recognized that in addition to GPCR arresting (hence the name arrestin). β-Arrestins are adaptor proteins that control the recruitment, activation, and scaffolding of numerous cytoplasmic signaling complexes and assist in G-protein receptor signaling, thus bringing them into close proximity. They have participated in various cellular processes such as cell proliferation, migration, apoptosis, and transcription via canonical and noncanonical pathways. Despite their significant recognition in several physiological processes, these activities are also involved in the onset and progression of various cancers. This review delivers a concise overview of the role of β-arrestins with a primary emphasis on the signaling processes which underlie the mechanism of β-arrestins in the onset of cancer. Understanding these processes has important implications for understanding the therapeutic intervention and treatment of cancer in the future.
Collapse
Affiliation(s)
- Bari Aamna
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed to be University), Bhubaneswar, Odisha, India
| | - Aritra Kumar Dan
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed to be University), Bhubaneswar, Odisha, India
| | - Raghaba Sahu
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Santosh Kumar Behera
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, India
| | - Sagarika Parida
- Department of Botany, Centurion University of Technology and Management, Odisha, India
| |
Collapse
|
15
|
Masih M, Agarwal S, Kaur R, Gautam PK. Role of chemokines in breast cancer. Cytokine 2022; 155:155909. [PMID: 35597171 DOI: 10.1016/j.cyto.2022.155909] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 05/02/2022] [Accepted: 05/09/2022] [Indexed: 02/06/2023]
Abstract
Chemokines belong to a family of chemoattractant cytokines and are well known to have an essential role in various cancer aetiologies. Multiplesubsets of immune cells are recruited and enrolled into the tumor microenvironment through interactions between chemokines and their specific receptors. These populations and their interactions have a distinct impact on tumor growth, progression, and treatment outcomes. While it is clear that many chemokines and their cognate receptors can be detected in breast and other cancers, the role of each chemokine and receptor has yet to be determined. This review focuses on the main chemokines that play a crucial role in the tumor microenvironment, emphasizing breast cancer. We have also discussed the techniques used to identify the chemokines and their future implication in the early diagnosis of cancer. In-depth knowledge of chemokines and their role in breast cancer progression can provide specific targets for breast cancer biotherapy.
Collapse
Affiliation(s)
- Marilyn Masih
- Department of Biochemistry, AIIMS, New Delhi -110029, India.
| | - Sonam Agarwal
- Department of Biochemistry, AIIMS, New Delhi -110029, India.
| | - Rupinder Kaur
- Department of Biochemistry, AIIMS, New Delhi -110029, India.
| | | |
Collapse
|
16
|
Cheng H, Guo P, Su T, Jiang C, Zhu Z, Wei W, Zhang L, Wang Q. G protein-coupled receptor kinase type 2 and β-arrestin2: Key players in immune cell functions and inflammation. Cell Signal 2022; 95:110337. [PMID: 35461901 DOI: 10.1016/j.cellsig.2022.110337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/15/2022] [Accepted: 04/15/2022] [Indexed: 02/07/2023]
|
17
|
Joo M, Heo JB, Kim S, Kim N, Jeon HJ, An Y, Song GY, Kim JM, Lee HJ. Decursin inhibits tumor progression in head and neck squamous cell carcinoma by downregulating CXCR7 expression in vitro. Oncol Rep 2022; 47:39. [PMID: 34958113 PMCID: PMC8759107 DOI: 10.3892/or.2021.8250] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/06/2021] [Indexed: 11/06/2022] Open
Abstract
CXC chemokine receptor 7 (CXCR7) is frequently overexpressed in cancer and plays a significant role in tumor growth and metastasis. Consequently, inhibition of CXCR7 is important for treatment strategies. However, little is known concerning the biological role of CXCR7 and its underlying mechanisms in head and neck squamous cell carcinoma (HNSCC). The present study investigated the role of CXCR7 in HNSCC, as well as the effects of decursin, a pyranocoumarin compound isolated from Angelica gigas Nakai, on CXCR7 and its downstream signaling. Expression levels of CXCR7 in HNSCC cells were examined using flow cytometry, reverse transcriptase PCR, western blot analysis, and immunofluorescence. The effects of CXCR7 on cell proliferation, migration, and invasion were studied using CCK‑8, gap closure, and transwell assays. The results revealed that decursin significantly reduced CXCR7 expression and inhibited cell proliferation, migration, and invasion of human HNSCC cell lines. In addition, decursin induced G0/G1 cell cycle arrest in CXCR7‑overexpressing cells and decreased the levels of cyclin A, cyclin E, and CDK2. Furthermore, CXCR7 promoted cancer progression via the STAT3/c‑Myc pathway in HNSCC; suppression of CXCR7 with decursin prevented this effect. These results suggest that CXCR7 promotes cancer progression through the STAT3/c‑Myc pathway and that the natural compound decursin targets CXCR7 and may be valuable in the treatment of HNSCC.
Collapse
Affiliation(s)
- Mina Joo
- Department of Medical Science, College of Pharmacy, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jong Beom Heo
- College of Pharmacy, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Solbi Kim
- Department of Medical Science, College of Pharmacy, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Nayoung Kim
- Department of Medical Science, College of Pharmacy, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Heung Jin Jeon
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| | - Yueun An
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| | - Gyu-Yong Song
- College of Pharmacy, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Jin-Man Kim
- Department of Pathology, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| | - Hyo Jin Lee
- Infection Control Convergence Research Center, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
- Department of Internal Medicine, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| |
Collapse
|
18
|
TLR2 and TLR4 Signaling Pathways and Gastric Cancer: Insights from Transcriptomics and Sample Validation. IRANIAN BIOMEDICAL JOURNAL 2022; 26:36-43. [PMID: 34773930 PMCID: PMC8784901 DOI: 10.52547/ibj.26.1.36] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Pattern recognition receptors, especially toll-like receptors (TLRs), as the first line of defense for pathogen detection, were found to be associated with H.¬ pylori infection and gastric cancer (GC). However, the expression levels of TLRs, i.e. TLR2 and TLR4, as the main receptors sensed by H.¬ pylori, still remain largely ambiguous. We aimed to investigate the patterns of key transcripts of TLR2 and TLR4 in 100 GC transcriptome data. Additionally, we evaluated TLR2 and TLR4 gene expressions in gastric biopsies of Iranian GC patients, in order to validate RNA-seq outputs. Methods For this study, 100 runs of GC samples and controls were processed and analyzed using map read to reference. Differential gene expression method was used to distinguish between GC and normal samples in the expression of TLRs and other innate immune molecules. Also, using qRT-PCR assay, transcripts of TLRs molecules for 15 GC and 15 control samples were analyzed based on the analysis of variance and least significant differences. Results The results clearly showed that all signaling pathways molecules of TLR4, especially TLR4 (p = 0.019), NF-κB (p ¬= 0.047), IL-1β (p = 0.0096), and TNF-α (p = 0.048), were upregulated in a cancerous condition in different parts and at various stages of GC. Conclusion Our findings suggested that molecules involved in inflammation, including TLR4 and its related pro-inflammatory cytokines, may be responsible for the development and progression of GC. Accordingly, the control of H. pylori infection reduces inflammation in the gastric system and can play an important role in preventing gastrointestinal disorders.
Collapse
|
19
|
Endothelial ACKR3 drives atherosclerosis by promoting immune cell adhesion to vascular endothelium. Basic Res Cardiol 2022; 117:30. [PMID: 35674847 PMCID: PMC9177477 DOI: 10.1007/s00395-022-00937-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 01/31/2023]
Abstract
Atherosclerosis is the foundation of potentially fatal cardiovascular diseases and it is characterized by plaque formation in large arteries. Current treatments aimed at reducing atherosclerotic risk factors still allow room for a large residual risk; therefore, novel therapeutic candidates targeting inflammation are needed. The endothelium is the starting point of vascular inflammation underlying atherosclerosis and we could previously demonstrate that the chemokine axis CXCL12-CXCR4 plays an important role in disease development. However, the role of ACKR3, the alternative and higher affinity receptor for CXCL12 remained to be elucidated. We studied the role of arterial ACKR3 in atherosclerosis using western diet-fed Apoe-/- mice lacking Ackr3 in arterial endothelial as well as smooth muscle cells. We show for the first time that arterial endothelial deficiency of ACKR3 attenuates atherosclerosis as a result of diminished arterial adhesion as well as invasion of immune cells. ACKR3 silencing in inflamed human coronary artery endothelial cells decreased adhesion molecule expression, establishing an initial human validation of ACKR3's role in endothelial adhesion. Concomitantly, ACKR3 silencing downregulated key mediators in the MAPK pathway, such as ERK1/2, as well as the phosphorylation of the NF-kB p65 subunit. Endothelial cells in atherosclerotic lesions also revealed decreased phospho-NF-kB p65 expression in ACKR3-deficient mice. Lack of smooth muscle cell-specific as well as hematopoietic ACKR3 did not impact atherosclerosis in mice. Collectively, our findings indicate that arterial endothelial ACKR3 fuels atherosclerosis by mediating endothelium-immune cell adhesion, most likely through inflammatory MAPK and NF-kB pathways.
Collapse
|
20
|
Cxcl10 chemokine induces migration of ING4-deficient breast cancer cells via a novel crosstalk mechanism between the Cxcr3 and Egfr receptors. Mol Cell Biol 2021; 42:e0038221. [PMID: 34871062 DOI: 10.1128/mcb.00382-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The chemokine Cxcl10 has been associated with poor prognosis in breast cancer, but the mechanism is not well understood. Our previous study have shown that CXCL10 was repressed by the ING4 tumor suppressor, suggesting a potential inverse functional relationship. We thus investigated a role for Cxcl10 in the context of ING4 deficiencies in breast cancer. We first analyzed public gene expression datasets and found that patients with CXCL10-high/ING4-low expressing tumors had significantly reduced disease-free survival in breast cancer. In vitro, Cxcl10 induced migration of ING4-deleted breast cancer cells, but not of ING4-intact cells. Using inhibitors, we found that Cxcl10-induced migration of ING4-deleted cells required Cxcr3, Egfr, and the Gβγ subunits downstream of Cxcr3, but not Gαi. Immunofluorescent imaging showed that Cxcl10 induced early transient colocalization between Cxcr3 and Egfr in both ING4-intact and ING4-deleted cells, which recurred only in ING4-deleted cells. A peptide agent that binds to the internal juxtamembrane domain of Egfr inhibited Cxcr3/Egfr colocalization and cell migration. Taken together, these results presented a novel mechanism of Cxcl10 that elicits migration of ING4-deleted cells, in part by inducing a physical or proximal association between Cxcr3 and Egfr and signaling downstream via Gβγ. These results further indicated that ING4 plays a critical role in the regulation of Cxcl10 signaling that enables breast cancer progression.
Collapse
|
21
|
Saxena S, Singh RK. Chemokines orchestrate tumor cells and the microenvironment to achieve metastatic heterogeneity. Cancer Metastasis Rev 2021; 40:447-476. [PMID: 33959849 PMCID: PMC9863248 DOI: 10.1007/s10555-021-09970-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/22/2021] [Indexed: 01/26/2023]
Abstract
Chemokines, a subfamily of the cell cytokines, are low molecular weight proteins known to induce chemotaxis in leukocytes in response to inflammatory and pathogenic signals. A plethora of literature demonstrates that chemokines and their receptors regulate tumor progression and metastasis. With these diverse functionalities, chemokines act as a fundamental link between the tumor cells and their microenvironment. Recent studies demonstrate that the biology of chemokines and their receptor in metastasis is complex as numerous chemokines are involved in regulating site-specific tumor growth and metastasis. Successful treatment of disseminated cancer is a significant challenge. The most crucial problem for treating metastatic cancer is developing therapy regimes capable of overcoming heterogeneity problems within primary tumors and among metastases and within metastases (intralesional). This heterogeneity of malignant tumor cells can be related to metastatic potential, response to chemotherapy or specific immunotherapy, and many other factors. In this review, we have emphasized the role of chemokines in the process of metastasis and metastatic heterogeneity. Individual chemokines may not express the full potential to address metastatic heterogeneity, but chemokine networks need exploration. Understanding the interplay between chemokine-chemokine receptor networks between the tumor cells and their microenvironment is a novel approach to overcome the problem of metastatic heterogeneity. Recent advances in the understanding of chemokine networks pave the way for developing a potential targeted therapeutic strategy to treat metastatic cancer.
Collapse
Affiliation(s)
- Sugandha Saxena
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Rakesh K Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA.
| |
Collapse
|
22
|
Li CX, Zheng Y, Zhu H, Li CW, He Z, Wang C, Ding JH, Hu G, Lu M. β-arrestin 2 is essential for fluoxetine-mediated promotion of hippocampal neurogenesis in a mouse model of depression. Acta Pharmacol Sin 2021; 42:679-690. [PMID: 33526871 PMCID: PMC8115338 DOI: 10.1038/s41401-020-00576-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/08/2020] [Indexed: 12/15/2022]
Abstract
Over the last decade, the roles of β-arrestins in the treatment of neuropsychological diseases have become increasingly appreciated. Fluoxetine is the first selective serotonin reuptake inhibitor developed and is approved for the clinical treatment of depression. Emerging evidence suggests that fluoxetine can directly combine with the 5-HT receptor, which is a member of the G protein-coupled receptor (GPCR) family, in addition to suppressing the serotonin transporter. In this study, we prepared a chronic mild stress (CMS)-induced depression model with β-arrestin2-/- mice and cultured adult neural stem cells (ANSCs) to investigate the involvement of the 5-HT receptor-β-arrestin axis in the pathogenesis of depression and in the therapeutic effect of fluoxetine. We found that β-arrestin2 deletion abolished the fluoxetine-mediated improvement in depression-like behaviors and monoamine neurotransmitter levels, although β-arrestin2 knockout did not aggravate CMS-induced changes in mouse behaviors and neurotransmitters. Notably, the β-arrestin2-/- mice had a shortened dendritic length and reduced dendritic spine density, as well as decreased neural precursor cells, compared to the WT mice under both basal and CMS conditions. We further found that β-arrestin2 knockout decreased the number of proliferating cells in the hippocampal dentate gyrus and suppressed the proliferative capability of ANSCs in vitro. Moreover, β-arrestin2 knockout aggravated the impairment of cell proliferation induced by corticosterone and further blocked the fluoxetine-mediated promotion of mouse hippocampal neurogenesis. Mechanistically, we found that the 5-HT2BR-β-arrestin2-PI3K/Akt axis is essential to maintain the modulation of hippocampal neurogenesis in depressed mice. Our study may provide a promising target for the development of new antidepressant drugs.
Collapse
Affiliation(s)
- Chen-Xin Li
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ying Zheng
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hong Zhu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China
| | - Cheng-Wu Li
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhang He
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cong Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China
| | - Jian-Hua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China.
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China.
- Neuroprotective Drug Discovery Key Laboratory, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
23
|
Santagata S, Ieranò C, Trotta AM, Capiluongo A, Auletta F, Guardascione G, Scala S. CXCR4 and CXCR7 Signaling Pathways: A Focus on the Cross-Talk Between Cancer Cells and Tumor Microenvironment. Front Oncol 2021; 11:591386. [PMID: 33937018 PMCID: PMC8082172 DOI: 10.3389/fonc.2021.591386] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
The chemokine receptor 4 (CXCR4) and 7 (CXCR7) are G-protein-coupled receptors (GPCRs) activated through their shared ligand CXCL12 in multiple human cancers. They play a key role in the tumor/tumor microenvironment (TME) promoting tumor progression, targeting cell proliferation and migration, while orchestrating the recruitment of immune and stromal cells within the TME. CXCL12 excludes T cells from TME through a concentration gradient that inhibits immunoactive cells access and promotes tumor vascularization. Thus, dual CXCR4/CXCR7 inhibition will target different cancer components. CXCR4/CXCR7 antagonism should prevent the development of metastases by interfering with tumor cell growth, migration and chemotaxis and favoring the frequency of T cells in TME. Herein, we discuss the current understanding on the role of CXCL12/CXCR4/CXCR7 cross-talk in tumor progression and immune cells recruitment providing support for a combined CXCR4/CXCR7 targeting therapy. In addition, we consider emerging approaches that coordinately target both immune checkpoints and CXCL12/CXCR4/CXCR7 axis.
Collapse
Affiliation(s)
- Sara Santagata
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Caterina Ieranò
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Anna Maria Trotta
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Anna Capiluongo
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Federica Auletta
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Giuseppe Guardascione
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| | - Stefania Scala
- Research Department, Microenvironment Molecular Targets, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Napoli, Italy
| |
Collapse
|
24
|
Xu L, Li C, Hua F, Liu X. The CXCL12/CXCR7 signalling axis promotes proliferation and metastasis in cervical cancer. Med Oncol 2021; 38:58. [DOI: 10.1007/s12032-021-01481-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/13/2021] [Indexed: 02/07/2023]
|
25
|
O'Bryan SM, Mathis JM. CXCL12 Retargeting of an Oncolytic Adenovirus Vector to the Chemokine CXCR4 and CXCR7 Receptors in Breast Cancer. ACTA ACUST UNITED AC 2021; 12:311-336. [PMID: 34178415 DOI: 10.4236/jct.2021.126029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Breast cancer is the most frequently diagnosed cancer in women under 60, and the second most diagnosed cancer in women over 60. While significant progress has been made in developing targeted therapies for breast cancer, advanced breast cancer continues to have high mortality, with poor 5-year survival rates. Thus, current therapies are insufficient in treating advanced stages of breast cancer; new treatments are sorely needed to address the complexity of advanced-stage breast cancer. Oncolytic virotherapy has been explored as a therapeutic approach capable of systemic administration, targeting cancer cells, and sparing normal tissue. In particular, oncolytic adenoviruses have been exploited as viral vectors due to their ease of manipulation, production, and demonstrated clinical safety profile. In this study, we engineered an oncolytic adenovirus to target the chemokine receptors CXCR4 and CXCR7. The overexpression of CXCR4 and CXCR7 is implicated in the initiation, survival, progress, and metastasis of breast cancer. Both receptors bind to the ligand, CXCL12 (SDF-1), which has been identified to play a crucial role in the metastasis of breast cancer cells. This study incorporated a T4 fibritin protein fused to CXCL12 into the tail domain of an adenovirus fiber to retarget the vector to the CXCR4 and CXCR7 chemokine receptors. We showed that the modified virus targets and infects CXCR4- and CXCR7-overexpressing breast cancer cells more efficiently than a wild-type control vector. In addition, the substitution of the wild-type fiber and knob with the modified chimeric fiber did not interfere with oncolytic capability. Overall, the results of this study demonstrate the feasibility of retargeting adenovirus vectors to chemokine receptor-positive tumors.
Collapse
Affiliation(s)
- Samia M O'Bryan
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana USA
| | - J Michael Mathis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana USA.,University of North Texas Health Science Center, Graduate School of Biomedical Sciences, Fort Worth, Texas, USA
| |
Collapse
|
26
|
Koch C, Engele J. Functions of the CXCL12 Receptor ACKR3/CXCR7-What Has Been Perceived and What Has Been Overlooked. Mol Pharmacol 2020; 98:577-585. [PMID: 32883765 DOI: 10.1124/molpharm.120.000056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022] Open
Abstract
The CXCL12 system is central to the development of many organs and is further crucially engaged in pathophysiological processes underlying cancer, inflammation, and cardiovascular disorders. This disease-associated role presently focuses major interest on the two CXCL12 receptors, CXCR4 and atypical chemokine receptor 3 (ACKR3)/CXCR7, as promising therapeutic targets. Major obstacles in these ongoing efforts are confusing reports on the differential use of either ACKR3/CXCR7 and/or CXCR4 across various cells as well as on the specific function(s) of ACKR3/CXCR7. Although basically no doubts remain that CXCR4 represents a classic chemokine receptor, functions assigned to ACKR3/CXCR7 range from those of a strictly silent scavenger receptor eventually modulating CXCR4 signaling to an active and independent signaling receptor. In this review, we depict a thorough analysis of our present knowledge on different modes of organization and functions of the cellular CXCL12 system. We further highlight the potential role of ACKR3/CXCR7 as a "crosslinker" of different receptor systems. Finally, we discuss mechanisms with the potency to impinge on the cellular organization of the CXCL12 system and hence might represent additional future therapeutic targets. SIGNIFICANCE STATEMENT: Delineating the recognized functions of atypical chemokine receptor 3 and CXCR4 in CXCL12 signaling is central to the more detailed understanding of the role of the CXCL12 system in health and disease and will help to guide future research efforts.
Collapse
Affiliation(s)
- Christian Koch
- Institute of Anatomy, University of Leipzig, Medical Faculty, Leipzig, Germany
| | - Jürgen Engele
- Institute of Anatomy, University of Leipzig, Medical Faculty, Leipzig, Germany
| |
Collapse
|
27
|
Fumagalli A, Heuninck J, Pizzoccaro A, Moutin E, Koenen J, Séveno M, Durroux T, Junier MP, Schlecht-Louf G, Bachelerie F, Schütz D, Stumm R, Smit MJ, Guérineau NC, Chaumont-Dubel S, Marin P. The atypical chemokine receptor 3 interacts with Connexin 43 inhibiting astrocytic gap junctional intercellular communication. Nat Commun 2020; 11:4855. [PMID: 32978390 PMCID: PMC7519114 DOI: 10.1038/s41467-020-18634-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
The atypical chemokine receptor 3 (ACKR3) plays a pivotal role in directing the migration of various cellular populations and its over-expression in tumors promotes cell proliferation and invasiveness. The intracellular signaling pathways transducing ACKR3-dependent effects remain poorly characterized, an issue we addressed by identifying the interactome of ACKR3. Here, we report that recombinant ACKR3 expressed in HEK293T cells recruits the gap junction protein Connexin 43 (Cx43). Cx43 and ACKR3 are co-expressed in mouse brain astrocytes and human glioblastoma cells and form a complex in embryonic mouse brain. Functional in vitro studies show enhanced ACKR3 interaction with Cx43 upon ACKR3 agonist stimulation. Furthermore, ACKR3 activation promotes β-arrestin2- and dynamin-dependent Cx43 internalization to inhibit gap junctional intercellular communication in primary astrocytes. These results demonstrate a functional link between ACKR3 and gap junctions that might be of pathophysiological relevance. The atypical chemokine receptor 3 (ACKR3) is known to regulate cell migration, but the underlying mechanisms are unclear. Here, the authors show, from an interactome analysis, ACKR3 association with the gap junction protein Connexin 43 in vivo and ACKR3-mediated inhibition of astrocyte gap junctional communication.
Collapse
Affiliation(s)
- Amos Fumagalli
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joyce Heuninck
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Anne Pizzoccaro
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Enora Moutin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joyce Koenen
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France.,Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, 1081, HV, Amsterdam, The Netherlands
| | - Martial Séveno
- Biocampus Montpellier, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Thierry Durroux
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Marie-Pierre Junier
- CNRS UMR8246, Inserm U1130, Neuroscience Paris Seine-IBPS, Sorbonne Universités, Paris, France
| | - Géraldine Schlecht-Louf
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France
| | - Francoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France
| | - Dagmar Schütz
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747, Jena, Germany
| | - Ralf Stumm
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747, Jena, Germany
| | - Martine J Smit
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, 1081, HV, Amsterdam, The Netherlands
| | - Nathalie C Guérineau
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Séverine Chaumont-Dubel
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Philippe Marin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
28
|
Smit MJ, Schlecht-Louf G, Neves M, van den Bor J, Penela P, Siderius M, Bachelerie F, Mayor F. The CXCL12/CXCR4/ACKR3 Axis in the Tumor Microenvironment: Signaling, Crosstalk, and Therapeutic Targeting. Annu Rev Pharmacol Toxicol 2020; 61:541-563. [PMID: 32956018 DOI: 10.1146/annurev-pharmtox-010919-023340] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Elevated expression of the chemokine receptors CXCR4 and ACKR3 and of their cognate ligand CXCL12 is detected in a wide range of tumors and the tumor microenvironment (TME). Yet, the molecular mechanisms by which the CXCL12/CXCR4/ACKR3 axis contributes to the pathogenesis are complex and not fully understood. To dissect the role of this axis in cancer, we discuss its ability to impinge on canonical and less conventional signaling networks in different cancer cell types; its bidirectional crosstalk, notably with receptor tyrosine kinase (RTK) and other factors present in the TME; and the infiltration of immune cells that supporttumor progression. We discuss current and emerging avenues that target the CXCL12/CXCR4/ACKR3 axis. Coordinately targeting both RTKs and CXCR4/ACKR3 and/or CXCL12 is an attractive approach to consider in multitargeted cancer therapies. In addition, inhibiting infiltrating immune cells or reactivating the immune system along with modulating the CXCL12/CXCR4/ACKR3 axis in the TME has therapeutic promise.
Collapse
Affiliation(s)
- Martine J Smit
- Department of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, Netherlands;
| | - Géraldine Schlecht-Louf
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140 Clamart, France
| | - Maria Neves
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140 Clamart, France.,Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (CSIC/UAM), 28049 Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Jelle van den Bor
- Department of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, Netherlands;
| | - Petronila Penela
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (CSIC/UAM), 28049 Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Marco Siderius
- Department of Medicinal Chemistry, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Faculty of Science, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, Netherlands;
| | - Françoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140 Clamart, France
| | - Federico Mayor
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (CSIC/UAM), 28049 Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| |
Collapse
|
29
|
Sherif MF, Ismail IM, Ata SMS. Expression of CXCR7 in colorectal adenoma and adenocarcinoma: Correlation with clinicopathological parameters. Ann Diagn Pathol 2020; 49:151621. [PMID: 32949893 DOI: 10.1016/j.anndiagpath.2020.151621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 08/13/2020] [Accepted: 09/02/2020] [Indexed: 01/05/2023]
Abstract
Colorectal carcinoma (CRC) is one of the most lethal malignancies, it ranks third in cancer-related morbidity and mortality. Although great progress has been made in early diagnosis and combined treatment of CRC, the prognosis of patients remains poor owing to the high rate of recurrence and distant metastasis. CXCR7 belongs to chemokine receptor family and has been identified as a novel receptor for CXCL12. It plays an important role in development and in progression of cancer to metastatic stage. THE AIM OF STUDY To evaluate the immunohistochemical expression of CXCR7 in colorectal adenoma and carcinoma and to analyze its correlation with clinicopathological factors. This is retrospective study including 58 colonic adenocarcinoma specimens and 18 cases of colonic adenoma. RESULTS CXCR7 showed positive cytoplasmic expression in two out 18 cases of colorectal adenoma (11%) and 42 out of 58 cases of CRC (72.4%) with a significant difference between both (p < 0.001). We found a significant correlation between upregulation of CXCR7 and presence of lymphovascular tumor emboli, presence of lymph node metastasis and advanced TNM stage of the CRC. The association of the CXCR7 with patient age, sex, tumor size, depth of invasion and tumor cell differentiation was found to be non-significant. Regarding colonic adenoma, we found no significant association between CXCR7 expression on one hand and patient age, sex, tumor size, histologic type and degree of dysplasia on the other hand. CONCLUSION CXCR7 in CRC may act as a novel predictive indicator for prognosis and even be a potential molecular target for anticancer therapies.
Collapse
|
30
|
Jiang BC, Liu T, Gao YJ. Chemokines in chronic pain: cellular and molecular mechanisms and therapeutic potential. Pharmacol Ther 2020; 212:107581. [DOI: 10.1016/j.pharmthera.2020.107581] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/15/2020] [Indexed: 02/08/2023]
|
31
|
Morein D, Erlichman N, Ben-Baruch A. Beyond Cell Motility: The Expanding Roles of Chemokines and Their Receptors in Malignancy. Front Immunol 2020; 11:952. [PMID: 32582148 PMCID: PMC7287041 DOI: 10.3389/fimmu.2020.00952] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/23/2020] [Indexed: 01/10/2023] Open
Abstract
The anti-tumor activities of some members of the chemokine family are often overcome by the functions of many chemokines that are strongly and causatively linked with increased tumor progression. Being key leukocyte attractants, chemokines promote the presence of inflammatory pro-tumor myeloid cells and immune-suppressive cells in tumors and metastases. In parallel, chemokines elevate additional pro-cancerous processes that depend on cell motility: endothelial cell migration (angiogenesis), recruitment of mesenchymal stem cells (MSCs) and site-specific metastasis. However, the array of chemokine activities in cancer expands beyond such “typical” migration-related processes and includes chemokine-induced/mediated atypical functions that do not activate directly motility processes; these non-conventional chemokine functions provide the tumor cells with new sets of detrimental tools. Within this scope, this review article addresses the roles of chemokines and their receptors at atypical levels that are exerted on the cancer cell themselves: promoting tumor cell proliferation and survival; controlling tumor cell senescence; enriching tumors with cancer stem cells; inducing metastasis-related functions such as epithelial-to-mesenchymal transition (EMT) and elevated expression of matrix metalloproteinases (MMPs); and promoting resistance to chemotherapy and to endocrine therapy. The review also describes atypical effects of chemokines at the tumor microenvironment: their ability to up-regulate/stabilize the expression of inhibitory immune checkpoints and to reduce the efficacy of their blockade; to induce bone remodeling and elevate osteoclastogenesis/bone resorption; and to mediate tumor-stromal interactions that promote cancer progression. To illustrate this expanding array of atypical chemokine activities at the cancer setting, the review focuses on major metastasis-promoting inflammatory chemokines—including CXCL8 (IL-8), CCL2 (MCP-1), and CCL5 (RANTES)—and their receptors. In addition, non-conventional activities of CXCL12 which is a key regulator of tumor progression, and its CXCR4 receptor are described, alongside with the other CXCL12-binding receptor CXCR7 (RDC1). CXCR7, a member of the subgroup of atypical chemokine receptors (ACKRs) known also as ACKR3, opens the gate for discussion of atypical activities of additional ACKRs in cancer: ACKR1 (DARC, Duffy), ACKR2 (D6), and ACKR4 (CCRL1). The mechanisms involved in chemokine activities and the signals delivered by their receptors are described, and the clinical implications of these findings are discussed.
Collapse
Affiliation(s)
- Dina Morein
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nofar Erlichman
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Adit Ben-Baruch
- School of Molecular Cell Biology and Biotechnology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
32
|
Groblewska M, Litman-Zawadzka A, Mroczko B. The Role of Selected Chemokines and Their Receptors in the Development of Gliomas. Int J Mol Sci 2020; 21:ijms21103704. [PMID: 32456359 PMCID: PMC7279280 DOI: 10.3390/ijms21103704] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Among heterogeneous primary tumors of the central nervous system (CNS), gliomas are the most frequent type, with glioblastoma multiforme (GBM) characterized with the worst prognosis. In their development, certain chemokine/receptor axes play important roles and promote proliferation, survival, metastasis, and neoangiogenesis. However, little is known about the significance of atypical receptors for chemokines (ACKRs) in these tumors. The objective of the study was to present the role of chemokines and their conventional and atypical receptors in CNS tumors. Therefore, we performed a thorough search for literature concerning our investigation via the PubMed database. We describe biological functions of chemokines/chemokine receptors from various groups and their significance in carcinogenesis, cancer-related inflammation, neo-angiogenesis, tumor growth, and metastasis. Furthermore, we discuss the role of chemokines in glioma development, with particular regard to their function in the transition from low-grade to high-grade tumors and angiogenic switch. We also depict various chemokine/receptor axes, such as CXCL8-CXCR1/2, CXCL12-CXCR4, CXCL16-CXCR6, CX3CL1-CX3CR1, CCL2-CCR2, and CCL5-CCR5 of special importance in gliomas, as well as atypical chemokine receptors ACKR1-4, CCRL2, and PITPMN3. Additionally, the diagnostic significance and usefulness of the measurement of some chemokines and their receptors in the blood and cerebrospinal fluid (CSF) of glioma patients is also presented.
Collapse
Affiliation(s)
- Magdalena Groblewska
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland;
| | - Ala Litman-Zawadzka
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland;
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland;
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland;
- Correspondence: ; Tel.: +48-85-831-8785
| |
Collapse
|
33
|
Sjöberg E, Meyrath M, Chevigné A, Östman A, Augsten M, Szpakowska M. The diverse and complex roles of atypical chemokine receptors in cancer: From molecular biology to clinical relevance and therapy. Adv Cancer Res 2020; 145:99-138. [PMID: 32089166 DOI: 10.1016/bs.acr.2019.12.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chemokines regulate directed cell migration, proliferation and survival and are key components in cancer biology. They exert their functions by interacting with seven-transmembrane domain receptors that signal through G proteins (GPCRs). A subgroup of four chemokine receptors known as the atypical chemokine receptors (ACKRs) has emerged as essential regulators of the chemokine functions. ACKRs play diverse and complex roles in tumor biology from tumor initiation to metastasis, including cancer cell proliferation, adherence to endothelium, epithelial-mesenchymal transition (EMT), extravasation from blood vessels, tumor-associated angiogenesis or protection from immunological responses. This chapter gives an overview on the established and emerging roles that the atypical chemokine receptors ACKR1, ACKR2, ACKR3 and ACKR4 play in the different phases of cancer development and dissemination, their clinical relevance, as well as on the hurdles to overcome in ACKRs targeting as cancer therapy.
Collapse
Affiliation(s)
- Elin Sjöberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Max Meyrath
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Andy Chevigné
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Martyna Szpakowska
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
34
|
Wong M, Newton LR, Hartmann J, Hennrich ML, Wachsmuth M, Ronchi P, Guzmán-Herrera A, Schwab Y, Gavin AC, Gilmour D. Dynamic Buffering of Extracellular Chemokine by a Dedicated Scavenger Pathway Enables Robust Adaptation during Directed Tissue Migration. Dev Cell 2020; 52:492-508.e10. [PMID: 32059773 DOI: 10.1016/j.devcel.2020.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 11/22/2019] [Accepted: 01/13/2020] [Indexed: 01/16/2023]
Abstract
How tissues migrate robustly through changing guidance landscapes is poorly understood. Here, quantitative imaging is combined with inducible perturbation experiments to investigate the mechanisms that ensure robust tissue migration in vivo. We show that tissues exposed to acute "chemokine floods" halt transiently before they perfectly adapt, i.e., return to the baseline migration behavior in the continued presence of elevated chemokine levels. A chemokine-triggered phosphorylation of the atypical chemokine receptor Cxcr7b reroutes it from constitutive ubiquitination-regulated degradation to plasma membrane recycling, thus coupling scavenging capacity to extracellular chemokine levels. Finally, tissues expressing phosphorylation-deficient Cxcr7b migrate normally in the presence of physiological chemokine levels but show delayed recovery when challenged with elevated chemokine concentrations. This work establishes that adaptation to chemokine fluctuations can be "outsourced" from canonical GPCR signaling to an autonomously acting scavenger receptor that both senses and dynamically buffers chemokine levels to increase the robustness of tissue migration.
Collapse
Affiliation(s)
- Mie Wong
- Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany.
| | - Lionel R Newton
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Jonas Hartmann
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Marco L Hennrich
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Malte Wachsmuth
- Luxendo GmbH, Kurfürsten-Anlage 58, 69115 Heidelberg, Germany
| | - Paolo Ronchi
- Electron Microscopy Core Facility, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Alejandra Guzmán-Herrera
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Yannick Schwab
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany; Electron Microscopy Core Facility, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Anne-Claude Gavin
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany; Department for Cell Physiology and Metabolism, University of Geneva, 1 rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Darren Gilmour
- Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany.
| |
Collapse
|
35
|
Lokeshwar BL, Kallifatidis G, Hoy JJ. Atypical chemokine receptors in tumor cell growth and metastasis. Adv Cancer Res 2020; 145:1-27. [PMID: 32089162 DOI: 10.1016/bs.acr.2019.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atypical chemokine receptors (ACKRs) are seven-transmembrane cell surface protein receptors expressed in immune cells, normal mesenchymal cells, and several tumor cells. As of this writing, six ACKRs have been characterized by diverse activities. They bind both cysteine-cysteine (CC) type and cysteine-X-cysteine (CXC)-type chemokines, either alone, or together with a ligand bound-functional G-protein coupled (typical) chemokine receptor. The major structural difference between ACKRs and typical chemokine receptors is the substituted DRYLAIV amino acid motif in the second intracellular loop of the ACKR. Due to this substitution, these receptors cannot bind Gαi-type G-proteins responsible for intracellular calcium mobilization and cellular chemotaxis. Although initially characterized as non-signaling transmembrane receptors (decoy receptors) that attenuate ligand-induced signaling by GPCRs, studies of all ACKRs have shown ligand-independent and ligand-dependent transmembrane signaling in both non-tumor and tumor cells. The precise function and mechanism of the differential expression of ACKRs in many tumors are not understood well. The use of antagonists of ACKRs ligands has shown limited antitumor potential; however, depleting ACKR expression resulted in a reduction in experimental tumor growth and metastasis. The ACKRs represent a unique class of transmembrane signaling proteins that regulate growth, survival, and metastatic processes in tumor cells, affecting multiple pathways of tumor growth. Therefore, closer investigations of ACKRs have a high potential for identifying therapeutics which affect the intracellular signaling, preferentially via the ligand-independent mechanism.
Collapse
Affiliation(s)
- Bal L Lokeshwar
- Georgia Cancer Center, Augusta University, Augusta, GA, United States; Research Service, Charlie Norwood Veterans Administration Medical Center, Augusta, GA, United States.
| | - Georgios Kallifatidis
- Georgia Cancer Center, Augusta University, Augusta, GA, United States; Research Service, Charlie Norwood Veterans Administration Medical Center, Augusta, GA, United States; Department of Biological Sciences, Augusta University, Augusta, GA, United States
| | - James J Hoy
- LCMB Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
36
|
Xun Y, Yang H, Li J, Wu F, Liu F. CXC Chemokine Receptors in the Tumor Microenvironment and an Update of Antagonist Development. Rev Physiol Biochem Pharmacol 2020; 178:1-40. [PMID: 32816229 DOI: 10.1007/112_2020_35] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chemokine receptors, a diverse group within the seven-transmembrane G protein-coupled receptor superfamily, are frequently overexpressed in malignant tumors. Ligand binding activates multiple downstream signal transduction cascades that drive tumor growth and metastasis, resulting in poor clinical outcome. These receptors are thus considered promising targets for anti-tumor therapy. This article reviews recent studies on the expression and function of CXC chemokine receptors in various tumor microenvironments and recent developments in cancer therapy using CXC chemokine receptor antagonists.
Collapse
Affiliation(s)
- Yang Xun
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Hua Yang
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Jiekai Li
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Fuling Wu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Fang Liu
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China.
| |
Collapse
|
37
|
Neves M, Fumagalli A, van den Bor J, Marin P, Smit MJ, Mayor F. The Role of ACKR3 in Breast, Lung, and Brain Cancer. Mol Pharmacol 2019; 96:819-825. [PMID: 30745320 DOI: 10.1124/mol.118.115279] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/30/2019] [Indexed: 12/24/2022] Open
Abstract
Recent reports regarding the significance of chemokine receptors in disease have put a spotlight on atypical chemokine receptor 3 (ACKR3). This atypical chemokine receptor is overexpressed in numerous cancer types and has been involved in the modulation of tumor cell proliferation and migration, tumor angiogenesis, or resistance to drugs, thus contributing to cancer progression and metastasis occurrence. Here, we focus on the clinical significance and potential mechanisms underlying the pathologic role of ACKR3 in breast, lung, and brain cancer and discuss its possible relevance as a prognostic factor and potential therapeutic target in these contexts.
Collapse
Affiliation(s)
- Maria Neves
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma Madrid, Madrid, Spain (M.N., F.M.); Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.B., M.J.S.); and CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain (F.M.)
| | - Amos Fumagalli
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma Madrid, Madrid, Spain (M.N., F.M.); Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.B., M.J.S.); and CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain (F.M.)
| | - Jelle van den Bor
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma Madrid, Madrid, Spain (M.N., F.M.); Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.B., M.J.S.); and CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain (F.M.)
| | - Philippe Marin
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma Madrid, Madrid, Spain (M.N., F.M.); Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.B., M.J.S.); and CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain (F.M.)
| | - Martine J Smit
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma Madrid, Madrid, Spain (M.N., F.M.); Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.B., M.J.S.); and CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain (F.M.)
| | - Federico Mayor
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma Madrid, Madrid, Spain (M.N., F.M.); Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (J.B., M.J.S.); and CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain (F.M.)
| |
Collapse
|
38
|
Fumagalli A, Zarca A, Neves M, Caspar B, Hill SJ, Mayor F, Smit MJ, Marin P. CXCR4/ACKR3 Phosphorylation and Recruitment of Interacting Proteins: Key Mechanisms Regulating Their Functional Status. Mol Pharmacol 2019; 96:794-808. [PMID: 30837297 DOI: 10.1124/mol.118.115360] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/21/2019] [Indexed: 01/14/2023] Open
Abstract
The C-X-C motif chemokine receptor type 4 (CXCR4) and the atypical chemokine receptor 3 (ACKR3/CXCR7) are class A G protein-coupled receptors (GPCRs). Accumulating evidence indicates that GPCR subcellular localization, trafficking, transduction properties, and ultimately their pathophysiological functions are regulated by both interacting proteins and post-translational modifications. This has encouraged the development of novel techniques to characterize the GPCR interactome and to identify residues subjected to post-translational modifications, with a special focus on phosphorylation. This review first describes state-of-the-art methods for the identification of GPCR-interacting proteins and GPCR phosphorylated sites. In addition, we provide an overview of the current knowledge of CXCR4 and ACKR3 post-translational modifications and an exhaustive list of previously identified CXCR4- or ACKR3-interacting proteins. We then describe studies highlighting the importance of the reciprocal influence of CXCR4/ACKR3 interactomes and phosphorylation states. We also discuss their impact on the functional status of each receptor. These studies suggest that deeper knowledge of the CXCR4/ACKR3 interactomes along with their phosphorylation and ubiquitination status would shed new light on their regulation and pathophysiological functions.
Collapse
Affiliation(s)
- Amos Fumagalli
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Aurélien Zarca
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Maria Neves
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Birgit Caspar
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Stephen J Hill
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Federico Mayor
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Martine J Smit
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Philippe Marin
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| |
Collapse
|
39
|
Heuninck J, Perpiñá Viciano C, Işbilir A, Caspar B, Capoferri D, Briddon SJ, Durroux T, Hill SJ, Lohse MJ, Milligan G, Pin JP, Hoffmann C. Context-Dependent Signaling of CXC Chemokine Receptor 4 and Atypical Chemokine Receptor 3. Mol Pharmacol 2019; 96:778-793. [PMID: 31092552 DOI: 10.1124/mol.118.115477] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/21/2019] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are regulated by complex molecular mechanisms, both in physiologic and pathologic conditions, and their signaling can be intricate. Many factors influence their signaling behavior, including the type of ligand that activates the GPCR, the presence of interacting partners, the kinetics involved, or their location. The two CXC-type chemokine receptors, CXC chemokine receptor 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3), both members of the GPCR superfamily, are important and established therapeutic targets in relation to cancer, human immunodeficiency virus infection, and inflammatory diseases. Therefore, it is crucial to understand how the signaling of these receptors works to be able to specifically target them. In this review, we discuss how the signaling pathways activated by CXCR4 and ACKR3 can vary in different situations. G protein signaling of CXCR4 depends on the cellular context, and discrepancies exist depending on the cell lines used. ACKR3, as an atypical chemokine receptor, is generally reported to not activate G proteins but can broaden its signaling spectrum upon heteromerization with other receptors, such as CXCR4, endothelial growth factor receptor, or the α 1-adrenergic receptor (α 1-AR). Also, CXCR4 forms heteromers with CC chemokine receptor (CCR) 2, CCR5, the Na+/H+ exchanger regulatory factor 1, CXCR3, α 1-AR, and the opioid receptors, which results in differential signaling from that of the monomeric subunits. In addition, CXCR4 is present on membrane rafts but can go into the nucleus during cancer progression, probably acquiring different signaling properties. In this review, we also provide an overview of the currently known critical amino acids involved in CXCR4 and ACKR3 signaling.
Collapse
Affiliation(s)
- Joyce Heuninck
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Cristina Perpiñá Viciano
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Ali Işbilir
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Birgit Caspar
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Davide Capoferri
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Stephen J Briddon
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Thierry Durroux
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Stephen J Hill
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Martin J Lohse
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Graeme Milligan
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Jean-Philippe Pin
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Carsten Hoffmann
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| |
Collapse
|
40
|
Yu C, Zhang Y. Characterization of the prognostic values of CXCR family in gastric cancer. Cytokine 2019; 123:154785. [PMID: 31344595 DOI: 10.1016/j.cyto.2019.154785] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 07/07/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The role of CXC chemokine receptors (CXCRs) in gastric cancer (GC) has been an increasing focus. However, comprehensive prognostic values of CXCR members in GC are yet to be clearly defined. METHODS Multiple public available datasets, including Kaplan-Meier (KM) plotter, oncomine, the cancer genome atlas (TCGA), SurvExpress platform and the tumor immune estimation resource (TIMER), were used for mRNA expression and prognostic characterization. Nomogram method was used for clinical model prediction. RESULTS CXCR3, CXCR4 and CXCR5 displayed significantly up-regulated expression in tumor compared to normal. High mRNA expression of CXCR2 (HR = 0.77, 95%CI: 0.62-0.95, p = 0.014), CXCR3 (HR = 0.74, 95%CI: 0.61-0.90, p = 0.0024), CXCR4 (HR = 0.7, 95%CI: 0.58-0.86, p = 0.00048), CXCR5 (HR = 0.72, 95%CI: 0.59-0.87, p = 0.00093) and CXCR6 (HR = 0.66, 95%CI: 0.54-0.81, p = 4.9e-05) was significantly associated with favorable overall survival (OS). The prognostic values of CXCR members were also explored in subtypes, including HER2 status, Lauren classification, pathological stages. The low risk group of CXCR signature displayed a significantly favorable OS compared to the high risk group (HR = 3.22, 95% CI = 2.21-4.69, p = 1.057e-09). Nomogram clinical models were established for both OS (C-index: 0.692; 95%CI: 0.648-0.736) and recurrence free survival (C-index: 0.731; 95%CI: 0.675-0.786). In addition, CXCR6 and CD8+T cells featured the highest correlation (partial-cor = 0.781, p = 4.17e-77). CONCLUSION This study identified distinct expression and prognostic values of CXCR members in GC using public databases.
Collapse
Affiliation(s)
- Chaoran Yu
- Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200025, PR China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200025, PR China; Department of Gastrointestinal Surgery, Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 20025, PR China.
| | - Yujie Zhang
- Department of Gastrointestinal Surgery, Tongji Hospital, Tongji Medical College in Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
41
|
Elmansi AM, Awad ME, Eisa NH, Kondrikov D, Hussein KA, Aguilar-Pérez A, Herberg S, Periyasamy-Thandavan S, Fulzele S, Hamrick MW, McGee-Lawrence ME, Isales CM, Volkman BF, Hill WD. What doesn't kill you makes you stranger: Dipeptidyl peptidase-4 (CD26) proteolysis differentially modulates the activity of many peptide hormones and cytokines generating novel cryptic bioactive ligands. Pharmacol Ther 2019; 198:90-108. [PMID: 30759373 PMCID: PMC7883480 DOI: 10.1016/j.pharmthera.2019.02.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dipeptidyl peptidase 4 (DPP4) is an exopeptidase found either on cell surfaces where it is highly regulated in terms of its expression and surface availability (CD26) or in a free/circulating soluble constitutively available and intrinsically active form. It is responsible for proteolytic cleavage of many peptide substrates. In this review we discuss the idea that DPP4-cleaved peptides are not necessarily inactivated, but rather can possess either a modified receptor selectivity, modified bioactivity, new antagonistic activity, or even a novel activity relative to the intact parent ligand. We examine in detail five different major DPP4 substrates: glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), peptide tyrosine-tyrosine (PYY), and neuropeptide Y (NPY), and stromal derived factor 1 (SDF-1 aka CXCL12). We note that discussion of the cleaved forms of these five peptides are underrepresented in the research literature, and are both poorly investigated and poorly understood, representing a serious research literature gap. We believe they are understudied and misinterpreted as inactive due to several factors. This includes lack of accurate and specific quantification methods, sample collection techniques that are inherently inaccurate and inappropriate, and a general perception that DPP4 cleavage inactivates its ligand substrates. Increasing evidence points towards many DPP4-cleaved ligands having their own bioactivity. For example, GLP-1 can work through a different receptor than GLP-1R, DPP4-cleaved GIP can function as a GIP receptor antagonist at high doses, and DPP4-cleaved PYY, NPY, and CXCL12 can have different receptor selectivity, or can bind novel, previously unrecognized receptors to their intact ligands, resulting in altered signaling and functionality. We believe that more rigorous research in this area could lead to a better understanding of DPP4's role and the biological importance of the generation of novel cryptic ligands. This will also significantly impact our understanding of the clinical effects and side effects of DPP4-inhibitors as a class of anti-diabetic drugs that potentially have an expanding clinical relevance. This will be specifically relevant in targeting DPP4 substrate ligands involved in a variety of other major clinical acute and chronic injury/disease areas including inflammation, immunology, cardiology, stroke, musculoskeletal disease and injury, as well as cancer biology and tissue maintenance in aging.
Collapse
Affiliation(s)
- Ahmed M Elmansi
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States
| | - Mohamed E Awad
- Department of Oral Biology, School of Dentistry, Augusta University, Augusta, GA 30912, United States
| | - Nada H Eisa
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, United States; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Dmitry Kondrikov
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States
| | - Khaled A Hussein
- Department of Surgery and Medicine, National Research Centre, Cairo, Egypt
| | - Alexandra Aguilar-Pérez
- Department of Anatomy and Cell Biology, Indiana University School of Medicine in Indianapolis, IN, United States; Department of Cellular and Molecular Biology, School of Medicine, Universidad Central del Caribe, Bayamon, 00956, Puerto Rico; Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Samuel Herberg
- Departments of Ophthalmology & Cell and Dev. Bio., SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | | | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States
| | - Mark W Hamrick
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States
| | - Meghan E McGee-Lawrence
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States
| | - Carlos M Isales
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States; Division of Endocrinology, Diabetes and Metabolism, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Brian F Volkman
- Biochemistry Department, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - William D Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29403, United States; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States; Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Center for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, 30912, United States.
| |
Collapse
|
42
|
C-X-C Chemokine Receptor Type 7 (CXCR-7) Expression in Invasive Ductal Carcinoma of Breast in Association with Clinicopathological Features. Pathol Oncol Res 2019; 26:1015-1020. [PMID: 30955173 DOI: 10.1007/s12253-019-00649-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 03/20/2019] [Indexed: 10/27/2022]
Abstract
C-X-C chemokine receptor type 7 (CXCR-7) is an atypical receptor for chemokines whose role in different stages of carcinogenesis has been evaluated in breast cancer cell lines and animal models. Moreover, it has been demonstrated to be a target of regulation by the tumor suppressor microRNA (miR)-100. In the present study, we assessed CXCR-7 expression in 60 breast cancer patients in association with clinicopathological and demographic data of patients. We also extracted the results of our previous work on miR-100 expression in the same cohort of patients to assess the correlation between miR-100 and CXCR-7 expression levels. Transcript levels of CXCR-7 were significantly higher in tumoral tissues compared with adjacent non-cancerous tissues (ANCTs) (Tumoral vs. ANCTs: 3.64 ± 1.8 vs. 0.73 ± 1.3, P = 0.000). A significant negative correlation was detected between CXCR-7 protein and miR-100 transcript levels (r = -0.526, P < 0.05). High CXCR-7 mRNA levels were significantly associated with tumor size (P = 0.01). Besides, high protein levels were more prevalent in higher TNM stages (P = 0.000). Moreover, high CXCR-7 protein levels were significantly associated with ER (P = 0.005) and PR (P = 0.02) status. The present work provides further evidence for the role of CXCR-7 in breast cancer and proposes the elimination of inhibitory effects of miR-100 on CXCR-7 expression as a mechanism for its up-regulation in breast cancer tissues.
Collapse
|
43
|
Liu B, Song S, Setroikromo R, Chen S, Hu W, Chen D, van der Wekken AJ, Melgert BN, Timens W, van den Berg A, Saber A, Haisma HJ. CX Chemokine Receptor 7 Contributes to Survival of KRAS-Mutant Non-Small Cell Lung Cancer upon Loss of Epidermal Growth Factor Receptor. Cancers (Basel) 2019; 11:cancers11040455. [PMID: 30935067 PMCID: PMC6520904 DOI: 10.3390/cancers11040455] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/25/2019] [Accepted: 03/27/2019] [Indexed: 12/24/2022] Open
Abstract
KRAS-driven non-small cell lung cancer (NSCLC) patients have no effective targeted treatment. In this study, we aimed to investigate targeting epidermal growth factor receptor (EGFR) as a therapeutic approach in KRAS-driven lung cancer cells. We show that ablation of EGFR significantly suppressed tumor growth in KRAS-dependent cells and induced significantly higher expression of CX chemokine receptor 7 (CXCR7) and activation of MAPK (ERK1/2). Conversely, rescue of EGFR led to CXCR7 downregulation in EGFR−/− cells. Dual EGFR and CXCR7 inhibition led to substantial reduction of MAPK (pERK) and synergistic inhibition of cell growth. Analysis of two additional EGFR knockout NSCLC cell lines using CRISPR/Cas9 revealed genotype dependency of CXCR7 expression. In addition, treatment of different cells with gefitinib increased CXCR7 expression in EGFRwt but decreased it in EGFRmut cells. CXCR7 protein expression was detected in all NSCLC patient samples, with higher levels in adenocarcinoma as compared to squamous cell lung carcinoma and healthy control cases. In conclusion, EGFR and CXCR7 have a crucial interaction in NSCLC, and dual inhibition may be a potential therapeutic option for NSCLC patients.
Collapse
Affiliation(s)
- Bin Liu
- Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology, University of Groningen, 9713 AV Groningen, The Netherlands.
| | - Shanshan Song
- Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology, University of Groningen, 9713 AV Groningen, The Netherlands.
- Toxicology and Targeting Groningen Research Institute for Pharmacy, Department of Pharmacokinetics, University of Groningen, 9713 AV Groningen, The Netherlands.
| | - Rita Setroikromo
- Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology, University of Groningen, 9713 AV Groningen, The Netherlands.
| | - Siwei Chen
- Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology, University of Groningen, 9713 AV Groningen, The Netherlands.
| | - Wenteng Hu
- Department of Thoracic Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| | - Deng Chen
- Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology, University of Groningen, 9713 AV Groningen, The Netherlands.
| | - Anthonie J van der Wekken
- Department of Pulmonary Diseases, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Barbro N Melgert
- Toxicology and Targeting Groningen Research Institute for Pharmacy, Department of Pharmacokinetics, University of Groningen, 9713 AV Groningen, The Netherlands.
- GRIAC- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands.
| | - Ali Saber
- Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology, University of Groningen, 9713 AV Groningen, The Netherlands.
| | - Hidde J Haisma
- Groningen Research Institute of Pharmacy, Department of Chemical and Pharmaceutical Biology, University of Groningen, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
44
|
Xu S, Tang J, Wang C, Liu J, Fu Y, Luo Y. CXCR7 promotes melanoma tumorigenesis via Src kinase signaling. Cell Death Dis 2019; 10:191. [PMID: 30804329 PMCID: PMC6389959 DOI: 10.1038/s41419-019-1442-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 01/19/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022]
Abstract
Chemokine receptors have been documented to exert critical functions in melanoma progression. However, current drugs targeting these receptors have limited efficacy in clinical applications, suggesting the urgency to further explore the roles of chemokine receptors in melanoma. Here we found that C–X–C chemokine receptor 7 (CXCR7) was the most highly expressed chemokine receptor in murine melanoma cell lines. In addition, the expression level of CXCR7 was positively correlated with melanoma progression in the clinical samples. High CXCR7 expression was associated with shorter overall survival in melanoma patients. Increased expression of CXCR7 augmented melanoma proliferation in vitro and tumor growth in vivo, whereas knockout of CXCR7 exhibited significant inhibitory effects. Moreover, our data elucidated that CXCR7 activated Src kinase phosphorylation in a β-arrestin2-dependent manner. The administration of the Src kinase inhibitor PP1 or siRNA specific for β-arrestin2 abolished CXCR7-promoted cell proliferation. Importantly, CXCR7 also regulated melanoma angiogenesis and the secretion of vascular endothelial growth factor (VEGF). Subsequent investigations revealed a novel event that the activation of the CXCR7-Src axis stimulated the phosphorylation of eukaryotic translation initiation factor 4E (eIF4E) to accelerate the translation of hypoxia-inducible factor 1α (HIF-1α), which enhanced the secretion of VEGF from melanoma cells. Collectively, our results illuminate the crucial roles of CXCR7 in melanoma tumorigenesis, and indicate the potential of targeting CXCR7 as new therapeutic strategies for melanoma treatment.
Collapse
Affiliation(s)
- Siran Xu
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program (PTN), School of Life Sciences, Peking University, Beijing, China.,The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jiaze Tang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Chunying Wang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jie Liu
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yan Fu
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yongzhang Luo
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, China. .,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, China. .,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
45
|
Fan H, Wang W, Yan J, Xiao L, Yang L. Prognostic significance of CXCR7 in cancer patients: a meta-analysis. Cancer Cell Int 2018; 18:212. [PMID: 30574021 PMCID: PMC6300004 DOI: 10.1186/s12935-018-0702-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/07/2018] [Indexed: 02/07/2023] Open
Abstract
Background CXC chemokine receptor 7 (CXCR7) is frequently overexpressed in a variety of tumors. Nevertheless, whether CXCR7 can be used as a tumor prognosis marker has not been systematically assessed. The current meta-analysis was performed to obtain an accurate evaluation of the relationship between CXCR7 level and the prognosis of cancer patients. Methods Embase, Web of Science, and PubMed were systematically searched according to a defined search strategy up to June 11, 2018. Then, the required data were extracted from all qualified studies which were screened out based on the defined inclusion and exclusion criteria. Finally, the hazard ratios (HR) with 95% confidence intervals (CI) were used to evaluate the prognostic significance of CXCR7 in tumor patients. Results A total of 28 original research studies comprising 33 cohorts and 5685 patients were included in this meta-analysis. The results showed that CXCR7 overexpression was significantly related to worse overall survival (OS) (HR 1.72; 95% CI 1.49–1.99), disease-free survival (DFS) (HR 5.58; 95% CI 3.16–9.85), progression-free survival (PFS) (HR 2.83; 95% CI 1.66–4.85) and recurrence-free survival (RFS) (HR 1.58; 95% CI 1.34–1.88) in cancer patients. Furthermore, for certain types of cancer, significant associations between higher CXCR7 expression and worse OS of glioma (HR 1.77; 95% CI 1.43–2.19), breast cancer (HR 1.45; 95% CI 1.28–1.63), esophageal cancer (HR 2.72; 95% CI 1.11–6.66) and pancreatic cancer (HR 1.46; 95% CI 1.12–1.90) were found. However, for lung cancer and hepatocellular cancer, there was no significant relationship between CXCR7 expression level and OS, (HR 2.40; 95% CI 0.34–17.07) and (HR 1.37; 95% CI 0.84–2.24) respectively. Conclusions Increased CXCR7 level could predict poor prognosis of tumor patients and might be regarded as a novel prognostic biomarker for tumor patients.
Collapse
Affiliation(s)
- Huiqian Fan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijun Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Yan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Xiao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Wang R, Li J, Yin C, Zhao D, Yin L. Identification of differentially expressed genes and typical fusion genes associated with three subtypes of breast cancer. Breast Cancer 2018; 26:305-316. [PMID: 30446971 DOI: 10.1007/s12282-018-0924-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/15/2018] [Indexed: 11/25/2022]
Abstract
BACKGROUND This study aimed to identify the differentially expressed genes (DEGs) and the typical fusion genes in different types of breast cancers using RNA-seq. METHODS GSE52643 was downloaded from Gene Expression Omnibus, which included 1 normal sample (MCF10A) and 7 breast cancer samples (BT-474, BT-20, MCF7, MDA-MB-231, MDA-MB-468, T47D, and ZR-75-1). The transcript abundance and the DEGs screening were performed by Cufflinks. The functional and pathway enrichment was analyzed by Gostats. SnowShoes-FTD was applied to identify the fusion genes. RESULTS We screened 430, 445, 397, 417, 369, 557, and 375 DEGs in BT-474, BT-20, MCF7, DA-MB-231, MDA-MB-468, T47D, and ZR-75-1, respectively, compared with MCF10A. DEGs in each comparison group (such as CD40 and CDH1) were significantly enriched in the functions of cell adhesion and extracellular matrix organization and pathways of CAMs and ECM receptor interaction. UCP2 was a common DEG in the 7 comparison groups. SFRP1 and MMP7 were significantly enriched in wnt/-catenin signaling pathway in MDA-MB-231. FAS was significantly enriched in autoimmune thyroid disease pathway in BT-474. Besides, we screened 96 fusion genes, such as ESR1-C6orf97 in ZR-75-1, COBRA1-C9orf167 in BT-20, and VAPB-IKZF3 and ACACA-STAC2 in BT-474. CONCLUSIONS The DEGs such as SFRP1, MMP7, CDH1, FAS, and UCP2 might be the potential biomarkers in breast cancer. Furthermore, some pivotal fusion genes like ESR1-C6orf97 with COBRA1-C9orf167 and VAPB-IKZF3 with ACACA-STAC2 were found in Luminal A and Luminal B breast cancer, respectively.
Collapse
Affiliation(s)
- Rong Wang
- National Research Institute for Health and Family Planning, Beijing, 100081, China
| | - Jinbin Li
- Core Laboratory of Translational Medicine, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853, China
| | - Chunyu Yin
- Core Laboratory of Translational Medicine, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853, China
| | - Di Zhao
- Dermatological of Department, The 309 Hospital of Chinese PLA, Beijing, 100091, China
| | - Ling Yin
- Core Laboratory of Translational Medicine, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
47
|
Borroni EM, Savino B, Bonecchi R, Locati M. Chemokines sound the alarmin: The role of atypical chemokine in inflammation and cancer. Semin Immunol 2018; 38:63-71. [DOI: 10.1016/j.smim.2018.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/08/2018] [Indexed: 12/17/2022]
|
48
|
Zhang B, Sun J, Yao X, Li J, Tu Y, Yao F, Sun S. Knockdown of B7H6 inhibits tumor progression in triple-negative breast cancer. Oncol Lett 2018; 16:91-96. [PMID: 29963127 PMCID: PMC6019890 DOI: 10.3892/ol.2018.8689] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 01/22/2018] [Indexed: 12/15/2022] Open
Abstract
The B7 family, the most common family of secondary signaling molecules, consists of eight cell-surface proteins, which regulate the T-cell mediated immune response by delivering co-inhibitory or co-stimulatory signals through their corresponding ligands. Among them, natural killer cell cytotoxicity receptor 3 ligand 1 (NCR3LG1, also known as B7H6) has been reported as a new member, and is involved in tumor progression of various types of human cancer. However, the role of B7H6 in triple-negative breast cancer (TNBC) remains unknown. In the present study, western blotting was performed to determine the protein expression levels of B7H6 in a normal mammary epithelial cell line (MCF-10A), non-TNBC breast cancer cell lines (MCF-7 and AU565) and TNBC cell lines (MDA-MB-231 and MDA-MB-468). B7H6 was knocked down using small interfering RNA, and an MTT assay was performed to determine proliferation ability, flow cytometry was used to analyze apoptosis, and Transwell and wound-healing assays were performed to measure migration ability. Expression of proliferation-associated proteins (SMAD family member 4 and β-catenin) and apoptosis-associated proteins (BCL2 associated X, BCL2 apoptosis regulator and caspase-3) were analyzed by western blotting. The results demonstrated that B7H6 was highly expressed in TNBC cells, and that knockdown of B7H6 inhibited cell proliferation and migration, and promoted apoptosis. Furthermore, the results revealed that proliferation and apoptosis-associated proteins were altered in the B7H6-knockdown MDA-MB-231 cells. In conclusion, the present study demonstrated that B7H6 may have significant roles in the regulation of cell proliferation, apoptosis and migration of TNBC cells.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jinzhong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaoli Yao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yi Tu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Feng Yao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
49
|
Wang W, Qiao Y, Li Z. New Insights into Modes of GPCR Activation. Trends Pharmacol Sci 2018; 39:367-386. [DOI: 10.1016/j.tips.2018.01.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
|
50
|
Yuan CL, Liu ZH, Zou N, Wang YL, Chen ZY. Relationship between expression of CXCR7 and NF-κB in breast cancer tissue and occurrence of breast cancer and lymphatic metastasis. Saudi J Biol Sci 2018; 24:1767-1770. [PMID: 29551920 PMCID: PMC5851893 DOI: 10.1016/j.sjbs.2017.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/03/2017] [Accepted: 11/05/2017] [Indexed: 12/01/2022] Open
Abstract
It is designed to discuss the relationship between the expression of chemokine receptor 7 (chemokine receptor 7, CXCR7) and nuclear transcription factor-κB (nuclear factor kappa B, NF-κB) and the occurrence of the breast cancer and lymphatic metastasis. Method: 80 samples were excised and confirmed as breast cancer through our hospital pathology from January 2014 to December 2016 and tumor tissues and normal mammary tissues 2 cm from the tumor edges were taken as an experimental group and a control group, respectively. The method of immunohistochemical is utilized to test the expression of CXCR7 and NF-κB in the breast cancer tissue, compared with the para-carcinoma tissue, and analyze its relevance with the clinicopathologic features of the breast cancer tissue, such as tumor size, TNM staging, lymphatic metastasis and other conditions. Results: both CXCR7 and NF-κB were highly expressed in the breast cancer tissue, the positive rate was significantly higher that that of paracancerous normal tissues, and the difference was statistically significant. And the expressions of CXCR7 and NF-κB were related to TNM staging of the breast cancer and lymphatic metastasis and unrelated to the tumor size, age, and expressions of ER, PR and HER2. Conclusion: both CXCR7 and NF-κB are related to the malignant grade of the breast cancer and lymphatic metastasis, which may be regarded as in important indicator to judge the prognosis of the breast cancer and be expected to be the new target of curing part of the breast cancer.
Collapse
Affiliation(s)
- Cheng-Liang Yuan
- Department of Clinical Laboratory, Deyang People's Hospital, Deyang 618000, China
| | - Zhao-Hong Liu
- Department of Clinical Laboratory, Deyang People's Hospital, Deyang 618000, China
| | - Ning Zou
- Department of Clinical Laboratory, Deyang People's Hospital, Deyang 618000, China
| | - Ya-Li Wang
- Department of Clinical Laboratory, Deyang People's Hospital, Deyang 618000, China
| | - Zhong-Yao Chen
- Department of Clinical Laboratory, Deyang People's Hospital, Deyang 618000, China
| |
Collapse
|