151
|
Shi Y, Riese DJ, Shen J. The Role of the CXCL12/CXCR4/CXCR7 Chemokine Axis in Cancer. Front Pharmacol 2020; 11:574667. [PMID: 33363463 PMCID: PMC7753359 DOI: 10.3389/fphar.2020.574667] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/29/2020] [Indexed: 12/27/2022] Open
Abstract
Chemokines are a family of small, secreted cytokines which regulate a variety of cell functions. The C-X-C motif chemokine ligand 12 (CXCL12) binds to C-X-C chemokine receptor type 4 (CXCR4) and C-X-C chemokine receptor type 7 (CXCR7). The interaction of CXCL12 and its receptors subsequently induces downstream signaling pathways with broad effects on chemotaxis, cell proliferation, migration, and gene expression. Accumulating evidence suggests that the CXCL12/CXCR4/CXCR7 axis plays a pivotal role in tumor development, survival, angiogenesis, metastasis, and tumor microenvironment. In addition, this chemokine axis promotes chemoresistance in cancer therapy via complex crosstalk with other pathways. Multiple small molecules targeting CXCR4/CXCR7 have been developed and used for preclinical and clinical cancer treatment. In this review, we describe the roles of the CXCL12/CXCR4/CXCR7 axis in cancer progression and summarize strategies to develop novel targeted cancer therapies.
Collapse
Affiliation(s)
| | | | - Jianzhong Shen
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| |
Collapse
|
152
|
Russo E, Santoni A, Bernardini G. Tumor inhibition or tumor promotion? The duplicity of CXCR3 in cancer. J Leukoc Biol 2020; 108:673-685. [PMID: 32745326 DOI: 10.1002/jlb.5mr0320-205r] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 03/23/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor tissue includes cancer cells and normal stromal cells such as vascular endothelial cells, connective tissue cells (cancer associated fibroblast, mesenchymal stem cell), and immune cells (tumor-infiltrating lymphocytes or TIL, dendritic cells, eosinophils, basophils, mast cells, tumor-associated macrophages or TAM, myeloid-derived suppressor cells or MDSC). Anti-tumor activity is mainly mediated by infiltration of NK cells, Th1 and CD8+ T cells, and correlates with expression of NK cell and T cell attracting chemokines. Nevertheless, cancer cells hijack tissue homeostasis through secretion of cytokines and chemokines that mediate not only the induction of an inflamed status that supports cancer cell survival and growth, but also the recruitment and/or activation of immune suppressive cells. CXCL9, CXCL10, and CXCL11 are known for their tumor-inhibiting properties, but their overexpression in several hematologic and solid tumors correlates with disease severity, suggesting a role in tumor promotion. The dichotomous nature of CXCR3 ligands activity mainly depends on several molecular mechanisms induced by cancer cells themselves able to divert immune responses and to alter the whole local environment. A deep understanding of the nature of such phenomenon may provide a rationale to build up a CXCR3/ligand axis targeting strategy. In this review, we will discuss the role of CXCR3 in cancer progression and in regulation of anti-tumor immune response and immunotherapy.
Collapse
Affiliation(s)
- Eleonora Russo
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Institute Pasteur-Italia, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Institute Pasteur-Italia, Rome, Italy.,IRCCS, Neuromed, Pozzilli, Isernia, Italy
| | - Giovanni Bernardini
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory affiliated to Institute Pasteur-Italia, Rome, Italy
| |
Collapse
|
153
|
Eiger DS, Boldizsar N, Honeycutt CC, Gardner J, Rajagopal S. Biased agonism at chemokine receptors. Cell Signal 2020; 78:109862. [PMID: 33249087 DOI: 10.1016/j.cellsig.2020.109862] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/07/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
In the human chemokine system, interactions between the approximately 50 known endogenous chemokine ligands and 20 known chemokine receptors (CKRs) regulate a wide range of cellular functions and biological processes including immune cell activation and homeostasis, development, angiogenesis, and neuromodulation. CKRs are a family of G protein-coupled receptors (GPCR), which represent the most common and versatile class of receptors in the human genome and the targets of approximately one third of all Food and Drug Administration-approved drugs. Chemokines and CKRs bind with significant promiscuity, as most CKRs can be activated by multiple chemokines and most chemokines can activate multiple CKRs. While these ligand-receptor interactions were previously regarded as redundant, it is now appreciated that many chemokine:CKR interactions display biased agonism, the phenomenon in which different ligands binding to the same receptor signal through different pathways with different efficacies, leading to distinct biological effects. Notably, these biased responses can be modulated through changes in ligand, receptor, and or the specific cellular context (system). In this review, we explore the biochemical mechanisms, functional consequences, and therapeutic potential of biased agonism in the chemokine system. An enhanced understanding of biased agonism in the chemokine system may prove transformative in the understanding of the mechanisms and consequences of biased signaling across all GPCR subtypes and aid in the development of biased pharmaceuticals with increased therapeutic efficacy and safer side effect profiles.
Collapse
Affiliation(s)
| | - Noelia Boldizsar
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA.
| | | | - Julia Gardner
- Trinity College of Arts and Sciences, Duke University, Durham, NC 27710, USA.
| | - Sudarshan Rajagopal
- Department of Biochemistry, Duke University, Durham, NC 27710, USA; Department of Medicine, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
154
|
Dalle Nogare DE, Natesh N, Vishwasrao HD, Shroff H, Chitnis AB. Zebrafish Posterior Lateral Line primordium migration requires interactions between a superficial sheath of motile cells and the skin. eLife 2020; 9:58251. [PMID: 33237853 PMCID: PMC7688310 DOI: 10.7554/elife.58251] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
The Zebrafish Posterior Lateral Line primordium migrates in a channel between the skin and somites. Its migration depends on the coordinated movement of its mesenchymal-like leading cells and trailing cells, which form epithelial rosettes, or protoneuromasts. We describe a superficial population of flat primordium cells that wrap around deeper epithelialized cells and extend polarized lamellipodia to migrate apposed to the overlying skin. Polarization of lamellipodia extended by both superficial and deeper protoneuromast-forming cells depends on Fgf signaling. Removal of the overlying skin has similar effects on superficial and deep cells: lamellipodia are lost, blebs appear instead, and collective migration fails. When skinned embryos are embedded in Matrigel, basal and superficial lamellipodia are recovered; however, only the directionality of basal protrusions is recovered, and migration is not rescued. These observations support a key role played by superficial primordium cells and the skin in directed migration of the Posterior Lateral Line primordium.
Collapse
Affiliation(s)
- Damian E Dalle Nogare
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Naveen Natesh
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Harshad D Vishwasrao
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, United States
| | - Hari Shroff
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, United States.,Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, United States
| | - Ajay B Chitnis
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| |
Collapse
|
155
|
Nguyen HT, Reyes-Alcaraz A, Yong HJ, Nguyen LP, Park HK, Inoue A, Lee CS, Seong JY, Hwang JI. CXCR7: a β-arrestin-biased receptor that potentiates cell migration and recruits β-arrestin2 exclusively through Gβγ subunits and GRK2. Cell Biosci 2020; 10:134. [PMID: 33292475 PMCID: PMC7686738 DOI: 10.1186/s13578-020-00497-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Some chemokine receptors referred to as atypical chemokine receptors (ACKRs) are thought to non-signaling decoys because of their inability to activate typical G-protein signaling pathways. CXCR7, also known as ACKR3, binds to only two chemokines, SDF-1α and I-TAC, and recruits β-arrestins. SDF-1α also binds to its own conventional receptor, CXCR4, involving in homeostatic modulation such as development and immune surveillance as well as pathological conditions such as inflammation, ischemia, and cancers. Recently, CXCR7 is suggested as a key therapeutic target together with CXCR4 in such conditions. However, the molecular mechanisms underlying cellular responses and functional relation with CXCR7 and CXCR4 have not been elucidated, despite massive studies. Therefore, we aimed to reveal the molecular networks of CXCR7 and CXCR4 and compare their effects on cell migration. METHODS Base on structural complementation assay using NanoBiT technology, we characterized the distinct mechanisms underlying β-arrestin2 recruitment by both CXCR4 and CXCR7. Crosslinking and immunoprecipitation were conducted to analyze complex formation of the receptors. Gene deletion using CRISPR and reconstitution of the receptors were applied to analysis of ligand-dependent ERK phosphorylation and cell migration. All experiments were performed in triplicate and repeated more than three times. Unpaired Student's t-tests or ANOVA using PRISM5 software were employed for statistical analyses. RESULTS Ligand binding to CXCR7 does not result in activation of typical signaling pathways via Gα subunits but activation of GRK2 via βγ subunits and receptor phosphorylation with subsequent β-arrestin2 recruitment. In contrast, CXCR4 induced Gαi activation and recruited β-arrestin2 through C-terminal phosphorylation by both GRK2 and GRK5. SDF-1α-stimulated ERK phosphorylation was facilitated by CXCR4, but not CXCR7. Heterodimerization of CXCR4 and CXCR7 was not confirmed in this study, while homodimerization of them was verified by crosslinking experiment and NanoBiT assay. Regarding chemotaxis, SDF-1α-stimulated cell migration was mediated by both CXCR4 and CXCR7. CONCLUSION This study demonstrates that SDF-1α-stimulated CXCR7 mediates β-arrestin2 recruitment via different molecular networking from that of CXCR4. CXCR7 may be neither a simple scavenger nor auxiliary receptor but plays an essential role in cell migration through cooperation with CXCR4.
Collapse
Affiliation(s)
- Huong Thi Nguyen
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | | | - Hyo Jeong Yong
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Lan Phuong Nguyen
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Hee-Kyung Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Cheol Soon Lee
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jae Young Seong
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jong-Ik Hwang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
156
|
Cun Y, Diao B, Zhang Z, Wang G, Yu J, Ma L, Rao Z. Role of the stromal cell derived factor-1 in the biological functions of endothelial progenitor cells and its underlying mechanisms. Exp Ther Med 2020; 21:39. [PMID: 33273969 PMCID: PMC7706408 DOI: 10.3892/etm.2020.9471] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022] Open
Abstract
Stromal cell derived factor-1 (SDF-1) is a chemokine that plays a critical role in the homing of stem and progenitor cells, including endothelial progenitor cells (EPCs). However, little research has been undertaken to evaluate the roles of SDF-1 in the biological functions of EPCs and related signaling pathways. The present study aimed to investigate the biological functions of EPCs in response to SDF-1, as well as the underlying mechanisms. The effects of SDF-1 treatment on EPC proliferation, migration and tube formation were assessed by performing MTS, Transwell and in vitro tube formation assays, respectively. The phosphorylation status of Akt and ERK was evaluated by western blotting. The present results indicated that SDF-1 treatment enhanced EPC proliferation, migration and tube formation compared with the control group. Furthermore, SDF-1-induced EPC proliferation was significantly reduced following treatment with a C-X-C Motif Chemokine Receptor 4 antagonist (AMD3100), a PI3K inhibitor (LY294002) and the mitogen-activated protein kinase kinase inhibitor (MEK; PD98059). SDF-1-induced migration and angiogenesis were significantly suppressed by the PI3K inhibitor, but not the MEK inhibitor. Moreover, SDF-1 significantly increased the protein expression levels of phosphorylated (p)-Akt and p-ERK; however, SDF-1-induced effects on protein expression were suppressed by AMD3100, LY294002 and PD98059. Thus, SDF-1-induced EPC proliferation was mediated by activation of the Akt and ERK signaling pathways, whereas SDF-1-mediated EPC migration and tube formation only involved activation of the Akt signaling pathway.
Collapse
Affiliation(s)
- Yanping Cun
- Department of Oncology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Bo Diao
- Department of Clinical Experiment, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Zhimin Zhang
- Department of Oncology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Gang Wang
- Department of Clinical Experiment, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Jing Yu
- Department of Clinical Experiment, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Lianting Ma
- Department of Neurosurgery, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Zhiguo Rao
- Department of Oncology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
157
|
Heck AM, Ishida T, Hadland B. Location, Location, Location: How Vascular Specialization Influences Hematopoietic Fates During Development. Front Cell Dev Biol 2020; 8:602617. [PMID: 33282876 PMCID: PMC7691428 DOI: 10.3389/fcell.2020.602617] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/30/2020] [Indexed: 01/22/2023] Open
Abstract
During embryonic development, sequential waves of hematopoiesis give rise to blood-forming cells with diverse lineage potentials and self-renewal properties. This process must accomplish two important yet divergent goals: the rapid generation of differentiated blood cells to meet the needs of the developing embryo and the production of a reservoir of hematopoietic stem cells to provide for life-long hematopoiesis in the adult. Vascular beds in distinct anatomical sites of extraembryonic tissues and the embryo proper provide the necessary conditions to support these divergent objectives, suggesting a critical role for specialized vascular niche cells in regulating disparate blood cell fates during development. In this review, we will examine the current understanding of how organ- and stage-specific vascular niche specialization contributes to the development of the hematopoietic system.
Collapse
Affiliation(s)
- Adam M. Heck
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Takashi Ishida
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Brandon Hadland
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
158
|
CXCR7, CXCR4, and Their Ligand Expression Profile in Traumatic Brain Injury. World Neurosurg 2020; 147:e16-e24. [PMID: 33189916 DOI: 10.1016/j.wneu.2020.11.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/05/2020] [Accepted: 11/05/2020] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Traumatic brain injury (TBI) is a health problem worldwide, and therapeutic strategies to enhance brain tissue repair to lessen neurologic sequels are imperative. We aimed to analyze the impact of the inflammatory process in TBI through CXCR4 and CXCR7 chemokine receptors and their ligands' CXCL11 and CXCL12 expression profile in search for potential new druggable targets. METHODS Twelve pericontusional tissues from severe TBI patients submitted to surgical treatment, and 20 control brain tissues from normal autopsy were analyzed for expression profile by real-time quantitative-polymerase chain reaction. CXCR7 and CXCR4 protein expressions were analyzed by immunohistochemistry. The findings were correlated with the clinical evolution. RESULTS Increased gene expression of both receptors and their ligands was observed in TBI compared with controls, presenting high sensitivity and specificity to differentiate TBI from normal control (area under the curve ranging from 0.85 to 0.98, P < 0.001). In particular, CXCR7 expression highly correlated with CXCR4 and both ligands' expressions in TBI. Higher immunoreactions for CXCR7 and CXCR4 were identified in neurons and endothelial cells of TBI samples compared with controls. The patients presenting upregulated chemokine expression levels showed a trend toward favorable clinical evolution at up to 6 months of follow-up. CONCLUSIONS The neuroprotective trend of CXCR4, CXCR7, CXCL11, and CXCL12 in TBI observed in this initial analysis warrants further studies with more patients, analyzing the involved signaling pathways for the development of new therapeutic strategies for TBI.
Collapse
|
159
|
Cao H, Gao Y, Wang R, Guo Q, Hui H. Wogonin reverses the drug resistance of chronic myelogenous leukemia cells to imatinib through CXCL12-CXCR4/7 axis in bone marrow microenvironment. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1046. [PMID: 33145265 PMCID: PMC7575956 DOI: 10.21037/atm-20-1166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background In the current study, chronic myeloid leukemia (CML) cells (K562 and KU812) co-cultured with human bone marrow stromal cells (BMSCs) were significantly less sensitive to imatinib (IM). The activation of the CXCL12-CXCR4/7 axis plays an important role in the protective effect of the bone marrow microenvironment (BME) on CML cells. The aim of this study was to investigate whether Wogonin could increase the sensitivity of CML cells to IM when they were co-cultured with BME and explore its underlying mechanism. Methods A model of CML cells co-cultured with BMSCs was applied in vitro. Flow cytometric, western blotting, immunofluorescence, and RT-PCR assays were used to explore the protective effects of BME on CML cells. Results The results showed that Wogonin could reverse the resistance of CML cells to IM under co-culture conditions by inhibiting Transforming growth factor-β (TGF-β) secretion in the BME, preventing the translocation of Smad4 into nucleus and subsequently reducing the expression of CXCR4 and CXCR7 in CML cells. Moreover, the reverse effect of Wogonin was demonstrated by inhibiting the activation of CXCL12-CXCR4/7 axis via restraining the TGF-β/Smad4/Id3 pathway in vitro. In vivo studies also showed that Wogonin decreased the expression of CXCR4 and CXCR7 in mice bone marrow with low systemic toxicity, and the mechanism was consistent with the in vitro study. Conclusions Wogonin increases the sensitivity of CML cells to IM in BME by controlling the TGF-β/Smad4/Id3 pathway and decreasing the expression of CXCR4 and CXCR7. These results co-supported the point that Wogonin could be a potential candidate of reversal agents on treatment of IM-resistant CML.
Collapse
Affiliation(s)
- Hanbo Cao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yuan Gao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Ruixuan Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Hui Hui
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
160
|
Matti C, D'Uonnolo G, Artinger M, Melgrati S, Salnikov A, Thelen S, Purvanov V, Strobel TD, Spannagel L, Thelen M, Legler DF. CCL20 is a novel ligand for the scavenging atypical chemokine receptor 4. J Leukoc Biol 2020; 107:1137-1154. [PMID: 32533638 DOI: 10.1002/jlb.2ma0420-295rrr] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022] Open
Abstract
The chemokine CCL20 is broadly produced by endothelial cells in the liver, the lung, in lymph nodes and mucosal lymphoid tissues, and recruits CCR6 expressing leukocytes, particularly dendritic cells, mature B cells, and subpopulations of T cells. How CCL20 is systemically scavenged is currently unknown. Here, we identify that fluorescently labeled human and mouse CCL20 are efficiently taken-up by the atypical chemokine receptor ACKR4. CCL20 shares ACKR4 with the homeostatic chemokines CCL19, CCL21, and CCL25, although with a lower affinity. We demonstrate that all 4 human chemokines recruit β-arrestin1 and β-arrestin2 to human ACKR4. Similarly, mouse CCL19, CCL21, and CCL25 equally activate the human receptor. Interestingly, at the same chemokine concentration, mouse CCL20 did not recruit β-arrestins to human ACKR4. Further cross-species analysis suggests that human ACKR4 preferentially takes-up human CCL20, whereas mouse ACKR4 similarly internalizes mouse and human CCL20. Furthermore, we engineered a fluorescently labeled chimeric chemokine consisting of the N-terminus of mouse CCL25 and the body of mouse CCL19, termed CCL25_19, which interacts with and is taken-up by human and mouse ACKR4.
Collapse
Affiliation(s)
- Christoph Matti
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Giulia D'Uonnolo
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Marc Artinger
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Serena Melgrati
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Angela Salnikov
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Sylvia Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Vladimir Purvanov
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Tobias D Strobel
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Lisa Spannagel
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg), University of Konstanz, Kreuzlingen, Switzerland.,Faculty of Biology, University of Konstanz, Konstanz, Germany.,Theodor Kocher Institute, University of Bern, Bern, Switzerland
| |
Collapse
|
161
|
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
|
162
|
Groover MK, Richmond JM. Potential therapeutic manipulations of the CXCR3 chemokine axis for the treatment of inflammatory fibrosing diseases. F1000Res 2020; 9:1197. [PMID: 33145014 PMCID: PMC7590900 DOI: 10.12688/f1000research.26728.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Chemokines play important roles in homeostasis and inflammatory processes. While their roles in leukocyte recruitment are well-appreciated, chemokines play additional roles in the body, including mediating or regulating angiogenesis, tumor metastasis and wound healing. In this opinion article, we focus on the role of CXCR3 and its ligands in fibrotic processes. We emphasize differences of the effects of each ligand, CXCL9, CXCL10 and CXCL11, on fibroblasts in different tissues of the body. We include discussions of differences in signaling pathways that may account for protective or pro-fibrotic effects of each ligand in different experimental models and ex vivo analysis of human tissues. Our goal is to highlight potential reasons why there are disparate findings in different models, and to suggest ways in which this chemokine axis could be manipulated for the treatment of fibrosis.
Collapse
Affiliation(s)
- Morgan K. Groover
- Department of Dermatology, University of Massachussetts Medical School, Worcester, MA, 01605, USA
| | - Jillian M. Richmond
- Department of Dermatology, University of Massachussetts Medical School, Worcester, MA, 01605, USA
| |
Collapse
|
163
|
Jiang C, Li R, Ma X, Hu H, Wei L, Zhao J. Plerixafor stimulates adhesive activity and endothelial regeneration of endothelial progenitor cells via elevating CXCR7 expression. J Diabetes Complications 2020; 34:107654. [PMID: 32741660 DOI: 10.1016/j.jdiacomp.2020.107654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/14/2020] [Accepted: 06/14/2020] [Indexed: 01/28/2023]
Abstract
AIMS To assess the effects of plerixafor on function and endothelial regeneration of endothelial progenitor cells (EPCs). METHODS The proliferation and adhesion capacity of EPCs were evaluated in vitro. Furthermore, the expression levels of CXC chemokine receptor-7 (CXCR7) were detected before and after treatment with plerixafor. The CXCR7 expression of EPCs was knocked-down by RNA interference to evaluate the role of CXCR7 in regulating function of EPCs. A rat carotid artery injury model was established to assess the influences of plerixafor on endothelial regeneration. RESULTS Plerixafor stimulated adhesion capacity of EPCs, associating with upregulation of CXCR7 and activation of LFA-1 and VLA-4 molecules. Knockdown of CXCR7 slightly impaired proliferation capacity but significantly attenuated adhesion capacity of EPCs. Plerixafor facilitated endothelial repair at 7 days, while reduced neointimal hyperplasia at 7 and 14 days via recruiting more EPCs participating in endothelial reparation. CONCLUSIONS Plerixafor can positively regulate adhesion capacity of EPCs to HUVECs via elevating the expression level of CXCR7 and stimulating LFA-1 and VLA-4 molecules activation. Treatment with plerixafor accelerated re-endothelialization and inhibited neointimal hyperplasia after endoth elial injury, indicating that it can to be used for endothelial regeneration.
Collapse
Affiliation(s)
- Chunyu Jiang
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China
| | - Ruiting Li
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China
| | - Xu Ma
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China
| | - Hui Hu
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China
| | - Liming Wei
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China
| | - Jungong Zhao
- Department of Radiology, The Sixth People's Hospital, Affiliated to Shanghai Jiao Tong University, 600 Yi-Shan Road, Shanghai 200233, PR China..
| |
Collapse
|
164
|
Enciso J, Mendoza L, Álvarez-Buylla ER, Pelayo R. Dynamical modeling predicts an inflammation-inducible CXCR7+ B cell precursor with potential implications in lymphoid blockage pathologies. PeerJ 2020; 8:e9902. [PMID: 33062419 PMCID: PMC7531334 DOI: 10.7717/peerj.9902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/18/2020] [Indexed: 12/16/2022] Open
Abstract
Background The blockage at the early B lymphoid cell development pathway within the bone marrow is tightly associated with hematopoietic and immune diseases, where the disruption of basal regulatory networks prevents the continuous replenishment of functional B cells. Dynamic computational models may be instrumental for the comprehensive understanding of mechanisms underlying complex differentiation processes and provide novel prediction/intervention platforms to reinvigorate the system. Methods By reconstructing a three-module regulatory network including genetic transcription, intracellular transduction, and microenvironment communication, we have investigated the early B lineage cell fate decisions in normal and pathological settings. The early B cell differentiation network was simulated as a Boolean model and then transformed, using fuzzy logic, to a continuous model. We tested null and overexpression mutants to analyze the emergent behavior of the network. Due to its importance in inflammation, we investigated the effect of NFkB induction at different early B cell differentiation stages. Results While the exhaustive synchronous and asynchronous simulation of the early B cell regulatory network (eBCRN) reproduced the configurations of the hematopoietic progenitors and early B lymphoid precursors of the pathway, its simulation as a continuous model with fuzzy logics suggested a transient IL-7R+ ProB-to-Pre-B subset expressing pre-BCR and a series of dominant B-cell transcriptional factors. This conspicuous differentiating cell population up-regulated CXCR7 and reduced CXCR4 and FoxO1 expression levels. Strikingly, constant but intermediate NFkB signaling at specific B cell differentiation stages allowed stabilization of an aberrant CXCR7+ pre-B like phenotype with apparent affinity to proliferative signals, while under constitutive overactivation of NFkB, such cell phenotype was aberrantly exacerbated from the earliest stage of common lymphoid progenitors. Our mutant models revealed an abnormal delay in the BCR assembly upon NFkB activation, concomitant to sustained Flt3 signaling, down-regulation of Ebf1, Irf4 and Pax5 genes transcription, and reduced Ig recombination, pointing to a potential lineage commitment blockage. Discussion For the first time, an inducible CXCR7hi B cell precursor endowed with the potential capability of shifting central lymphoid niches, is inferred by computational modeling. Its phenotype is compatible with that of leukemia-initiating cells and might be the foundation that bridges inflammation with blockage-related malignancies and a wide range of immunological diseases. Besides the predicted differentiation impairment, inflammation-inducible phenotypes open the possibility of newly formed niches colonized by the reported precursor. Thus, emergent bone marrow ecosystems are predicted following a pro-inflammatory induction, that may lead to hematopoietic instability associated to blockage pathologies.
Collapse
Affiliation(s)
- Jennifer Enciso
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Metepec, Puebla, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, México.,Programa de Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Mexico City, México
| | - Luis Mendoza
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, México
| | | | - Rosana Pelayo
- Centro de Investigación Biomédica de Oriente, Delegación Puebla, Instituto Mexicano del Seguro Social, Metepec, Puebla, Mexico
| |
Collapse
|
165
|
Haque N, Fareez IM, Fong LF, Mandal C, Kasim NHA, Kacharaju KR, Soesilawati P. Role of the CXCR4-SDF1-HMGB1 pathway in the directional migration of cells and regeneration of affected organs. World J Stem Cells 2020. [DOI: 10.4252/wjsc.v12.i9.0000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
166
|
Haque N, Fareez IM, Fong LF, Mandal C, Abu Kasim NH, Kacharaju KR, Soesilawati P. Role of the CXCR4-SDF1-HMGB1 pathway in the directional migration of cells and regeneration of affected organs. World J Stem Cells 2020; 12:938-951. [PMID: 33033556 PMCID: PMC7524697 DOI: 10.4252/wjsc.v12.i9.938] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/18/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
In recent years, several studies have reported positive outcomes of cell-based therapies despite insufficient engraftment of transplanted cells. These findings have created a huge interest in the regenerative potential of paracrine factors released from transplanted stem or progenitor cells. Interestingly, this notion has also led scientists to question the role of proteins in the secretome produced by cells, tissues or organisms under certain conditions or at a particular time of regenerative therapy. Further studies have revealed that the secretomes derived from different cell types contain paracrine factors that could help to prevent apoptosis and induce proliferation of cells residing within the tissues of affected organs. This could also facilitate the migration of immune, progenitor and stem cells within the body to the site of inflammation. Of these different paracrine factors present within the secretome, researchers have given proper consideration to stromal cell-derived factor-1 (SDF1) that plays a vital role in tissue-specific migration of the cells needed for regeneration. Recently researchers recognized that SDF1 could facilitate site-specific migration of cells by regulating SDF1-CXCR4 and/or HMGB1-SDF1-CXCR4 pathways which is vital for tissue regeneration. Hence in this study, we have attempted to describe the role of different types of cells within the body in facilitating regeneration while emphasizing the HMGB1-SDF1-CXCR4 pathway that orchestrates the migration of cells to the site where regeneration is needed.
Collapse
Affiliation(s)
- Nazmul Haque
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Selangor 42610, Malaysia
| | - Ismail M Fareez
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Selangor 42610, Malaysia
| | - Liew Fong Fong
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Selangor 42610, Malaysia
| | - Chanchal Mandal
- Biotechnology and Genetic Engineering Discipline, Life Science, Khulna University, Khulna 9208, Bangladesh
| | - Noor Hayaty Abu Kasim
- Faculty of Dentistry, University Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
- Faculty of Dental Medicine, Universitas Airlangga, Surabaya 411007, Indonesia
| | - Kranthi Raja Kacharaju
- Department of Conservative Dentistry, Faculty of Dentistry MAHSA University, Selangor 42610, Malaysia
| | - Pratiwi Soesilawati
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya 60115, Indonesia
| |
Collapse
|
167
|
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
|
168
|
Niu J, Yan T, Guo W, Wang W, Zhao Z, Ren T, Huang Y, Zhang H, Yu Y, Liang X. Identification of Potential Therapeutic Targets and Immune Cell Infiltration Characteristics in Osteosarcoma Using Bioinformatics Strategy. Front Oncol 2020; 10:1628. [PMID: 32974202 PMCID: PMC7471873 DOI: 10.3389/fonc.2020.01628] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma is one of the most aggressive malignant bone tumors worldwide. Although great advancements have been made in its treatment owing to the advent of neoadjuvant chemotherapy, the problem of lung metastasis is a major obstacle in the improvement of survival outcomes. Thus, the aim of the present study is to screen novel and key biomarkers, which may act as potential prognostic markers and therapeutic targets in osteosarcoma. We utilized the robust rank aggregation (RRA) method to integrate three osteosarcoma microarray datasets downloaded from the Gene Expression Omnibus (GEO) database, and we identified the robust differentially expressed genes (DEGs) between primary and metastatic osteosarcoma tissues. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to explore the functions of robust DEGs. The results of enrichment analysis showed that the robust DEGs were closely associated with osteosarcoma development and progression. Immune cell infiltration analysis was also conducted by CIBERSORT algorithm, and we found that macrophages are the most principal infiltrating immune cells in osteosarcoma, especially macrophages M0 and M2. Then, the protein–protein interaction network and key modules were constructed by Cytoscape, and 10 hub genes were selected by plugin cytoHubba from the whole network. The survival analysis of hub genes was also carried out based on the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) database. The integrated bioinformatics analysis was utilized to provide new insight into osteosarcoma development and metastasis and identified EGR1, CXCL10, MYC, and CXCR4 as potential biomarkers for prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Jianfang Niu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Taiqiang Yan
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Wei Wang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Zhiqing Zhao
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Tingting Ren
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Yi Huang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Hongliang Zhang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Yiyang Yu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| | - Xin Liang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Peking University, Beijing, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China
| |
Collapse
|
169
|
Charan M, Verma AK, Hussain S, Misri S, Mishra S, Majumder S, Ramaswamy B, Ahirwar D, Ganju RK. Molecular and Cellular Factors Associated with Racial Disparity in Breast Cancer. Int J Mol Sci 2020; 21:5936. [PMID: 32824813 PMCID: PMC7460595 DOI: 10.3390/ijms21165936] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
Recent studies have demonstrated that racial differences can influence breast cancer incidence and survival rate. African American (AA) women are at two to three fold higher risk for breast cancer than other ethnic groups. AA women with aggressive breast cancers show worse prognoses and higher mortality rates relative to Caucasian (CA) women. Over the last few years, effective treatment strategies have reduced mortality from breast cancer. Unfortunately, the breast cancer mortality rate among AA women remains higher compared to their CA counterparts. The focus of this review is to underscore the racial differences and differential regulation/expression of genetic signatures in CA and AA women with breast cancer. Moreover, immune cell infiltration significantly affects the clinical outcome of breast cancer. Here, we have reviewed recent findings on immune cell recruitment in the tumor microenvironment (TME) and documented its association with breast cancer racial disparity. In addition, we have extensively discussed the role of cytokines, chemokines, and other cell signaling molecules among AA and CA breast cancer patients. Furthermore, we have also reviewed the distinct genetic and epigenetic changes in AA and CA patients. Overall, this review article encompasses various molecular and cellular factors associated with breast cancer disparity that affects mortality and clinical outcome.
Collapse
Affiliation(s)
- Manish Charan
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Ajeet K. Verma
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Shahid Hussain
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Swati Misri
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Sanjay Mishra
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Sarmila Majumder
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA; (S.M.); (B.R.)
| | - Bhuvaneswari Ramaswamy
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA; (S.M.); (B.R.)
| | - Dinesh Ahirwar
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
| | - Ramesh K. Ganju
- Department of Pathology, Ohio State University, Columbus, OH 43210, USA; (M.C.); (A.K.V.); (S.H.); (S.M.); (S.M.)
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA; (S.M.); (B.R.)
| |
Collapse
|
170
|
Caligiuri A, Pastore M, Lori G, Raggi C, Di Maira G, Marra F, Gentilini A. Role of Chemokines in the Biology of Cholangiocarcinoma. Cancers (Basel) 2020; 12:cancers12082215. [PMID: 32784743 PMCID: PMC7463556 DOI: 10.3390/cancers12082215] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
Cholangiocarcinoma (CCA), a heterogeneous tumor with poor prognosis, can arise at any level in the biliary tree. It may derive from epithelial cells in the biliary tracts and peribiliary glands and possibly from progenitor cells or even hepatocytes. Several risk factors are responsible for CCA onset, however an inflammatory milieu nearby the biliary tree represents the most common condition favoring CCA development. Chemokines play a key role in driving the immunological response upon liver injury and may sustain tumor initiation and development. Chemokine receptor-dependent pathways influence the interplay among various cellular components, resulting in remodeling of the hepatic microenvironment towards a pro-inflammatory, pro-fibrogenic, pro-angiogenic and pre-neoplastic setting. Moreover, once tumor develops, chemokine signaling may influence its progression. Here we review the role of chemokines in the regulation of CCA development and progression, and the modulation of angiogenesis, metastasis and immune control. The potential role of chemokines and their receptors as possible biomarkers and/or therapeutic targets for hepatobiliary cancer is also discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Fabio Marra
- Correspondence: (F.M.); (A.G.); Tel.: +39-055-2758095 or +39-055-2758498 or +39-055-2758499 (F.M.); +39-055-2751801 (A.G.)
| | - Alessandra Gentilini
- Correspondence: (F.M.); (A.G.); Tel.: +39-055-2758095 or +39-055-2758498 or +39-055-2758499 (F.M.); +39-055-2751801 (A.G.)
| |
Collapse
|
171
|
Huynh C, Dingemanse J, Meyer Zu Schwabedissen HE, Sidharta PN. Relevance of the CXCR4/CXCR7-CXCL12 axis and its effect in pathophysiological conditions. Pharmacol Res 2020; 161:105092. [PMID: 32758634 DOI: 10.1016/j.phrs.2020.105092] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 02/07/2023]
Abstract
The impact of the C-X-C receptor (CXCR) 7 and its close co-player CXCR4 in different physiological and pathophysiological processes has been extensively investigated within the last decades. Following activation by their shared ligand C-X-C ligand (CXCL) 12, both chemokine receptors can induce various routes of cell signaling and/or scavenge CXCL12 from the extracellular environment. This contributes to organ development and maintenance of homeostasis. Alterations of the CXCR4/CXCR7-CXCL12 axis have been detected in diseases such as cancer, central nervous system and cardiac disorders, and autoimmune diseases. These alterations include changes of the expression pattern, distribution, or downstream effects. The progression of the diseases can be regulated in preclinical models by the use of various modulators suggesting that this axis serves as a promising therapeutic target. It is therefore of great interest to investigate CXCR4/CXCR7/CXCL12 modulators in clinical development, with several CXCR4 and CXCL12 modulators such as plerixafor, ulocuplumab, balixafortide, and olaptesed pegol having already reached this stage. An overview is presented of the most important diseases whose outcomes can be positively or negatively regulated by the CXCR4/CXCR7-CXCL12 axis and summarizes preclinical and clinical data of modulators of that axis. Contrary to CXCR4 and CXCL12 modulators, CXCR7 modulators have, thus far, not been extensively studied. Therefore, more (pre)clinical investigations are needed.
Collapse
Affiliation(s)
- Christine Huynh
- Idorsia Pharmaceuticals Ltd, Department of Clinical Pharmacology, Hegenheimermattweg 91, 4123 Allschwil, Switzerland; Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Jasper Dingemanse
- Idorsia Pharmaceuticals Ltd, Department of Clinical Pharmacology, Hegenheimermattweg 91, 4123 Allschwil, Switzerland
| | | | - Patricia N Sidharta
- Idorsia Pharmaceuticals Ltd, Department of Clinical Pharmacology, Hegenheimermattweg 91, 4123 Allschwil, Switzerland.
| |
Collapse
|
172
|
DeCordova S, Shastri A, Tsolaki AG, Yasmin H, Klein L, Singh SK, Kishore U. Molecular Heterogeneity and Immunosuppressive Microenvironment in Glioblastoma. Front Immunol 2020; 11:1402. [PMID: 32765498 PMCID: PMC7379131 DOI: 10.3389/fimmu.2020.01402] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumor in adults, with a poor prognosis, despite surgical resection combined with radio- and chemotherapy. The major clinical obstacles contributing to poor GBM prognosis are late diagnosis, diffuse infiltration, pseudo-palisading necrosis, microvascular proliferation, and resistance to conventional therapy. These challenges are further compounded by extensive inter- and intra-tumor heterogeneity and the dynamic plasticity of GBM cells. The complex heterogeneous nature of GBM cells is facilitated by the local inflammatory tumor microenvironment, which mostly induces tumor aggressiveness and drug resistance. An immunosuppressive tumor microenvironment of GBM provides multiple pathways for tumor immune evasion. Infiltrating immune cells, mostly tumor-associated macrophages, comprise much of the non-neoplastic population in GBM. Further understanding of the immune microenvironment of GBM is essential to make advances in the development of immunotherapeutics. Recently, whole-genome sequencing, epigenomics and transcriptional profiling have significantly helped improve the prognostic and therapeutic outcomes of GBM patients. Here, we discuss recent genomic advances, the role of innate and adaptive immune mechanisms, and the presence of an established immunosuppressive GBM microenvironment that suppresses and/or prevents the anti-tumor host response.
Collapse
Affiliation(s)
- Syreeta DeCordova
- Biosciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Abhishek Shastri
- Central and North West London NHS Foundation Trust, London, United Kingdom
| | - Anthony G Tsolaki
- Biosciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, India
| | - Lukas Klein
- Department of Gastroenterology and Gastroenterology Oncology, University Medical Centre, Göttingen, Germany
| | - Shiv K Singh
- Department of Gastroenterology and Gastroenterology Oncology, University Medical Centre, Göttingen, Germany
| | - Uday Kishore
- Biosciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| |
Collapse
|
173
|
Chemokine receptor CXCR7 non-cell-autonomously controls pontine neuronal migration and nucleus formation. Sci Rep 2020; 10:11830. [PMID: 32678266 PMCID: PMC7367352 DOI: 10.1038/s41598-020-68852-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/30/2020] [Indexed: 11/28/2022] Open
Abstract
Long distance tangential migration transports neurons from their birth places to distant destinations to be incorporated into neuronal circuits. How neuronal migration is guided during these long journeys is still not fully understood. We address this issue by studying the migration of pontine nucleus (PN) neurons in the mouse hindbrain. PN neurons migrate from the lower rhombic lip first anteriorly and then turn ventrally near the trigeminal ganglion root towards the anterior ventral hindbrain. Previously we showed that in mouse depleted of chemokine receptor CXCR4 or its ligand CXCL12, PN neurons make their anterior-to-ventral turn at posteriorized positions. However, the mechanism that spatiotemporally controls the anterior-to-ventral turning is still unclear. Furthermore, the role of CXCR7, the atypical receptor of CXCL12, in pontine migration has yet to be examined. Here, we find that the PN is elongated in Cxcr7 knockout due to a broadened anterior-to-ventral turning positions. Cxcr7 is not expressed in migrating PN neurons en route to their destinations, but is strongly expressed in the pial meninges. Neuroepithelium-specific knockout of Cxcr7 does not recapitulate the PN phenotype in Cxcr7 knockout, suggesting that CXCR7 acts non-cell-autonomously possibly from the pial meninges. We show further that CXCR7 regulates pontine migration by modulating CXCL12 protein levels.
Collapse
|
174
|
Xin Q, Sun Q, Zhang CS, Zhang Q, Li CJ. Functions and mechanisms of chemokine receptor 7 in tumors of the digestive system. World J Clin Cases 2020; 8:2448-2463. [PMID: 32607322 PMCID: PMC7322425 DOI: 10.12998/wjcc.v8.i12.2448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/12/2020] [Accepted: 06/02/2020] [Indexed: 02/05/2023] Open
Abstract
Chemokine (C-X-C motif) receptor 7 (CXCR7), recently termed ACKR3, belongs to the G protein-coupled cell surface receptor family, binds to stromal cell-derived factor-1 [SDF-1, or chemokine (C-X-C motif) ligand 12] or chemokine (C-X-C motif) ligand 11, and is the most common chemokine receptor expressed in a variety of cancer cells. SDF-1 binds to its receptor chemokine (C-X-C motif) receptor 4 (CXCR4) and regulates cell proliferation, survival, angiogenesis and migration. In recent years, another new receptor for SDF-1, CXCR7, has been discovered, and CXCR7 has also been found to be expressed in a variety of tumor cells and tumor-related vascular endothelial cells. Many studies have shown that CXCR7 can promote the growth and metastasis of a variety of malignant tumor cells. Unlike CXCR4, CXCR7 exhibits a slight modification in the DRYLAIV motif and does not induce intracellular Ca2+ release following ligand binding, which is essential for recruiting and activating G proteins. CXCR7 is generally thought to work in three ways: (1) Recruiting β-arrestin 2; (2) Heterodimerizing with CXCR4; and (3) Acting as a “scavenger” of SDF-1, thus lowering the level of SDF-1 to weaken the activity of CXCR4. In the present review, the expression and role of CXCR7, as well as its prognosis in cancers of the digestive system, were investigated.
Collapse
Affiliation(s)
- Qi Xin
- Department of Pathology, Tianjin Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Quan Sun
- Department of Hepatobiliary Surgery, Tianjin Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
| | - Chuan-Shan Zhang
- Department of Pathology, Tianjin Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
| | - Qin Zhang
- Department of Pathology, Tianjin Third Central Hospital, The Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin 300170, China
| | - Chun-Jun Li
- Department of Endocrinology, Health Management Center, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin 300121, China
| |
Collapse
|
175
|
Meyrath M, Szpakowska M, Zeiner J, Massotte L, Merz MP, Benkel T, Simon K, Ohnmacht J, Turner JD, Krüger R, Seutin V, Ollert M, Kostenis E, Chevigné A. The atypical chemokine receptor ACKR3/CXCR7 is a broad-spectrum scavenger for opioid peptides. Nat Commun 2020; 11:3033. [PMID: 32561830 PMCID: PMC7305236 DOI: 10.1038/s41467-020-16664-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 05/15/2020] [Indexed: 12/29/2022] Open
Abstract
Endogenous opioid peptides and prescription opioid drugs modulate pain, anxiety and stress by activating opioid receptors, currently classified into four subtypes. Here we demonstrate that ACKR3/CXCR7, hitherto known as an atypical scavenger receptor for chemokines, is a broad-spectrum scavenger of opioid peptides. Phylogenetically, ACKR3 is intermediate between chemokine and opioid receptors and is present in various brain regions together with classical opioid receptors. Functionally, ACKR3 is a scavenger receptor for a wide variety of opioid peptides, especially enkephalins and dynorphins, reducing their availability for the classical opioid receptors. ACKR3 is not modulated by prescription opioids, but we show that an ACKR3-selective subnanomolar competitor peptide, LIH383, can restrain ACKR3’s negative regulatory function on opioid peptides in rat brain and potentiate their activity towards classical receptors, which may open alternative therapeutic avenues for opioid-related disorders. Altogether, our results reveal that ACKR3 is an atypical opioid receptor with cross-family ligand selectivity. Opioids modulate pain, anxiety and stress by activating four subtypes of opioid receptors. The authors show that atypical chemokine receptor 3 (ACKR3) is a scavenger for various endogenous opioid peptides regulating their availability without activating downstream signaling.
Collapse
Affiliation(s)
- Max Meyrath
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Martyna Szpakowska
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Julian Zeiner
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Laurent Massotte
- Neurophysiology Unit, GIGA Neurosciences, University of Liège, avenue de l'hopital, B-4000, Liège, Belgium
| | - Myriam P Merz
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Tobias Benkel
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany.,Research Training Group 1873, University of Bonn, Bonn, Germany
| | - Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Jochen Ohnmacht
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg.,Department of Life Sciences and Medicine, University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg
| | - Jonathan D Turner
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg.,Transversal Translational Medicine, Luxembourg Institute of Health (LIH), rue Thomas Edison 1A-B, L-1445, Strassen, Luxembourg
| | - Vincent Seutin
- Neurophysiology Unit, GIGA Neurosciences, University of Liège, avenue de l'hopital, B-4000, Liège, Belgium
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, 5000, Odense, Denmark
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Andy Chevigné
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
176
|
CXCR7 Inhibits Fibrosis via Wnt/ β-Catenin Pathways during the Process of Angiogenesis in Human Umbilical Vein Endothelial Cells. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1216926. [PMID: 32566651 PMCID: PMC7293734 DOI: 10.1155/2020/1216926] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/01/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023]
Abstract
Although SDF-1/CXCR7 plays an important role in angiogenesis, the function and the pathway of the SDF-1/CXCR7 axis might depend on the cell type or tissue origin and not fully understood. In this study, we investigated the effect of CXCR7 in SDF-1-induced proliferation, migration, apoptosis, tube formation, and endothelial-to-mesenchymal transition (EndMT) of human umbilical vein endothelial cells (HUVECs), and the potential pathway of SDF-1/CXCR7. We confirmed that the silencing of CXCR7 inhibited the proliferation of HUVECs and contributed the apoptosis, while overexpressed CXCR7 increased SDF-1-induced HUVECs migration and tube formation. However, upregulated CXCR7 inhibited the expression of α-SMA, suggesting that CXCR7 might attenuate EndMT. In addition, overexpressed CXCR7 activated AKT and ERK signaling pathways but suppressed Wnt/β-catenin pathways in HUVECs. The inhibition of Wnt/β-catenin pathways decreased the expression of α-SMA. Altogether, these results suggest that CXCR7 might inhibit fibrosis via Wnt/β-catenin pathways during the process of angiogenesis.
Collapse
|
177
|
Wan Y, Zhang C, Xu Y, Wang M, Rao Q, Xing H, Tian Z, Tang K, Mi Y, Wang Y, Wang J. Hyperfunction of CD4 CD25 regulatory T cells in de novo acute myeloid leukemia. BMC Cancer 2020; 20:472. [PMID: 32456622 PMCID: PMC7249438 DOI: 10.1186/s12885-020-06961-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is a common hematopoietic malignancy that has a high relapse rate, and the number of regulatory T cells (Tregs) in AML patients is significantly increased. The aim of this study was to clarify the role of Tregs in the immune escape of acute myeloid leukemia. Methods The frequencies of Tregs and the expression of PD-1, CXCR4 and CXCR7 were examined by flow cytometry. The expression of CTLA-4 and GITR was tested by MFI. Chemotaxis assays were performed to evaluate Treg migration. The concentrations of SDF-1α, IFN-γ and TNF-α were examined by ELISA. Coculture and crisscross coculture experiments were performed to examine Treg proliferation and apoptosis and the effect of regulatory B cells (Breg) conversion. Results The frequencies of Tregs in peripheral blood and bone marrow in AML patients were increased compared with those in healthy participants. AML Tregs had robust migration towards bone marrow due to increased expression of CXCR4. AML Treg-mediated immunosuppression of T cells was achieved through proliferation inhibition, apoptosis promotion and suppression of IFN-γ production in CD4+CD25− T cells. AML Bregs induced the conversion of CD4+CD25−T cells to Tregs. Conclusion In AML patients, the Breg conversion effect and robust CXCR4-induced migration led to Treg enrichment in bone marrow. AML Tregs downregulated the function of CD4+CD25− T cells, contributing to immune escape.
Collapse
Affiliation(s)
- Yuling Wan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 288, Nanjing Road, Tianjin, 300020, China
| | - Congxiao Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 288, Nanjing Road, Tianjin, 300020, China
| | - Yingxi Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 288, Nanjing Road, Tianjin, 300020, China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 288, Nanjing Road, Tianjin, 300020, China
| | - Qing Rao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 288, Nanjing Road, Tianjin, 300020, China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 288, Nanjing Road, Tianjin, 300020, China
| | - Zheng Tian
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 288, Nanjing Road, Tianjin, 300020, China
| | - Kejing Tang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 288, Nanjing Road, Tianjin, 300020, China
| | - Yingchang Mi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 288, Nanjing Road, Tianjin, 300020, China
| | - Ying Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 288, Nanjing Road, Tianjin, 300020, China.
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 288, Nanjing Road, Tianjin, 300020, China.
| |
Collapse
|
178
|
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
|
179
|
Ayakannu T, Taylor AH, Konje JC. Selection of Endogenous Control Reference Genes for Studies on Type 1 or Type 2 Endometrial Cancer. Sci Rep 2020; 10:8468. [PMID: 32439920 PMCID: PMC7242460 DOI: 10.1038/s41598-020-64663-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/02/2020] [Indexed: 02/02/2023] Open
Abstract
A panel of 32 candidate reference genes was used to identify the most stable genes for gene normalisation in quantitative RT-PCR studies using endometrial biopsies obtained from women with endometrial cancer (type 1 or type 2) and without cancer (controls). RNA from the biopsies was isolated, examined for purity and quality, and then reverse transcribed into cDNA before being subjected to real-time qRT-PCR analysis in triplicate within the TaqMan gene Expression Assay kit. The most 'stable' endogenous control genes were then identified using the geNorm qbase + 2 and NormFinder software packages. PSMC4, PUM1 and IPO8 were identified as the best reference genes combination for type 1 endometrial cancer (grades 1, 2 and 3), whereas for type 2 endometrial cancer (serous and carcinosarcoma), UBC, MRPL19, PGK1 and PPIA were the best reference genes combination. We conclude that the use of these normaliser combinations should provide accurate interpretation of gene expression at the transcript level in endometrial cancer studies especially for types 1 and 2 cancers.
Collapse
Affiliation(s)
- Thangesweran Ayakannu
- Faculty of Health and Life Sciences, University of Liverpool, Thompson Yates Brownlow Hill, Liverpool, L69 3GB, UK.
- Reproductive Sciences Section, Department of Cancer Studies and Molecular Medicine, University of Leicester, Leicester, UK.
- Gynaecology Oncology Cancer Centre, Liverpool Women's NHS Foundation Trust, Liverpool Women's Hospital, Liverpool, UK.
| | - Anthony H Taylor
- Faculty of Health and Life Sciences, University of Liverpool, Thompson Yates Brownlow Hill, Liverpool, L69 3GB, UK
- Department of Molecular and Cell Biology, University of Leicester, Leicester, Leicestershire, UK
| | - Justin C Konje
- Faculty of Health and Life Sciences, University of Liverpool, Thompson Yates Brownlow Hill, Liverpool, L69 3GB, UK
- Department of Obstetrics and Gynaecology, Sidra Medicine, Women's Wellness and Research Center, HMC, Doha, Qatar
| |
Collapse
|
180
|
Jaracz-Ros A, Bernadat G, Cutolo P, Gallego C, Gustavsson M, Cecon E, Baleux F, Kufareva I, Handel TM, Bachelerie F, Levoye A. Differential activity and selectivity of N-terminal modified CXCL12 chemokines at the CXCR4 and ACKR3 receptors. J Leukoc Biol 2020; 107:1123-1135. [PMID: 32374043 DOI: 10.1002/jlb.2ma0320-383rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 03/12/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Chemokines play critical roles in numerous physiologic and pathologic processes through their action on seven-transmembrane (TM) receptors. The N-terminal domain of chemokines, which is a key determinant of signaling via its binding within a pocket formed by receptors' TM helices, can be the target of proteolytic processing. An illustrative case of this regulatory mechanism is the natural processing of CXCL12 that generates chemokine variants lacking the first two N-terminal residues. Whereas such truncated variants behave as antagonists of CXCR4, the canonical G protein-coupled receptor of CXCL12, they are agonists of the atypical chemokine receptor 3 (ACKR3/CXCR7), suggesting the implication of different structural determinants in the complexes formed between CXCL12 and its two receptors. Recent analyses have suggested that the CXCL12 N-terminus first engages the TM helices of ACKR3 followed by the receptor N-terminus wrapping around the chemokine core. Here we investigated the first stage of ACKR3-CXCL12 interactions by comparing the activity of substituted or N-terminally truncated variants of CXCL12 toward CXCR4 and ACKR3. We showed that modification of the first two N-terminal residues of the chemokine (K1R or P2G) does not alter the ability of CXCL12 to activate ACKR3. Our results also identified the K1R variant as a G protein-biased agonist of CXCR4. Comparative molecular dynamics simulations of the complexes formed by ACKR3 either with CXCL12 or with the P2G variant identified interactions between the N-terminal 2-4 residues of CXCL12 and a pocket formed by receptor's TM helices 2, 6, and 7 as critical determinants for ACKR3 activation.
Collapse
Affiliation(s)
- Agnieszka Jaracz-Ros
- Université Paris-Saclay, INSERM, Inflammation, Microbiome and Immunosurveillance, Clamart, France
| | | | - Pasquale Cutolo
- Université Paris-Saclay, INSERM, Inflammation, Microbiome and Immunosurveillance, Clamart, France
| | - Carmen Gallego
- Université Paris-Saclay, INSERM, Inflammation, Microbiome and Immunosurveillance, Clamart, France
| | - Martin Gustavsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, Jolla, Louisiana, California, USA
| | - Erika Cecon
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Françoise Baleux
- Institut Pasteur, Unité de Chimie des Biomolécules, Paris, France
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, Jolla, Louisiana, California, USA
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, Jolla, Louisiana, California, USA
| | - Françoise Bachelerie
- Université Paris-Saclay, INSERM, Inflammation, Microbiome and Immunosurveillance, Clamart, France
| | - Angélique Levoye
- Université de Paris, PARCC, INSERM, Paris, France.,Université Sorbonne Paris Nord, Bobigny, France
| |
Collapse
|
181
|
Regenbogen S, Stagno MJ, Schleicher S, Schilbach K, Bösmüller H, Fuchs J, Schmid E, Seitz G. Cytotoxic drugs in combination with the CXCR4 antagonist AMD3100 as a potential treatment option for pediatric rhabdomyosarcoma. Int J Oncol 2020; 57:289-300. [PMID: 32377699 DOI: 10.3892/ijo.2020.5059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 03/26/2020] [Indexed: 11/05/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common type of pediatric soft tissue sarcoma. The prognosis of advanced stage RMS remains poor, and metastatic invasion is a major cause of treatment failure. Therefore, there is an urgent need for treatment alternatives focusing on metastatic invasion and drug resistance. The stromal cell‑derived factor‑1 (SDF‑1)/chemokine receptor 4 (CXCR4) axis is a crucial factor for metastatic invasion in RMS. Clinical data has revealed that high CXCR4 expression is associated with a poor outcome and a high metastatic rate in several malignancies, including RMS. Thus, targeting CXCR4 in addition to classical chemotherapy may improve the effectiveness of RMS treatment. In the present study, flow cytometry and reverse transcription‑quantitative PCR were used to assess the effects of the combined treatment with a CXCR4 antagonist and chemotherapy on CXCR4 expression in the embryonal RMS (RME) cell line RD and in the alveolar RMS (RMA) cell line RH30. The functional effect of CXCR4 expression on the migratory behavior of RMS cells was analyzed using Transwell assays. Treatment with cytotoxic agents modulated CXCR4 expression in RMS cells in a dose‑, drug‑ and cell line dependent manner; however, this was not observed in RD cells with vincristine. The expression levels of CXCR4 significantly increased the migratory behavior of RMA and did not affect RME cell migration towards stromal cell‑derived factor‑1α (SDF‑1α). AMD3100 markedly reduced the migration of RH30 cells in the Transwell assays compared with SDF‑1α alone, and the cytotoxic agents doxorubicin and vincristine increased this effect. The results of the combined treatment in RMS cells using the CXCR4 antagonist AMD3100 together with cytotoxic drugs demonstrated that this approach may be a promising alternative for the treatment of advanced stage pediatric RMS. The observed effects of circumventing metastatic invasion and drug resistance should be further investigated in vivo.
Collapse
Affiliation(s)
- Stephan Regenbogen
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, D‑72076 Tuebingen, Germany
| | - Matias Julian Stagno
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, D‑72076 Tuebingen, Germany
| | - Sabine Schleicher
- Department of Haematology and Oncology, University Children's Hospital, D‑72076 Tuebingen, Germany
| | - Karin Schilbach
- Department of Haematology and Oncology, University Children's Hospital, D‑72076 Tuebingen, Germany
| | - Hans Bösmüller
- Department of Pathology, University Hospital Tuebingen, D‑72076 Tuebingen, Germany
| | - Jörg Fuchs
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, D‑72076 Tuebingen, Germany
| | - Evi Schmid
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, D‑72076 Tuebingen, Germany
| | - Guido Seitz
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, D‑72076 Tuebingen, Germany
| |
Collapse
|
182
|
Atypical chemokine receptor ACKR3/CXCR7 controls postnatal vasculogenesis and arterial specification by mesenchymal stem cells via Notch signaling. Cell Death Dis 2020; 11:307. [PMID: 32366833 PMCID: PMC7198625 DOI: 10.1038/s41419-020-2512-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) are known to play a role in postnatal vasculogenesis and hold great promise for vascular regeneration. However, the mechanisms by which the endothelial differentiation and specification of MSCs remain unclear. We examined the potential role and molecular mechanisms of atypical chemokine receptor ACKR3/CXCR7 in MSC-mediated endothelial cell differentiation and specification. Here, we showed that vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) activate CXCR7 expression on MSCs through PDGF receptors, PDGFRα and PDGFRβ-mediated phosphoinositide 3-kinase (PI3K)/Akt signaling. Genetic and pharmacologic blockage of CXCR7 on MSCs suppressed the VEGF or stromal cell-derived factor 1 (SDF)-1-induced the capacity for vasculogenesis in vitro and in vivo. Moreover, CXCR7 gain of function markedly promoted vasculogenesis by MSCs in vitro and in vivo and induced endothelial differentiation along the arterial endothelial cell lineage via upregulation of Notch signaling. However, blockade of Notch signaling inhibited CXCR7-induced vasculogensis by MSCs. These results indicate CXCR7 is a critical regulator of MSC-mediated postnatal vasculogenesis and arterial specification via Notch signaling.
Collapse
|
183
|
Saaber F, Schütz D, Miess E, Abe P, Desikan S, Ashok Kumar P, Balk S, Huang K, Beaulieu JM, Schulz S, Stumm R. ACKR3 Regulation of Neuronal Migration Requires ACKR3 Phosphorylation, but Not β-Arrestin. Cell Rep 2020; 26:1473-1488.e9. [PMID: 30726732 DOI: 10.1016/j.celrep.2019.01.049] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/23/2018] [Accepted: 01/11/2019] [Indexed: 12/11/2022] Open
Abstract
Phosphorylation of heptahelical receptors is thought to regulate G protein signaling, receptor endocytosis, and non-canonical signaling via recruitment of β-arrestins. We investigated chemokine receptor functionality under phosphorylation-deficient and β-arrestin-deficient conditions by studying interneuron migration in the embryonic cortex. This process depends on CXCL12, CXCR4, G protein signaling and on the atypical CXCL12 receptor ACKR3. We found that phosphorylation was crucial, whereas β-arrestins were dispensable for ACKR3-mediated control of CXCL12 levels in vivo. Cortices of mice expressing phosphorylation-deficient ACKR3 exhibited a major interneuron migration defect, which was accompanied by excessive activation and loss of CXCR4. Cxcl12-overexpressing mice mimicked this phenotype. Excess CXCL12 caused lysosomal CXCR4 degradation, loss of CXCR4 responsiveness, and, ultimately, similar motility defects as Cxcl12 deficiency. By contrast, β-arrestin deficiency caused only a subtle migration defect mimicked by CXCR4 gain of function. These findings demonstrate that phosphorylation regulates atypical chemokine receptor function without β-arrestin involvement in chemokine sequestration and non-canonical signaling.
Collapse
Affiliation(s)
- Friederike Saaber
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Dagmar Schütz
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Elke Miess
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Philipp Abe
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Srinidhi Desikan
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Praveen Ashok Kumar
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Sara Balk
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Ke Huang
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Jean Martin Beaulieu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto M5S 1A8, ON, Canada
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Ralf Stumm
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany.
| |
Collapse
|
184
|
New Insights on the Emerging Genomic Landscape of CXCR4 in Cancer: A Lesson from WHIM. Vaccines (Basel) 2020; 8:vaccines8020164. [PMID: 32260318 PMCID: PMC7349554 DOI: 10.3390/vaccines8020164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/16/2022] Open
Abstract
Deciphering the molecular alterations leading to disease initiation and progression is currently crucial to identify the most relevant targets for precision therapy in cancer patients. Cancers express a complex chemokine network influencing leucocyte infiltration and angiogenesis. Moreover, malignant cells also express a selective repertoire of chemokine receptors that sustain their growth and spread. At present, different cancer types have been shown to overexpress C-X-C chemokine receptor type 4 (CXCR4) and to respond to its ligand C-X-C motif chemokine 12 (CXCL12). The CXCL12/CXCR4 axis influences cancer biology, promoting survival, proliferation, and angiogenesis, and plays a pivotal role in directing migration of cancer cells to sites of metastases, making it a prognostic marker and a therapeutic target. More recently, mutations in the C-terminus of CXCR4 have been identified in the genomic landscape of patients affected by Waldenstrom's macroglobulinemia, a rare B cell neoplasm. These mutations closely resemble those occurring in Warts, Hypogammaglobulinemia, Immunodeficiency, and Myelokathexis (WHIM) syndrome, an immunodeficiency associated with CXCR4 aberrant expression and activity and with chemotherapy resistance in clinical trials. In this review, we summarize the current knowledge on the relevance of CXCR4 mutations in cancer biology, focusing on its importance as predictors of clinical presentation and response to therapy.
Collapse
|
185
|
Serafin DS, Harris NR, Nielsen NR, Mackie DI, Caron KM. Dawn of a New RAMPage. Trends Pharmacol Sci 2020; 41:249-265. [PMID: 32115276 PMCID: PMC7236817 DOI: 10.1016/j.tips.2020.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 01/08/2023]
Abstract
Receptor activity-modifying proteins (RAMPs) interact with G-protein-coupled receptors (GPCRs) to modify their functions, imparting significant implications upon their physiological and therapeutic potentials. Resurging interest in identifying RAMP-GPCR interactions has recently been fueled by coevolution studies and orthogonal technological screening platforms. These new studies reveal previously unrecognized RAMP-interacting GPCRs, many of which expand beyond Class B GPCRs. The consequences of these interactions on GPCR function and physiology lays the foundation for new molecular therapeutic targets, as evidenced by the recent success of erenumab. Here, we highlight recent papers that uncovered novel RAMP-GPCR interactions, human RAMP-GPCR disease-causing mutations, and RAMP-related human pathologies, paving the way for a new era of RAMP-targeted drug development.
Collapse
Affiliation(s)
- D Stephen Serafin
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie R Harris
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Duncan I Mackie
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
186
|
CXCR7 suppression modulates macrophage phenotype and function to ameliorate post-myocardial infarction injury. Inflamm Res 2020; 69:523-532. [DOI: 10.1007/s00011-020-01335-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/25/2020] [Accepted: 03/07/2020] [Indexed: 12/22/2022] Open
|
187
|
Zargari R, Mahdifar M, Mohammadi A, Vahidi Z, Hassanshahi G, Rafatpanah H. The Role of Chemokines in the Pathogenesis of HTLV-1. Front Microbiol 2020; 11:421. [PMID: 32231656 PMCID: PMC7083101 DOI: 10.3389/fmicb.2020.00421] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/27/2020] [Indexed: 12/16/2022] Open
Abstract
Human T cell leukemia virus type 1 (HTLV-1) is a human retrovirus that is associated with two main diseases: HTLV-associated myelopathy/tropical spastic paraparesis (HAM/TSP) and adult T cell leukemia/lymphoma (ATL). Chemokines are highly specialized groups of cytokines that play important roles in organizing, trafficking, homing, and in the migration of immune cells to the bone marrow, lymphoid organs and sites of infection and inflammation. Aberrant expression or function of chemokines, or their receptors, has been linked to the protection against or susceptibility to specific infectious diseases, as well as increased the risk of autoimmune diseases and malignancy. Chemokines and their receptors participate in pathogenesis of HTLV-1 associated diseases from inflammation in the central nervous system (CNS) which occurs in cases of HAM/TSP to T cell immortalization and tissue infiltration observed in ATL patients. Chemokines represent viable effective prognostic biomarkers for HTLV-1-associated diseases which provide the early identification of high-risk, treatment possibilities and high-yielding clinical trials. This review focuses on the emerging roles of these molecules in the outcome of HTLV-1-associated diseases.
Collapse
Affiliation(s)
- Razieh Zargari
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Mahdifar
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asadollah Mohammadi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Zohreh Vahidi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Houshang Rafatpanah
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
188
|
Whitman MC, Miyake N, Nguyen EH, Bell JL, Matos Ruiz PM, Chan WM, Di Gioia SA, Mukherjee N, Barry BJ, Bosley TM, Khan AO, Engle EC. Decreased ACKR3 (CXCR7) function causes oculomotor synkinesis in mice and humans. Hum Mol Genet 2020; 28:3113-3125. [PMID: 31211835 DOI: 10.1093/hmg/ddz137] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/08/2019] [Accepted: 06/10/2019] [Indexed: 01/17/2023] Open
Abstract
Oculomotor synkinesis is the involuntary movement of the eyes or eyelids with a voluntary attempt at a different movement. The chemokine receptor CXCR4 and its ligand CXCL12 regulate oculomotor nerve development; mice with loss of either molecule have oculomotor synkinesis. In a consanguineous family with congenital ptosis and elevation of the ptotic eyelid with ipsilateral abduction, we identified a co-segregating homozygous missense variant (c.772G>A) in ACKR3, which encodes an atypical chemokine receptor that binds CXCL12 and functions as a scavenger receptor, regulating levels of CXCL12 available for CXCR4 signaling. The mutant protein (p.V258M) is expressed and traffics to the cell surface but has a lower binding affinity for CXCL12. Mice with loss of Ackr3 have variable phenotypes that include misrouting of the oculomotor and abducens nerves. All embryos show oculomotor nerve misrouting, ranging from complete misprojection in the midbrain, to aberrant peripheral branching, to a thin nerve, which aberrantly innervates the lateral rectus (as seen in Duane syndrome). The abducens nerve phenotype ranges from complete absence, to aberrant projections within the orbit, to a normal trajectory. Loss of ACKR3 in the midbrain leads to downregulation of CXCR4 protein, consistent with reports that excess CXCL12 causes ligand-induced degradation of CXCR4. Correspondingly, excess CXCL12 applied to ex vivo oculomotor slices causes axon misrouting, similar to inhibition of CXCR4. Thus, ACKR3, through its regulation of CXCL12 levels, is an important regulator of axon guidance in the oculomotor system; complete loss causes oculomotor synkinesis in mice, while reduced function causes oculomotor synkinesis in humans.
Collapse
Affiliation(s)
- Mary C Whitman
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Noriko Miyake
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Elaine H Nguyen
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Jessica L Bell
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Paola M Matos Ruiz
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Wai-Man Chan
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Boston Children's Hospital, Boston, MA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Silvio Alessandro Di Gioia
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Nisha Mukherjee
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Brenda J Barry
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Boston Children's Hospital, Boston, MA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - T M Bosley
- Department of Ophthalmology, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| | - Arif O Khan
- Division of Pediatric Ophthalmology, King Khaled Eye Specialist Hospital, Riyadh, Saudi Arabia
| | - Elizabeth C Engle
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Boston Children's Hospital, Boston, MA, USA.,Department of Neurology, Harvard Medical School, Boston, MA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
189
|
Lau S, Feitzinger A, Venkiteswaran G, Wang J, Lewellis SW, Koplinski CA, Peterson FC, Volkman BF, Meier-Schellersheim M, Knaut H. A negative-feedback loop maintains optimal chemokine concentrations for directional cell migration. Nat Cell Biol 2020; 22:266-273. [PMID: 32042179 PMCID: PMC7809593 DOI: 10.1038/s41556-020-0465-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022]
Abstract
Chemoattractant gradients frequently guide migrating cells. To achieve the most directional signal, such gradients should be maintained with concentrations around the dissociation constant (Kd)1-6 of the chemoreceptor. Whether this actually occurs in animals is unknown. Here we investigate whether a moving tissue, the zebrafish posterior lateral line primordium, buffers its attractant in this concentration range to achieve robust migration. We find that the Cxcl12 (also known as Sdf1) attractant gradient ranges from 0 to 12 nM, values similar to the 3.4 nM Kd of its receptor Cxcr4. When we increase the Kd of Cxcl12 for Cxcr4, primordium migration is less directional. Furthermore, a negative-feedback loop between Cxcl12 and its clearance receptor Ackr3 (also known as Cxcr7) regulates the Cxcl12 concentrations. Breaking this negative feedback by blocking the phosphorylation of the cytoplasmic tail of Ackr3 also results in less directional primordium migration. Thus, directed migration of the primordium is dependent on a close match between the Cxcl12 concentration and the Kd of Cxcl12 for Cxcr4, which is maintained by buffering of the chemokine levels. Quantitative modelling confirms the plausibility of this mechanism. We anticipate that buffering of attractant concentration is a general mechanism for ensuring robust cell migration.
Collapse
Affiliation(s)
- Stephanie Lau
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - Anna Feitzinger
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - Gayatri Venkiteswaran
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - John Wang
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - Stephen W Lewellis
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - Chad A Koplinski
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Francis C Peterson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Martin Meier-Schellersheim
- Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Holger Knaut
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
190
|
Lounsbury N. Advances in CXCR7 Modulators. Pharmaceuticals (Basel) 2020; 13:ph13020033. [PMID: 32098047 PMCID: PMC7169404 DOI: 10.3390/ph13020033] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
CXC chemokine receptor 7 (CXCR7) is a G-protein-coupled receptor that signals through the β-arrestin pathway. Its ligands include interferon-inducible T cell α chemoattractant (CXCL11) and stromal cell-derived factor-1 (CXCL12). It interacts with CXCR4, and the two are associated with various cancers, as well as other disease states such as coronary artery disease, stroke, inflammation and human immunodeficiency virus (HIV). Antibodies and small interfering RNA (siRNA) have shown the utility of antagonists of CXCR7 in these disease states. Although some small molecules were initially reported as antagonists due to their displayed activity, many function as agonists while still producing the desired pharmacologic effects. A potential reason for this contradiction is that effects may be due to elevated extracellular CXCL12 levels.
Collapse
Affiliation(s)
- Nicole Lounsbury
- Department of Pharmaceutical Sciences, Larkin University College of Pharmacy, Miami, FL 33169, USA
| |
Collapse
|
191
|
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
|
192
|
Expression and Functional Analysis of CXCL12 and Its Receptors in Human Term Trophoblast Cells. Reprod Sci 2020; 27:46-54. [PMID: 32046406 DOI: 10.1007/s43032-019-00134-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/17/2019] [Indexed: 12/20/2022]
Abstract
Chemokine CXCL12 and its receptors CXCR4/CXCR7 play a pivotal role in many physiological and pathological situations, while the expression and function in human term trophoblast cells remain largely unknown. In the study, the expression and function of CXCL12 and its receptors CXCR4/CXCR7 in human term trophoblast cells were investigated. Immunocytochemistry and flow cytometry showed that the expression of CXCL12/CXCR4/CXCR7 could be detected in term trophoblast cells while expression level differed. The secretion of CXCL12 in human term trophoblast cells was confirmed by enzyme-linked immunosorbent assay (ELISA). In order to reveal the function of CXCL12, exogenetic recombinant human CXCL12 protein (rhCXCL12) was added to the cultured term trophoblast cells; results showed that cell proliferation ability was increased while cell apoptosis rate was decreased. Moreover, the effects of rhCXCL12 on term trophoblast cells could be diminished or attenuated by antibodies against CXCL12, CXCR4, or CXCR7, respectively. Therefore, these results revealed the important role of CXCL12 on human term trophoblast cells. Our study will provide new insights into understanding the role of CXCL12 on human term trophoblast cells.
Collapse
|
193
|
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
|
194
|
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
|
195
|
Pluchino N, Mamillapalli R, Shaikh S, Habata S, Tal A, Gaye M, Taylor HS. CXCR4 or CXCR7 antagonists treat endometriosis by reducing bone marrow cell trafficking. J Cell Mol Med 2020; 24:2464-2474. [PMID: 31904910 PMCID: PMC7028867 DOI: 10.1111/jcmm.14933] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/29/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022] Open
Abstract
Adult stem cells have a major role in endometrial physiology, including remodelling and repair. However, they also have a critical role in the development and progression of endometriosis. Bone marrow-derived stem cells engraft eutopic endometrium and endometriotic lesions, differentiating to both stromal and epithelial cell fates. Using a mouse bone marrow transplantation model, we show that bone marrow-derived cells engrafting endometriosis express CXCR4 and CXCR7. Targeting either receptor by the administration of small molecule receptor antagonists AMD3100 or CCX771, respectively, reduced BM-derived stem cell recruitment into endometriosis implants. Endometriosis lesion size was decreased compared to vehicle controls after treatment with each antagonist in both an early growth and established lesion treatment model. Endometriosis lesion size was not effected when the local effects of CXCL12 were abrogated using uterine-specific CXCL12 null mice, suggesting an effect primarily on bone marrow cell migration rather than a direct endometrial effect. Antagonist treatment also decreased hallmarks of endometriosis physiopathology such as pro-inflammatory cytokine production and vascularization. CXCR4 and CXCR7 antagonists are potential novel, non-hormonal therapies for endometriosis.
Collapse
Affiliation(s)
- Nicola Pluchino
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Shafiq Shaikh
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Shutaro Habata
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Aya Tal
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Marie Gaye
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
196
|
Wang Y, Zheng B, Xu M, Cai S, Younseo J, Zhang C, Jiang B. Prediction and Analysis of Hub Genes in Renal Cell Carcinoma based on CFS Gene Selection Method Combined with Adaboost Algorithm. Med Chem 2020; 16:654-663. [PMID: 31584378 DOI: 10.2174/1573406415666191004100744] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/04/2019] [Accepted: 08/23/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Renal cell carcinoma (RCC) is the most common malignant tumor of the adult kidney. OBJECTIVE The aim of this study was to identify key genes signatures during RCC and uncover their potential mechanisms. METHODS Firstly, the gene expression profiles of GSE53757 which contained 144 samples, including 72 kidney cancer samples and 72 controls, were downloaded from the GEO database. And then differentially expressed genes (DEGs) between the kidney cancer samples and the controls were identified. After that, GO and KEGG enrichment analyses of DEGs were performed by DAVID. Furthermore, the correlation-based feature subset (CFS) method was applied to the selection of key genes of DEGs. In addition, the classification model between the kidney cancer samples and the controls was built by Adaboost based on the selected key genes. RESULTS 213 DEGs including 80 up-regulated and 133 down-regulated genes were selected as the feature genes to build the classification model between the kidney cancer samples and the controls by CFS method. The accuracy of the classification model by using 5-folds cross-validation test and independent set test is 84.4% and 83.3%, respectively. Besides, TYROBP, CD4163, CAV1, CXCL9, CXCL11 and CXCL13 also can be found in the top 20 hub genes screened by proteinprotein interaction (PPI) network. CONCLUSION It indicated that CFS is a useful tool to identify key genes in kidney cancer. Besides, we also predicted genes such as TYROBP, CD4163, CAV1, CXCL9, CXCL11 and CXCL13 that might target genes to diagnose the kidney cancer.
Collapse
Affiliation(s)
- Yina Wang
- Department of VIP Medical Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Benrong Zheng
- Department of VIP Medical Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Manbin Xu
- Department of Head and Neck Surgery, The Cancer Hospital of Shantou University Medical College, Shantou 515000, China
| | - Shaoping Cai
- Department of Acupuncture and Moxibustion Foshan Hospital of TCM, Foshan 528000, China
| | - Jeong Younseo
- Center for Bioinformatics and Computational Biology, Pai Chai University, Daejeon, South Korea
| | - Chi Zhang
- Huaxia Eye Hospital of Foshan, Huaxia Eye Hospital Group, Foshan, Guangdong, 528000, China
| | - Boxiong Jiang
- Department of VIP Medical Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| |
Collapse
|
197
|
Clauder F, Zitzmann FD, Friebe S, Mayr SG, Robitzki AA, Beck-Sickinger AG. Multifunctional coatings combining bioactive peptides and affinity-based cytokine delivery for enhanced integration of degradable vascular grafts. Biomater Sci 2020; 8:1734-1747. [DOI: 10.1039/c9bm01801h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mussel-derived surface coatings present integrin- and heparin-binding peptides for cell adhesion and modulator protein delivery to improve the endothelialization of biodegradable cardiovascular implants.
Collapse
Affiliation(s)
- Franziska Clauder
- Institute of Biochemistry
- Faculty of Life Sciences
- Leipzig University
- 04103 Leipzig
- Germany
| | - Franziska D. Zitzmann
- Centre for Biotechnology and Biomedicine (BBZ)/Institute of Biochemistry
- Faculty of Life Sciences
- Leipzig University
- 04103 Leipzig
- Germany
| | - Sabrina Friebe
- Leibniz-Institute of Surface Engineering (IOM)
- 04318 Leipzig
- Germany
| | - Stefan G. Mayr
- Leibniz-Institute of Surface Engineering (IOM)
- 04318 Leipzig
- Germany
| | - Andrea A. Robitzki
- Centre for Biotechnology and Biomedicine (BBZ)/Institute of Biochemistry
- Faculty of Life Sciences
- Leipzig University
- 04103 Leipzig
- Germany
| | | |
Collapse
|
198
|
Daniel SK, Seo YD, Pillarisetty VG. The CXCL12-CXCR4/CXCR7 axis as a mechanism of immune resistance in gastrointestinal malignancies. Semin Cancer Biol 2019; 65:176-188. [PMID: 31874281 DOI: 10.1016/j.semcancer.2019.12.007] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/03/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
Single agent checkpoint inhibitor therapy has not been effective for most gastrointestinal solid tumors, but combination therapy with drugs targeting additional immunosuppressive pathways is being attempted. One such pathway, the CXCL12-CXCR4/CXCR7 chemokine axis, has attracted attention due to its effects on tumor cell survival and metastasis as well as immune cell migration. CXCL12 is a small protein that functions in normal hematopoietic stem cell homing in addition to repair of damaged tissue. Binding of CXCL12 to CXCR4 leads to activation of G protein signaling kinases such as P13K/mTOR and MEK/ERK while binding to CXCR7 leads to β-arrestin mediated signaling. While some gastric and colorectal carcinoma cells have been shown to make CXCL12, the primary source in pancreatic cancer and peritoneal metastases is cancer-associated fibroblasts. Binding of CXCL12 to CXCR4 and CXCR7 on tumor cells leads to anti-apoptotic signaling through Bcl-2 and survivin upregulation, as well as promotion of the epithelial-to-mesechymal transition through the Rho-ROCK pathway and alterations in cell adhesion molecules. High levels of CXCL12 seen in the bone marrow, liver, and spleen could partially explain why these are popular sites of metastases for many tumors. CXCL12 is a chemoattractant for lymphocytes at lower levels, but becomes chemorepellant at higher levels; it is unclear exactly what gradient exists in the tumor microenvironment and how this influences tumor-infiltrating lymphocytes. AMD3100 (Plerixafor or Mozobil) is a small molecule CXCR4 antagonist and is the most frequently used drug targeting the CXCL12-CXCR4/CXCR7 axis in clinical trials for gastrointestinal solid tumors currently. Other small molecules and monoclonal antibodies against CXCR4 are being trialed. Further understanding of the CXCL12- CXCR4/CXCR7 chemokine axis in the tumor microenvironment will allow more effective targeting of this pathway in combination immunotherapy.
Collapse
Affiliation(s)
- Sara K Daniel
- University of Washington, Dept. of Surgery, Seattle, WA, USA
| | - Y David Seo
- University of Washington, Dept. of Surgery, Seattle, WA, USA
| | | |
Collapse
|
199
|
Yu L, Pham Q, Yu LL, Wang TTY. Modulation of CXC-motif chemokine receptor 7, but not 4, expression is related to migration of the human prostate cancer cell LNCaP: regulation by androgen and inflammatory stimuli. Inflamm Res 2019; 69:167-178. [PMID: 31865399 DOI: 10.1007/s00011-019-01305-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 11/08/2019] [Accepted: 12/05/2019] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE To elucidate the regulation, function of the chemokine CXC-motif ligand 12 (CXCL12) and its receptors (CXCR) 4 and 7 in prostate cancer tumor microenvironment. MATERIAL In-silico-analysis of expression in prostate cancer tissues. In-vitro comparison, testing of regulation in human prostate cancer cells LNCaP, DU145, and PC3. TREATMENT Dihydrotestosterone (DHT) treatments (0-10 nM) were for 0-48 h. The inflammatory agent Flagellin treatment (20 ng/ml) was for 2 h. Migration assays were performed for 24 h using 10 ng/ml CXCL12. METHODS Real-time PCR, western analysis, and migration assays were used to determine mRNA, protein, and functional changes, respectively. RESULTS Malignant prostate cancer tissues exhibit higher CXCR4/7 mRNA ratio, and higher CXCR7 mRNA levels were detected in the androgen-responsive LNCaP cells. Putative androgen-responsive elements were identified in CXCR4, 7 gene, and exposure to DHT, flagellin increased CXCR4 mRNA but decreased CXCR7 mRNA levels in LNCaP cells. Androgen receptor siRNA significantly attenuated the effects of DHT on CXCR4, 7 mRNA in LNCaP cells. However, DHT and flagellin only decrease CXCR7 protein and additively increased migration of LNCaP cells towards CXCL12. CONCLUSIONS Down regulation of CXCR7 protein by DHT and flagellin increased migration, supporting CXCR7 as decoy receptor counteracting CXCL12/CXCR4-mediated migration in prostate cancer cells.
Collapse
Affiliation(s)
- Lu Yu
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA
| | - Quynhchi Pham
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, ARS, USDA, 10300 Baltimore Ave., Bldg. 307C, Rm 132, Beltsville, MD, 20705, USA
| | - Liangli Lucy Yu
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA
| | - Thomas T Y Wang
- Diet, Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, ARS, USDA, 10300 Baltimore Ave., Bldg. 307C, Rm 132, Beltsville, MD, 20705, USA.
| |
Collapse
|
200
|
Effect of CXCL12 and Its Receptors on Unpredictable Renal Cell Carcinoma. Clin Genitourin Cancer 2019; 18:e337-e342. [PMID: 31882334 DOI: 10.1016/j.clgc.2019.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 01/10/2023]
Abstract
Chemokines are chemotactic cytokines that participate in numerous cell functions during hematopoiesis, morphogenesis, inflammation, neovascularization, and autoimmune diseases and cancer. They achieve their functions on binding to their G protein-coupled receptors. CXCL12, or stromal cell-derived factor-1, is a homeostatic chemokine secreted by fibroblasts, macrophages, and endothelial cells. It binds to CXC receptor 4 (CXCR4), also known as fusin (CD184), and alternate CXC receptor 7 (CXCR7), also known as atypical chemokine receptor 3. The CXCL12/CXCR4 axis participates in homing of hematopoietic stem cells and the development and production of B and T lymphocytes, plasmacytoid dendritic cells, and natural killer cells. It has been examined in > 20 different malignancies. CXCL12 plays an important role in tumor metastasis because it mediates the migration of tumor cells through the endothelial vessel wall and extracellular matrix. Its expression has been highest in common metastatic sites such as the brain, bone marrow, lymph nodes, and liver. CXCR4 is expressed by tumor cells in prostate, breast, lung, and other malignancies. Numerous studies have shown its correlation with a poor prognosis, recurrence-free survival, and poor overall survival. The present review has addressed the structure and function of CXCL12 and its receptors and the effect CXCL12/CXCR4 axis has on the pathogenesis and clinical development of renal cell carcinoma, one of the most aggressive cancers in urology, with limited therapeutic options.
Collapse
|