1
|
Yang L, Fang A, Zhou S, Liu H. -RAMP3 promotes hepatocellular carcinoma tumor cell-mediated CCL2 degradation by supporting membrane distribution of ACKR2. Int Immunopharmacol 2024; 143:113419. [PMID: 39437486 DOI: 10.1016/j.intimp.2024.113419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/12/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
This study aimed to explore the potential bind of Receptor Activity-Modifying Protein 3 (RAMP3) with atypical chemokine receptor 2 (ACKR2), and their cooperative regulation on the degradation of the immunosuppressive chemokine CCL2 in the tumor microenvironment of HCC. Bioinformatic analysis was conducted using available bulk-tissue RNA-seq, single-cell RNA-seq, and protein-protein interaction datasets. Human HCC cell line Huh7 and HepG2 and mouse HCC cell line Hepa1-6 were utilized for experiments. Results showed that RAMP3 binds with ACKR2 in HCC tumor cells and promotes the membrane distribution of ACKR2 through RAB4-positive vesicles. RAMP3 promotes CCL2 scavenging through ACKR2 in HCC cells. Mouse RAMP3 inhibited the proliferation of mouse liver cancer cell line (Hepa1-6)-derived syngeneic tumors through ACKR2, reduced the intratumoral concentration of CCL2 in the tumor, and inhibited the phosphorylation of Signal Transducer and Activator of Transcription 3 (STAT3) and protein kinase B (AKT). In addition, mouse RAMP3 inhibited CD11b+/Gr-1 + myeloid cell infiltration and neovascularization in the tumors through ACKR2. In TCGA-LIHC, RAMP3low/ACKR2low group had the worst progression-free interval (PFI), while the RAMP3high/ACKR2high group had the best overall survival (OS). In summary, restoring RAMP3 expression in HCC cells may generate synergistic support for the anticancer effect of ACKR2.
Collapse
Affiliation(s)
- Lan Yang
- Department of Oncology Centre, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Aiping Fang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, 610072 Chengdu, China
| | - Shijie Zhou
- Jinruijie Biotechnology Center, Chengdu 610041, China.
| | - Hao Liu
- Department of Oncology Centre, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
2
|
Sachdev KR, Lynch KJ, Barreto GE. Exploration of novel ligands to target C-C Motif Chemokine Receptor 2 (CCR2) as a promising pharmacological treatment against traumatic brain injury. Biomed Pharmacother 2022; 151:113155. [PMID: 35598371 DOI: 10.1016/j.biopha.2022.113155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/13/2022] [Accepted: 05/15/2022] [Indexed: 11/02/2022] Open
Abstract
It is widely reported that the overexpression of the C-C Motif Chemokine Receptor 2 (CCR2) has negative implications in neuroinflammatory diseases such as traumatic brain injury (TBI), although promising drugs to tackle this have been less forthcoming. As of 2016, only 2 drugs specifically targeting this receptor have made their way to market, with unsuccessful outcome unfortunately, suggesting that the search for more specific and precise ligands is utterly necessary. In this paper we hypothesized that by targeting Glu291, Met295, Trp98, Leu45 and Val189 amino acids, essential in the binding of CCR2 with C-C Motif Chemokine Ligand 2 (CCL2), the endogenous substrate, mitigates its activity in TBI. We used a pharmacophore model to screen for suitable ligands that may bind to CCR2, which returned 871 ligands. Docking and molecular dynamics results uncovered that two ligands (A102) and (A435) contained several of those important residues and showed a stability and compactness when in complex with CCR2, with these results confirmed by MMGBSA calculations with A102 recording a better interaction compared to A435. Finally, a PPI network was built to explore downstream signaling being regulated by both ligands in TBI, showing amyloid precursor protein (APP) as a key target and neuroactive-ligand receptor interaction (1.80E-27) the top functional annotated category. In conclusion, for the first time we report novel ligands A102 and A435 targeting CCR2 as a potential new pharmacological approach to target inflammation post-TBI.
Collapse
Affiliation(s)
- Kilian R Sachdev
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Kevin J Lynch
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| |
Collapse
|
3
|
Yan B, Guo Y, Gui Y, Jiang ZS, Zheng XL. Multifunctional RNase MCPIP1 and its Role in Cardiovascular Diseases. Curr Med Chem 2021; 28:3385-3405. [PMID: 33191882 DOI: 10.2174/0929867327999201113100918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 09/20/2020] [Accepted: 10/09/2020] [Indexed: 11/22/2022]
Abstract
Monocyte chemoattractant protein-1 induced protein 1 (MCPIP1), one of the MCPIP family members, is characterized by the presence of both C-x8-C-x5-C-x3-H (CCCH)- type zinc finger and PilT-N-terminal domains. As a potent regulator of innate immunity, MCPIP1 exerts anti-inflammatory effects through its ribonuclease (RNase) and deubiquitinating enzyme activities to degrade cytokine mRNAs and inhibit nuclear factor- kappa B (NF-κB), respectively. MCPIP1 is expressed not only in immune cells but also in many other cell types, including cardiomyocytes, vascular endothelial cells (ECs) and smooth muscle cells (SMCs). Increasing evidence indicates that MCPIP1 plays a role in the regulation of cardiac functions and is involved in the processes of vascular diseases, such as ischemia-reperfusion (I/R) and atherosclerosis. To better understand the emerging roles of MCPIP1 in the cardiovascular system, we reviewed the current literature with respect to MCPIP1 functions and discussed its association with the pathogenesis of cardiovascular diseases and the implication as a therapeutic target.
Collapse
Affiliation(s)
- Binjie Yan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, China
| | - Yanan Guo
- Departments of Biochemistry & Molecular Biology and Physiology & Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, The University of Calgary, 3330 Hospital Drive N.W., Calgary, ABT2N 4N1, Canada
| | - Yu Gui
- Departments of Biochemistry & Molecular Biology and Physiology & Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, The University of Calgary, 3330 Hospital Drive N.W., Calgary, ABT2N 4N1, Canada
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, China
| | - Xi-Long Zheng
- Departments of Biochemistry & Molecular Biology and Physiology & Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, The University of Calgary, 3330 Hospital Drive N.W., Calgary, ABT2N 4N1, Canada
| |
Collapse
|
4
|
K 2P2.1 (TREK-1) potassium channel activation protects against hyperoxia-induced lung injury. Sci Rep 2020; 10:22011. [PMID: 33319831 PMCID: PMC7738539 DOI: 10.1038/s41598-020-78886-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022] Open
Abstract
No targeted therapies exist to counteract Hyperoxia (HO)-induced Acute Lung Injury (HALI). We previously found that HO downregulates alveolar K2P2.1 (TREK-1) K+ channels, which results in worsening lung injury. This decrease in TREK-1 levels leaves a subset of channels amendable to pharmacological intervention. Therefore, we hypothesized that TREK-1 activation protects against HALI. We treated HO-exposed mice and primary alveolar epithelial cells (AECs) with the novel TREK-1 activators ML335 and BL1249, and quantified physiological, histological, and biochemical lung injury markers. We determined the effects of these drugs on epithelial TREK-1 currents, plasma membrane potential (Em), and intracellular Ca2+ (iCa) concentrations using fluorometric assays, and blocked voltage-gated Ca2+ channels (CaV) as a downstream mechanism of cytokine secretion. Once-daily, intra-tracheal injections of HO-exposed mice with ML335 or BL1249 improved lung compliance, histological lung injury scores, broncho-alveolar lavage protein levels and cell counts, and IL-6 and IP-10 concentrations. TREK-1 activation also decreased IL-6, IP-10, and CCL-2 secretion from primary AECs. Mechanistically, ML335 and BL1249 induced TREK-1 currents in AECs, counteracted HO-induced cell depolarization, and lowered iCa2+ concentrations. In addition, CCL-2 secretion was decreased after L-type CaV inhibition. Therefore, Em stabilization with TREK-1 activators may represent a novel approach to counteract HALI.
Collapse
|
5
|
Arimont M, Sun SL, Leurs R, Smit M, de Esch IJP, de Graaf C. Structural Analysis of Chemokine Receptor-Ligand Interactions. J Med Chem 2017; 60:4735-4779. [PMID: 28165741 PMCID: PMC5483895 DOI: 10.1021/acs.jmedchem.6b01309] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
This
review focuses on the construction and application of structural chemokine
receptor models for the elucidation of molecular determinants of chemokine
receptor modulation and the structure-based discovery and design of
chemokine receptor ligands. A comparative analysis of ligand binding
pockets in chemokine receptors is presented, including a detailed
description of the CXCR4, CCR2, CCR5, CCR9, and US28 X-ray structures,
and their implication for modeling molecular interactions of chemokine
receptors with small-molecule ligands, peptide ligands, and large
antibodies and chemokines. These studies demonstrate how the integration
of new structural information on chemokine receptors with extensive
structure–activity relationship and site-directed mutagenesis
data facilitates the prediction of the structure of chemokine receptor–ligand
complexes that have not been crystallized. Finally, a review of structure-based
ligand discovery and design studies based on chemokine receptor crystal
structures and homology models illustrates the possibilities and challenges
to find novel ligands for chemokine receptors.
Collapse
Affiliation(s)
- Marta Arimont
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Shan-Liang Sun
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Rob Leurs
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Martine Smit
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Chris de Graaf
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
6
|
Matsushita S, Mochizuki S, Sakurai K, Kawano T. Prevention of copper-induced cell death by GC-rich DNA oligomers in murine macrophage-like RAW264.7 cells. Commun Integr Biol 2016; 8:e1017173. [PMID: 27066170 PMCID: PMC4802767 DOI: 10.1080/19420889.2015.1017173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/13/2015] [Indexed: 01/01/2023] Open
Abstract
Impact of redox active transition metals on activation of cell death signaling in plant cells have been documented to date. We have recently reported that GC-rich DNA oligomers with high affinity for binding of copper and catalytic activity for removal of ROS as novel plant cell-protecting agents. Here, we show that similar DNA oligomers protect the mouse macrophage-like RAW264.7 cells from copper-induced cell death, suggesting that the phenomenon firstly observed in plant model can be expanded to a wider range of cells and/or organisms including mammalian cells.
Collapse
Affiliation(s)
- Sakiko Matsushita
- Faculty and Graduate School of Environmental Engineering; The University of Kitakyushu ; Kitakyushu, Japan
| | - Shinichi Mochizuki
- Faculty and Graduate School of Environmental Engineering; The University of Kitakyushu ; Kitakyushu, Japan
| | - Kazuo Sakurai
- Faculty and Graduate School of Environmental Engineering; The University of Kitakyushu ; Kitakyushu, Japan
| | - Tomonori Kawano
- Faculty and Graduate School of Environmental Engineering; The University of Kitakyushu ; Kitakyushu, Japan
| |
Collapse
|
7
|
Auvynet C, Baudesson de Chanville C, Hermand P, Dorgham K, Piesse C, Pouchy C, Carlier L, Poupel L, Barthélémy S, Felouzis V, Lacombe C, Sagan S, Chemtob S, Quiniou C, Salomon B, Deterre P, Sennlaub F, Combadière C. ECL1i, d(LGTFLKC), a novel, small peptide that specifically inhibits CCL2-dependent migration. FASEB J 2016; 30:2370-81. [PMID: 26979087 DOI: 10.1096/fj.201500116] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/25/2016] [Indexed: 11/11/2022]
Abstract
CC chemokine receptor type 2 (CCR2) is a key molecule in inflammatory diseases and is an obvious drug target for the treatment of inflammation. A number of nonpeptidic, competitive CCR2 antagonists have been developed, but none has yet been approved for clinical use. Our aim was to identify a short peptide that showed allosteric antagonism against human and mouse CCR2. On the basis of sequence analysis and 3-dimensional modeling, we identified an original 7-d-amino acid peptidic CCR2 inhibitor that we have called extracellular loop 1 inverso (ECL1i), d(LGTFLKC). In vitro, ECL1i selectively and potently inhibits CC chemokine ligand type 2 (CCL2)-triggered chemotaxis (IC50, 2 µM) but no other conventional CCL2-associated events. We used the classic competitive CCR2 antagonist, BMS22 {2-[(isopropylaminocarbonyl)amino]-N-[2-[[cis-2-[[4-(methylthio)benzoyl]amino]cyclohexyl]amino]-2-oxoethyl]-5-(trifluoromethyl)benzamide}, as positive control and inhibited CCL2-dependent chemotaxis with an IC50 of 18 nM. As negative control, we used a peptide with the same composition as ECL1i, but in a different sequence, d(FKLTLCG). In vivo, ECL1i (4 mg/kg) interfered with CCR2-positive cell recruitment and attenuated disease progression in experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis. This study establishes ECL1i as the first allosteric inhibitor of CCR2 with functional selectivity. ECL1i is a promising new agent in therapeutic development, and it may, by its selective effect, increase our understanding of CCR2 signaling pathways and functions.-Auvynet, C., Baudesson de Chanville, C., Hermand, P., Dorgham, K., Piesse, C., Pouchy, C., Carlier, L., Poupel, L., Barthélémy, S., Felouzis, V., Lacombe, C., Sagan, S., Salomon, B., Deterre, P., Sennlaub, F., Combadière, C. ECL1i, d(LGTFLKC), a novel, small peptide that specifically inhibits CCL2-dependent migration.
Collapse
Affiliation(s)
- Constance Auvynet
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Camille Baudesson de Chanville
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Patricia Hermand
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Karim Dorgham
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Christophe Piesse
- Sorbonne Universités, UPMC/Univ Paris 06, Institut de Biologie Paris-Seine (IBPS) 3631, CNRS, Service de Synthése Peptidique, Paris, France
| | - Charlotte Pouchy
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Ludovic Carlier
- Sorbonne Universités, UPMC/Univ Paris 06, CNRS, UMR 7203, Laboratoire des Biomolécules, Paris, France
| | - Lucie Poupel
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Sandrine Barthélémy
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Virginie Felouzis
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Claire Lacombe
- Sorbonne Universités, UPMC/Univ Paris 06, CNRS, UMR 7203, Laboratoire des Biomolécules, Paris, France; Ecole Normale Supérieure-Université de Recherche Paris Sciences et Lettres, Département de Chimie, Paris, France; Faculté des Sciences et Technologie, Université Paris Est Créteil-Val de Marne, Créteil, France
| | - Sandrine Sagan
- Sorbonne Universités, UPMC/Univ Paris 06, CNRS, UMR 7203, Laboratoire des Biomolécules, Paris, France
| | | | | | - Benoit Salomon
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Philippe Deterre
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Florian Sennlaub
- Sorbonne Universités, UPMC/ Univ Paris 06, UMRS 968, INSERM, U968, Institut de la Vision, Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-Direction des Hôpitaux et de l'Offre de Soins (DHOS), Centre d'Investigation Clinique 503, Paris, France
| | - Christophe Combadière
- *Sorbonne Universités, Université Pierre et Marie Curie (UPMC)/Univ Paris 06, Unité Mixte de Recherche Scientifique (UMRS) 1135, INSERM Unité 1135, Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée (ERL) 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France;
| |
Collapse
|
8
|
Zhang RL, Zhang JP, Wang QQ. Recombinant Treponema pallidum protein Tp0965 activates endothelial cells and increases the permeability of endothelial cell monolayer. PLoS One 2014; 9:e115134. [PMID: 25514584 PMCID: PMC4267829 DOI: 10.1371/journal.pone.0115134] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 11/19/2014] [Indexed: 02/07/2023] Open
Abstract
The recombinant Treponema pallidum protein Tp0965 (rTp0965), one of the many proteins derived from the genome of T. pallidum subsp. pallidum, shows strong immunogenicity and immunoreactivity. In this study, we investigated the effects of rTp0965 on the endothelial barrier. Treatment of human umbilical vein endothelial cells (HUVECs) with rTp0965 resulted in increased levels of ICAM-1, E-selectin, and MCP-1 mRNA and protein expression. These increases contributed to the adhesion and chemataxis of monocytes (THP-1 cells) to HUVECs preincubated with rTp0965. In addition, rTp0965 induced reorganization of F-actin and decreased expression of claudin-1 in HUVECs. Interestingly, inhibition of the RhoA/ROCK signal pathway protected against rTp0965-induced higher endothelial permeability as well as transendothelial migration of monocytes. These data indicate that Tp0965 protein may play an important role in the immunopathogenesis of syphilis.
Collapse
Affiliation(s)
- Rui-Li Zhang
- Department of Dermatology, Wuxi Second Affiliated Hospital of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Jing-Ping Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, & National Center for STD Control, China Centers for Diseases Control and Prevention, Nanjing, Jiangsu Province, China
| | - Qian-Qiu Wang
- Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, & National Center for STD Control, China Centers for Diseases Control and Prevention, Nanjing, Jiangsu Province, China
- * E-mail:
| |
Collapse
|
9
|
Kothandan G, Gadhe CG, Cho SJ. Investigation of the Binding Site of CCR2 using 4-Azetidinyl-1-aryl-cyclohexane Derivatives: A Membrane Modeling and Molecular Dynamics Study. B KOREAN CHEM SOC 2013. [DOI: 10.5012/bkcs.2013.34.11.3429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
10
|
Binding site characterization of G protein-coupled receptor by alanine-scanning mutagenesis using molecular dynamics and binding free energy approach: application to C-C chemokine receptor-2 (CCR2). Mol Divers 2012; 16:401-13. [PMID: 22528270 DOI: 10.1007/s11030-012-9368-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 03/16/2012] [Indexed: 10/28/2022]
Abstract
The C-C chemokine receptor 2 (CCR2) was proved as a multidrug target in many diseases like diabetes, inflammation and AIDS, but rational drug design on this target is still lagging behind as the information on the exact binding site and the crystal structure is not yet available. Therefore, for a successful structure-based drug design, an accurate receptor model in ligand-bound state is necessary. In this study, binding-site residues of CCR2 was determined using in silico alanine scanning mutagenesis and the interactions between TAK-779 and the developed homology model of CCR2. Molecular dynamic simulation and Molecular Mechanics-Generalized Born Solvent Area method was applied to calculate binding free energy difference between the template and mutated protein. Upon mutating 29 amino acids of template protein and comparison of binding free energy with wild type, six residues were identified as putative hot spots of CCR2.
Collapse
|
11
|
Kothandan G, Gadhe CG, Cho SJ. Structural insights from binding poses of CCR2 and CCR5 with clinically important antagonists: a combined in silico study. PLoS One 2012; 7:e32864. [PMID: 22479344 PMCID: PMC3314010 DOI: 10.1371/journal.pone.0032864] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 01/31/2012] [Indexed: 11/19/2022] Open
Abstract
Chemokine receptors are G protein-coupled receptors that contain seven transmembrane domains. In particular, CCR2 and CCR5 and their ligands have been implicated in the pathophysiology of a number of diseases, including rheumatoid arthritis and multiple sclerosis. Based on their roles in disease, they have been attractive targets for the pharmaceutical industry, and furthermore, targeting both CCR2 and CCR5 can be a useful strategy. Owing to the importance of these receptors, information regarding the binding site is of prime importance. Structural studies have been hampered due to the lack of X-ray crystal structures, and templates with close homologs for comparative modeling. Most of the previous models were based on the bovine rhodopsin and β2-adrenergic receptor. In this study, based on a closer homolog with higher resolution (CXCR4, PDB code: 3ODU 2.5 Å), we constructed three-dimensional models. The main aim of this study was to provide relevant information on binding sites of these receptors. Molecular dynamics simulation was done to refine the homology models and PROCHECK results indicated that the models were reasonable. Here, binding poses were checked with some established inhibitors of high pharmaceutical importance against the modeled receptors. Analysis of interaction modes gave an integrated interpretation with detailed structural information. The binding poses confirmed that the acidic residues Glu291 (CCR2) and Glu283 (CCR5) are important, and we also found some additional residues. Comparisons of binding sites of CCR2/CCR5 were done sequentially and also by docking a potent dual antagonist. Our results can be a starting point for further structure-based drug design.
Collapse
Affiliation(s)
- Gugan Kothandan
- Department of Bio-New Drug Development, College of Medicine, Chosun University, Gwangju, Korea
| | - Changdev G. Gadhe
- Department of Bio-New Drug Development, College of Medicine, Chosun University, Gwangju, Korea
| | - Seung Joo Cho
- Department of Bio-New Drug Development, College of Medicine, Chosun University, Gwangju, Korea
- Department of Cellular Molecular Medicine, Research Center for Resistant Cells, College of Medicine, Chosun University, Gwangju, Korea
- * E-mail:
| |
Collapse
|
12
|
Kothandan G, Gadhe CG, Madhavan T, Cho SJ. Binding Site Analysis of CCR2 Through In Silico Methodologies: Docking, CoMFA, and CoMSIA. Chem Biol Drug Des 2011; 78:161-74. [DOI: 10.1111/j.1747-0285.2011.01095.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
13
|
Laborde E, Macsata RW, Meng F, Peterson BT, Robinson L, Schow SR, Simon RJ, Xu H, Baba K, Inagaki H, Ishiwata Y, Jomori T, Matsumoto Y, Miyachi A, Nakamura T, Okamoto M, Handel TM, Bernard CCA. Discovery, optimization, and pharmacological characterization of novel heteroaroylphenylureas antagonists of C-C chemokine ligand 2 function. J Med Chem 2011; 54:1667-81. [PMID: 21341682 PMCID: PMC3229226 DOI: 10.1021/jm1012903] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Through the application of TRAP (target-related affinity profiling), we identified a novel class of heteroaroylphenylureas that inhibit human CCL2-induced chemotaxis of monocytes/macrophages both in vitro and in vivo. This inhibition was concentration-dependent and selective with regard to other chemokines. The compounds, however, did not antagonize the binding of (125)I-labeled CCL2 to the CCR2 receptor nor did they block CCR2-mediated signal transduction responses such as calcium mobilization. Optimization of early leads for potency and pharmacokinetic parameters resulted in the identification of 17, a potent inhibitor of chemotaxis (IC(50) = 80 nM) with excellent oral bioavailability in rats (F = 60%). Compound 17 reduced swelling and joint destruction in two rat models of rheumatoid arthritis and delayed disease onset and produced near complete resolution of symptoms in a mouse model of multiple sclerosis.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis
- Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/pathology
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/pathology
- Biological Availability
- Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis
- Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- CHO Cells
- Cell Line, Tumor
- Chemokine CCL2/antagonists & inhibitors
- Chemotaxis/drug effects
- Cricetinae
- Cricetulus
- Humans
- Joints/drug effects
- Joints/pathology
- Macrophages/drug effects
- Macrophages/physiology
- Mice
- Mice, Inbred ICR
- Monocytes/drug effects
- Monocytes/physiology
- Multiple Sclerosis/drug therapy
- Phenylurea Compounds/chemical synthesis
- Phenylurea Compounds/pharmacokinetics
- Phenylurea Compounds/pharmacology
- Radioligand Assay
- Rats
- Receptors, CCR2/metabolism
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Edgardo Laborde
- Telik, Inc., 700 Hansen Way, Palo Alto, California 94304, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Beyond rhodopsin: G protein-coupled receptor structure and modeling incorporating the beta2-adrenergic and adenosine A(2A) crystal structures. Methods Mol Biol 2010; 672:359-86. [PMID: 20838977 DOI: 10.1007/978-1-60761-839-3_15] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
For quite some time, the majority of GPCR models have been based on a single template structure: dark-adapted bovine rhodopsin. The recent solution of β2AR, β1AR and adenosine A(2A) receptor crystal structures has dramatically expanded the GPCR structural landscape and provided many new insights into receptor conformation and ligand binding. They will serve as templates for the next generation of GPCR models, but also allow direct validation of previous models and computational techniques. This review summarizes key findings from the new structures, comparison of existing models to these structures and highlights new models constructed from these templates.
Collapse
|
15
|
Zhang J, Patel L, Pienta KJ. Targeting chemokine (C-C motif) ligand 2 (CCL2) as an example of translation of cancer molecular biology to the clinic. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 95:31-53. [PMID: 21075328 DOI: 10.1016/b978-0-12-385071-3.00003-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chemokines are a family of small and secreted proteins that play pleiotropic roles in inflammation-related pathological diseases, including cancer. Among the identified 50 human chemokines, chemokine (C-C motif) ligand 2 (CCL2) is of particular importance in cancer development since it serves as one of the key mediators of interactions between tumor and host cells. CCL2 is produced by cancer cells and multiple different host cells within the tumor microenvironment. CCL2 mediates tumorigenesis in many different cancer types. For example, CCL2 has been reported to promote prostate cancer cell proliferation, migration, invasion, and survival, via binding to its functional receptor CCR2. Furthermore, CCL2 induces the recruitment of macrophages and induces angiogenesis and matrix remodeling. Targeting CCL2 has been demonstrated as an effective therapeutic approach in preclinical prostate cancer models, and currently, neutralizing monoclonal antibody against CCL2 has entered into clinical trials in prostate cancer. In this chapter, targeting CCL2 in prostate cancer will be used as an example to show translation of laboratory findings from cancer molecular biology to the clinic.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Medicine, Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
16
|
Chapter 12 The Use of Receptor Homology Modeling to Facilitate the Design of Selective Chemokine Receptor Antagonists. Methods Enzymol 2009; 461:249-79. [DOI: 10.1016/s0076-6879(09)05412-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
17
|
Loberg RD, Day LL, Harwood J, Ying C, St John LN, Giles R, Neeley CK, Pienta KJ. CCL2 is a potent regulator of prostate cancer cell migration and proliferation. Neoplasia 2006; 8:578-86. [PMID: 16867220 PMCID: PMC1601934 DOI: 10.1593/neo.06280] [Citation(s) in RCA: 200] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tumor cells in the bone interact with the microenvironment to promote tumor cell survival and proliferation, resulting in a lethal phenotype for patients with advanced prostate cancer. Monocyte chemoattractant protein 1 (CCL2) is a member of the CC chemokine family and is known to promote monocyte chemotaxis to sites of inflammation. Here we have shown that human bone marrow endothelial (HBME) cells secrete significantly higher levels of CCL2 compared to human aortic endothelial cells and human dermal microvascular endothelial cells. Furthermore, we demonstrate that CCL2 is a potent chemoattractant of prostate cancer epithelial cells, and that stimulation of PC-3 and VCaP cells resulted in a dose-dependent activation of PI3 kinase/Akt signaling pathway. Activation of the PI3 kinase/Akt pathway was found to be vital to the proliferative effects of CCL2 stimulation of both PC-3 and VCaP cells. Additionally, CCL2 stimulated the phosphorylation of p70-S6 kinase (a downstream target of Akt) and induced actin rearrangement, resulting in a dynamic morphologic change indicative of microspike formation. These data suggest that bone marrow endothelial cells are a major source of CCL2, and that an elevated secretion of CCL2 recruits prostate cancer epithelial cells to the bone microenvironment and regulates their proliferation rate.
Collapse
Affiliation(s)
- Robert D Loberg
- Department of Urology, University of Michigan Urology Center, Ann Arbor, MI 48109-0946, USA.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Zhou L, Azfer A, Niu J, Graham S, Choudhury M, Adamski FM, Younce C, Binkley PF, Kolattukudy PE. Monocyte chemoattractant protein-1 induces a novel transcription factor that causes cardiac myocyte apoptosis and ventricular dysfunction. Circ Res 2006; 98:1177-85. [PMID: 16574901 PMCID: PMC1523425 DOI: 10.1161/01.res.0000220106.64661.71] [Citation(s) in RCA: 213] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monocyte chemoattractant protein-1 (MCP-1; CCL2)-mediated inflammation plays a critical role in the development of ischemic heart disease (IHD). However, the gene expression changes caused by signal transduction, triggered by MCP-1 binding to its receptor CCR2, and their possible role in the development of IHD are not understood. We present evidence that MCP-1 binding to CCR2 induces a novel transcription factor (MCP-induced protein [MCPIP]) that causes cell death. Gene microarray analysis showed that when expressed in hiuman embryonic kidney 293 cells, MCPIP induced apoptotic gene families before causing cell death. Mutagenesis studies showed that the structural features required for transcription factor-like activity were also required for causing cell death. Activation of caspase-3 was detected after MCPIP transfection and Z-VAD-fmk partially inhibited cell death. Cardiomyocyte-targeted expression of MCP-1 in mice caused death by heart failure at 6 months of age. MCPIP expression increased in parallel with the development of ventricular dysfunction. In situ hybridization showed the presence of MCPIP transcripts in the cardiomyocytes and immunohistochemistry showed that MCPIP was associated with the cardiomyocyte nuclei of apoptotic cardiomyocytes. CCR2 expression in cardiomyocytes increased with the development of IHD. MCPIP production induced by MCP-1 binding to CCR2 in the cardiomyocytes is probably involved in the development of IHD in this murine model. MCPIP transcript levels were much higher in the explanted human hearts with IHD than with nonischemic heart disease. These results provide a molecular insight into how chronic inflammation and exposure to MCP-1 contributes to heart failure and suggest that MCPIP could be a potential target for therapeutic intervention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Pappachan E. Kolattukudy
- Correspondence to Dr P.E. Kolattukudy, Burnett College of Biomedical Sciences, University of Central Florida, Building 20, Room 136, Orlando, FL 32816-2364. E-mail
| |
Collapse
|
19
|
Zhang Y, DeVries ME, Skolnick J. Structure modeling of all identified G protein-coupled receptors in the human genome. PLoS Comput Biol 2006; 2:e13. [PMID: 16485037 PMCID: PMC1364505 DOI: 10.1371/journal.pcbi.0020013] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Accepted: 01/11/2005] [Indexed: 12/22/2022] Open
Abstract
G protein–coupled receptors (GPCRs), encoded by about 5% of human genes, comprise the largest family of integral membrane proteins and act as cell surface receptors responsible for the transduction of endogenous signal into a cellular response. Although tertiary structural information is crucial for function annotation and drug design, there are few experimentally determined GPCR structures. To address this issue, we employ the recently developed threading assembly refinement (TASSER) method to generate structure predictions for all 907 putative GPCRs in the human genome. Unlike traditional homology modeling approaches, TASSER modeling does not require solved homologous template structures; moreover, it often refines the structures closer to native. These features are essential for the comprehensive modeling of all human GPCRs when close homologous templates are absent. Based on a benchmarked confidence score, approximately 820 predicted models should have the correct folds. The majority of GPCR models share the characteristic seven-transmembrane helix topology, but 45 ORFs are predicted to have different structures. This is due to GPCR fragments that are predominantly from extracellular or intracellular domains as well as database annotation errors. Our preliminary validation includes the automated modeling of bovine rhodopsin, the only solved GPCR in the Protein Data Bank. With homologous templates excluded, the final model built by TASSER has a global Cα root-mean-squared deviation from native of 4.6 Å, with a root-mean-squared deviation in the transmembrane helix region of 2.1 Å. Models of several representative GPCRs are compared with mutagenesis and affinity labeling data, and consistent agreement is demonstrated. Structure clustering of the predicted models shows that GPCRs with similar structures tend to belong to a similar functional class even when their sequences are diverse. These results demonstrate the usefulness and robustness of the in silico models for GPCR functional analysis. All predicted GPCR models are freely available for noncommercial users on our Web site (http://www.bioinformatics.buffalo.edu/GPCR). G protein–coupled receptors (GPCRs) are a large superfamily of integral membrane proteins that transduce signals across the cell membrane. Because of the breadth and importance of the physiological roles undertaken by the GPCR family, many of its members are important pharmacological targets. Although the knowledge of a protein's native structure can provide important insight into understanding its function and for the design of new drugs, the experimental determination of the three-dimensional structure of GPCR membrane proteins has proved to be very difficult. This is demonstrated by the fact that there is only one solved GPCR structure (from bovine rhodopsin) deposited in the Protein Data Bank library. In contrast, there are no human GPCR structures in the Protein Data Bank. To address the need for the tertiary structures of human GPCRs, using just sequence information, the authors use a newly developed threading-assembly-refinement method to generate models for all 907 registered GPCRs in the human genome. About 820 GPCRs are anticipated to have correct topology and transmembrane helix arrangement. A subset of the resulting models is validated by comparison with mutagenesis experimental data, and consistent agreement is demonstrated.
Collapse
Affiliation(s)
- Yang Zhang
- Center of Excellence in Bioinformatics, University at Buffalo, Buffalo, New York, United States of America
| | - Mark E DeVries
- Center of Excellence in Bioinformatics, University at Buffalo, Buffalo, New York, United States of America
| | - Jeffrey Skolnick
- Center of Excellence in Bioinformatics, University at Buffalo, Buffalo, New York, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|