1
|
Russo RC, Ryffel B. The Chemokine System as a Key Regulator of Pulmonary Fibrosis: Converging Pathways in Human Idiopathic Pulmonary Fibrosis (IPF) and the Bleomycin-Induced Lung Fibrosis Model in Mice. Cells 2024; 13:2058. [PMID: 39768150 PMCID: PMC11674266 DOI: 10.3390/cells13242058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and lethal interstitial lung disease (ILD) of unknown origin, characterized by limited treatment efficacy and a fibroproliferative nature. It is marked by excessive extracellular matrix deposition in the pulmonary parenchyma, leading to progressive lung volume decline and impaired gas exchange. The chemokine system, a network of proteins involved in cellular communication with diverse biological functions, plays a crucial role in various respiratory diseases. Chemokine receptors trigger the activation, proliferation, and migration of lung-resident cells, including pneumocytes, endothelial cells, alveolar macrophages, and fibroblasts. Around 50 chemokines can potentially interact with 20 receptors, expressed by both leukocytes and non-leukocytes such as tissue parenchyma cells, contributing to processes such as leukocyte mobilization from the bone marrow, recirculation through lymphoid organs, and tissue influx during inflammation or immune response. This narrative review explores the complexity of the chemokine system in the context of IPF and the bleomycin-induced lung fibrosis mouse model. The goal is to identify specific chemokines and receptors as potential therapeutic targets. Recent progress in understanding the role of the chemokine system during IPF, using experimental models and molecular diagnosis, underscores the complex nature of this system in the context of the disease. Despite advances in experimental models and molecular diagnostics, discovering an effective therapy for IPF remains a significant challenge in both medicine and pharmacology. This work delves into microarray results from lung samples of IPF patients and murine samples at different stages of bleomycin-induced pulmonary fibrosis. By discussing common pathways identified in both IPF and the experimental model, we aim to shed light on potential targets for therapeutic intervention. Dysregulation caused by abnormal chemokine levels observed in IPF lungs may activate multiple targets, suggesting that chemokine signaling plays a central role in maintaining or perpetuating lung fibrogenesis. The highlighted chemokine axes (CCL8-CCR2, CCL19/CCL21-CCR7, CXCL9-CXCR3, CCL3/CCL4/CCL5-CCR5, and CCL20-CCR6) present promising opportunities for advancing IPF treatment research and uncovering new pharmacological targets within the chemokine system.
Collapse
Affiliation(s)
- Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte 31270-901, MG, Brazil
| | - Bernhard Ryffel
- Laboratory of Immuno-Neuro Modulation (INEM), UMR7355 Centre National de la Recherche Scientifique (CNRS), University of Orleans, 45071 Orleans, France
| |
Collapse
|
2
|
Lee J, Goeckel ME, Levitas A, Colijn S, Shin J, Hindes A, Mun G, Burton Z, Chintalapati B, Yin Y, Abello J, Solnica-Krezel L, Stratman AN. CXCR3-CXCL11 Signaling Restricts Angiogenesis and Promotes Pericyte Recruitment. Arterioscler Thromb Vasc Biol 2024; 44:2577-2595. [PMID: 39360413 PMCID: PMC11594002 DOI: 10.1161/atvbaha.124.321434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Endothelial cell (EC)-pericyte interactions are known to remodel in response to hemodynamic forces; yet there is a lack of mechanistic understanding of the signaling pathways that underlie these events. Here, we have identified a novel signaling network regulated by blood flow in ECs-the chemokine receptor CXCR3 (CXC motif chemokine receptor 3) and one of its ligands, CXCL11 (CXC motif chemokine ligand 11)-that delimits EC angiogenic potential and promotes pericyte recruitment to ECs during development. METHODS We investigated the role of CXCR3 on vascular development using both 2- and 3-dimensional in vitro assays, to study EC-pericyte interactions and EC behavioral responses to blood flow. Additionally, genetic mutants and pharmacological modulators were used in zebrafish in vivo to study the impacts of CXCR3 loss and gain of function on vascular development. RESULTS In vitro modeling of EC-pericyte interactions demonstrates that suppression of EC-specific CXCR3 signaling leads to loss of pericyte association with EC tubes. In vivo, phenotypic defects are particularly noted in the cranial vasculature, where we see a loss of pericyte association with ECs and expansion of the vasculature in zebrafish treated with the Cxcr3 inhibitor AMG487 or in homozygous cxcr3.1/3.2/3.3 triple mutants. We also demonstrate that CXCR3-deficient ECs are more elongated, move more slowly, and have impaired EC-EC junctions compared with their control counterparts. CONCLUSIONS Our results suggest that CXCR3 signaling in ECs helps promote vascular stabilization events during development by preventing EC overgrowth and promoting pericyte recruitment.
Collapse
Affiliation(s)
- Jihui Lee
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Megan E. Goeckel
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Allison Levitas
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Sarah Colijn
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Jimann Shin
- Department of Developmental Biology (J.S., A.H., L.S.-K.), Washington University School of Medicine, St. Louis, MO
| | - Anna Hindes
- Department of Developmental Biology (J.S., A.H., L.S.-K.), Washington University School of Medicine, St. Louis, MO
| | - Geonyoung Mun
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Zarek Burton
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Bharadwaj Chintalapati
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Ying Yin
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Javier Abello
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology (J.S., A.H., L.S.-K.), Washington University School of Medicine, St. Louis, MO
- Center of Regenerative Medicine (L.S.-K.), Washington University School of Medicine, St. Louis, MO
| | - Amber N. Stratman
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
3
|
Mendes N, Zanesco A, Aguiar C, Rodrigues-Luiz GF, Silva D, Campos J, Camara NOS, Moraes-Vieira P, Araujo E, Velloso LA. CXCR3-expressing myeloid cells recruited to the hypothalamus protect against diet-induced body mass gain and metabolic dysfunction. eLife 2024; 13:RP95044. [PMID: 39535032 PMCID: PMC11560133 DOI: 10.7554/elife.95044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Microgliosis plays a critical role in diet-induced hypothalamic inflammation. A few hours after a high-fat diet (HFD), hypothalamic microglia shift to an inflammatory phenotype, and prolonged fat consumption leads to the recruitment of bone marrow-derived cells to the hypothalamus. However, the transcriptional signatures and functions of these cells remain unclear. Using dual-reporter mice, this study reveals that CX3CR1-positive microglia exhibit minimal changes in response to a HFD, while significant transcriptional differences emerge between microglia and CCR2-positive recruited myeloid cells, particularly affecting chemotaxis. These recruited cells also show sex-specific transcriptional differences impacting neurodegeneration and thermogenesis. The chemokine receptor CXCR3 is emphasized for its role in chemotaxis, displaying notable differences between recruited cells and resident microglia, requiring further investigation. Central immunoneutralization of CXCL10, a ligand for CXCR3, resulted in increased body mass and decreased energy expenditure, especially in females. Systemic chemical inhibition of CXCR3 led to significant metabolic changes, including increased body mass, reduced energy expenditure, elevated blood leptin, glucose intolerance, and decreased insulin levels. This study elucidates the transcriptional differences between hypothalamic microglia and CCR2-positive recruited myeloid cells in diet-induced inflammation and identifies CXCR3-expressing recruited immune cells as protective in metabolic outcomes linked to HFD consumption, establishing a new concept in obesity-related hypothalamic inflammation.
Collapse
Affiliation(s)
- Natalia Mendes
- School of Medical Sciences, Department of Translational Medicine (Section of Pharmacology), University of CampinasCampinasBrazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Ariane Zanesco
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Cristhiane Aguiar
- Laboratory of Immunometabolism, Institute of Biology - University of Campinas, BrazilCampinasBrazil
| | - Gabriela F Rodrigues-Luiz
- Department of Microbiology, Immunology and Parasitology, Federal University of Santa CatarinaFlorianópolisBrazil
| | - Dayana Silva
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Jonathan Campos
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
| | - Niels Olsen Saraiva Camara
- Laboratory for Transplantation Immunobiology, Institute of Biomedical Sciences, University of Sao PauloSao PauloBrazil
| | - Pedro Moraes-Vieira
- Laboratory of Immunometabolism, Institute of Biology - University of Campinas, BrazilCampinasBrazil
| | - Eliana Araujo
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
- Faculty of Nursing, University of CampinasCampinasBrazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of CampinasCampinasBrazil
- National Institute of Science and Technology on NeuroimmunomodulationRio de JaneiroBrazil
| |
Collapse
|
4
|
Xing X, Zhao C, Cai S, Wang J, Zhang J, Sun F, Huang M, Zhang L. Deciphering the mediating role of CXCL10 in hypothyroidism-induced idiopathic pulmonary fibrosis in European ancestry: a Mendelian randomization study. Front Immunol 2024; 15:1379480. [PMID: 39185418 PMCID: PMC11341476 DOI: 10.3389/fimmu.2024.1379480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease characterized by progressive fibrosis, leading to impaired gas exchange and high mortality. The etiology of IPF is complex, with potential links to autoimmune disorders such as hypothyroidism. This study explores the relationship between hypothyroidism and IPF, focusing on the mediating role of plasma proteins. Methods A two-sample Mendelian randomization (MR) approach was employed to determine the impact of hypothyroidism on IPF and the mediating role of 4,907 plasma proteins, all in individuals of European ancestry. Sensitivity analyses, external validation, and reverse causality tests were conducted to ensure the robustness of the findings. Additionally, the function of causal SNPs was evaluated through gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Conclusion The findings suggest that hypothyroidism, through altered plasma protein expression, particularly CXCL10, may contribute to the pathogenesis of IPF. This novel insight highlights the potential of CXCL10 as a therapeutic target in IPF, especially in patients with hypothyroidism. The study emphasizes the need for further research into the complex interplay between autoimmune disorders and IPF, with a view towards developing targeted interventions for IPF management.
Collapse
Affiliation(s)
- Xiaoming Xing
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Cong Zhao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Song Cai
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fang Sun
- Department of respiration, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mao Huang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lishan Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
5
|
Xu Y, Cohen E, Johnson CN, Parent CA, Coulombe PA. Repeated stress to the skin amplifies neutrophil infiltration in a keratin 17- and PKCα-dependent manner. PLoS Biol 2024; 22:e3002779. [PMID: 39159283 PMCID: PMC11361748 DOI: 10.1371/journal.pbio.3002779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 08/29/2024] [Accepted: 07/31/2024] [Indexed: 08/21/2024] Open
Abstract
Neutrophils are the first immune cells to reach inflamed sites and contribute to the pathogenesis of chronic inflammatory skin diseases. Yet, little is known about the pattern of neutrophil infiltration in inflamed skin in vivo and the mechanisms mediating their recruitment. Here, we provide insight into the dynamics of neutrophil infiltration in skin in response to acute or repeated inflammatory stress, highlighting a novel keratinocyte- and keratin 17 (K17)-dependent mechanism that regulates neutrophil recruitment to inflamed skin. We used the phorbol ester TPA and UVB, alone or in combination, to induce sterile inflammation in mouse skin. A single TPA treatment results in a neutrophil influx in the dermis that peaks at 12 h and resolves within 24 h. A subsequent TPA treatment or a UVB challenge, when applied 24 h but not 48 h later, accelerates, amplifies, and prolongs neutrophil infiltration. This transient amplification response (TAR) is mediated by local signals in inflamed skin, can be recapitulated in ex vivo culture, and involves the K17-dependent sustainment of protein kinase Cα (PKCα) activity and release of chemoattractants by stressed keratinocytes. K17 binds RACK1, a scaffold protein essential for PKCα activity. The N-terminal head domain of K17 is crucial for its association with RACK1 and regulation of PKCα activity. Analysis of RNAseq data reveals a signature consistent with TAR and PKCα activation in inflammatory skin diseases. These findings uncover a novel, keratin-dependent mechanism that amplifies neutrophil recruitment in skin under stress, with direct implications for inflammatory skin disorders.
Collapse
Affiliation(s)
- Yang Xu
- Graduate Program in Pharmacology and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Erez Cohen
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Craig N. Johnson
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Carole A. Parent
- Graduate Program in Pharmacology and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Life Science Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Pierre A. Coulombe
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
6
|
Jiao H, Pang B, Liu A, Chen Q, Pan Q, Wang X, Xu Y, Chiang YC, Ren R, Hu H. Structural insights into the activation and inhibition of CXC chemokine receptor 3. Nat Struct Mol Biol 2024; 31:610-620. [PMID: 38177682 PMCID: PMC11026165 DOI: 10.1038/s41594-023-01175-5] [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: 01/30/2023] [Accepted: 11/03/2023] [Indexed: 01/06/2024]
Abstract
The chemotaxis of CD4+ type 1 helper cells and CD8+ cytotoxic lymphocytes, guided by interferon-inducible CXC chemokine 9-11 (CXCL9-11) and CXC chemokine receptor 3 (CXCR3), plays a critical role in type 1 immunity. Here we determined the structures of human CXCR3-DNGi complexes activated by chemokine CXCL11, peptidomimetic agonist PS372424 and biaryl-type agonist VUF11222, and the structure of inactive CXCR3 bound to noncompetitive antagonist SCH546738. Structural analysis revealed that PS372424 shares a similar orthosteric binding pocket to the N terminus of CXCL11, while VUF11222 buries deeper and activates the receptor in a distinct manner. We showed an allosteric binding site between TM5 and TM6, accommodating SCH546738 in the inactive CXCR3. SCH546738 may restrain the receptor at an inactive state by preventing the repacking of TM5 and TM6. By revealing the binding patterns and the pharmacological properties of the four modulators, we present the activation mechanisms of CXCR3 and provide insights for future drug development.
Collapse
Affiliation(s)
- Haizhan Jiao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China
| | - Bin Pang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Aijun Liu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China
| | - Qiang Chen
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China
| | - Qi Pan
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China
| | - Xiankun Wang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China
| | - Yunong Xu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China
| | - Ying-Chih Chiang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China.
| | - Ruobing Ren
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, China.
| |
Collapse
|
7
|
Drews SJ. Commentary on "Inhibition of cellular activation induced by platelet factor 4 via the CXCR3 pathway ameliorates Japanese encephalitis and dengue viral infections". J Thromb Haemost 2024; 22:609-612. [PMID: 38417982 DOI: 10.1016/j.jtha.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 03/01/2024]
Affiliation(s)
- Steven J Drews
- Canadian Blood Services, Microbiology Department, Donor Policy and Studies, Edmonton, Alberta, Canada; Division of Diagnostic and Applied Microbiology, Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
8
|
Livne-Bar I, Maurya S, Gronert K, Sivak JM. Lipoxins A 4 and B 4 inhibit glial cell activation via CXCR3 signaling in acute retinal neuroinflammation. J Neuroinflammation 2024; 21:18. [PMID: 38212822 PMCID: PMC10782675 DOI: 10.1186/s12974-024-03010-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/01/2024] [Indexed: 01/13/2024] Open
Abstract
Lipoxins are small lipids that are potent endogenous mediators of systemic inflammation resolution in a variety of diseases. We previously reported that Lipoxins A4 and B4 (LXA4 and LXB4) have protective activities against neurodegenerative injury. Yet, lipoxin activities and downstream signaling in neuroinflammatory processes are not well understood. Here, we utilized a model of posterior uveitis induced by lipopolysaccharide endotoxin (LPS), which results in rapid retinal neuroinflammation primarily characterized by activation of resident macroglia (astrocytes and Müller glia), and microglia. Using this model, we observed that each lipoxin reduces acute inner retinal inflammation by affecting endogenous glial responses in a cascading sequence beginning with astrocytes and then microglia, depending on the timing of exposure; prophylactic or therapeutic. Subsequent analyses of retinal cytokines and chemokines revealed inhibition of both CXCL9 (MIG) and CXCL10 (IP10) by each lipoxin, compared to controls, following LPS injection. CXCL9 and CXCL10 are common ligands for the CXCR3 chemokine receptor, which is prominently expressed in inner retinal astrocytes and ganglion cells. We found that CXCR3 inhibition reduces LPS-induced neuroinflammation, while CXCR3 agonism alone induces astrocyte reactivity. Together, these data uncover a novel lipoxin-CXCR3 pathway to promote distinct anti-inflammatory and proresolution cascades in endogenous retinal glia.
Collapse
Affiliation(s)
- Izhar Livne-Bar
- Department of Vision Sciences, Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada
- Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Canada
| | - Shubham Maurya
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | - Karsten Gronert
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
- Vision Science Program, University of California Berkeley, Berkeley, CA, USA
- Infectious Disease and Immunity Program, University of California Berkeley, Berkeley, CA, USA
| | - Jeremy M Sivak
- Department of Vision Sciences, Donald K Johnson Eye Institute, Krembil Research Institute, University Health Network, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada.
- Department of Ophthalmology and Vision Science, University of Toronto School of Medicine, Toronto, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto School of Medicine, Toronto, Canada.
| |
Collapse
|
9
|
Yuan Z. Research progress of CXCR3 inhibitors. Anticancer Drugs 2024; 35:36-45. [PMID: 37694856 DOI: 10.1097/cad.0000000000001543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The human CXCR3 receptor was initially identified and cloned in the mid-1990s. In the process of understanding CXCR3, it gradually found that it plays an important role in the process of a variety of diseases, including inflammation, immune diseases, cancer, cardiovascular diseases, central nervous system diseases, etc., which attracted the attention of many researchers. Subsequently, some small molecule inhibitors targeting CXCR3 receptors were also developed. Unfortunately, no CXCR3 inhibitors have been approved for marketing by FDA. Up to now, only one CXCR3 small molecule inhibitor has entered the clinical trial stage, but it has not achieved ideal results in the end. Therefore, there is still much to think about and explore for the development of CXCR3 inhibitors. This article reviews the important role of CXCR3 in various physiological and pathological processes and some small molecule inhibitors of CXCR3.
Collapse
Affiliation(s)
- Zhuo Yuan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Wang S, Ballard TE, Christopher LJ, Foti RS, Gu C, Khojasteh SC, Liu J, Ma S, Ma B, Obach RS, Schadt S, Zhang Z, Zhang D. The Importance of Tracking "Missing" Metabolites: How and Why? J Med Chem 2023; 66:15586-15612. [PMID: 37769129 DOI: 10.1021/acs.jmedchem.3c01293] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Technologies currently employed to find and identify drug metabolites in complex biological matrices generally yield results that offer a comprehensive picture of the drug metabolite profile. However, drug metabolites can be missed or are captured only late in the drug development process. This could be due to a variety of factors, such as metabolism that results in partial loss of the molecule, covalent bonding to macromolecules, the drug being metabolized in specific human tissues, or poor ionization in a mass spectrometer. These scenarios often draw a great deal of attention from chemistry, safety assessment, and pharmacology. This review will summarize scenarios of missing metabolites, why they are missing, and associated uncovering strategies from deeper investigations. Uncovering previously missed metabolites can have ramifications in drug development with toxicological and pharmacological consequences, and knowledge of these can help in the design of new drugs.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - T Eric Ballard
- Takeda Development Center Americas, Inc., 35 Landsdowne St, Cambridge, Massachusetts 02139, United States
| | - Lisa J Christopher
- Department of Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol-Myers Squibb, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Robert S Foti
- Preclinical Development, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Chungang Gu
- Drug Metabolism and Pharmacokinetics, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Joyce Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shuguang Ma
- Drug Metabolism and Pharmacokinetics, Pliant Therapeutics, 260 Littlefield Avenue, South San Francisco, California 94080, United States
| | - Bin Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - R Scott Obach
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Simone Schadt
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacher Strasse 124, 4070 Basel, Switzerland
| | - Zhoupeng Zhang
- DMPK Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Donglu Zhang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
11
|
Russo RC, Quesniaux VFJ, Ryffel B. Homeostatic chemokines as putative therapeutic targets in idiopathic pulmonary fibrosis. Trends Immunol 2023; 44:1014-1030. [PMID: 37951789 DOI: 10.1016/j.it.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal chronic interstitial lung disease (ILD) that affects lung mechanical functions and gas exchange. IPF is caused by increased fibroblast activity and collagen deposition that compromise the alveolar-capillary barrier. Identifying an effective therapy for IPF remains a clinical challenge. Chemokines are key proteins in cell communication that have functions in immunity as well as in tissue homeostasis, damage, and repair. Chemokine receptor signaling induces the activation and proliferation of lung-resident cells, including alveolar macrophages (AMs) and fibroblasts. AMs are an important source of chemokines and cytokines during IPF. We highlight the complexity of this system and, based on insights from genetic and transcriptomic studies, propose a new role for homeostatic chemokine imbalance in IPF, with implications for putative therapeutic targets.
Collapse
Affiliation(s)
- Remo C Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| | - Valerie F J Quesniaux
- Experimental and Molecular Immunology and Neurogenetics (INEM), Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7355, University of Orleans, Orleans 45071, France.
| | - Bernhard Ryffel
- Experimental and Molecular Immunology and Neurogenetics (INEM), Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7355, University of Orleans, Orleans 45071, France.
| |
Collapse
|
12
|
Jiao H, Pang B, Chiang YC, Chen Q, Pan Q, Ren R, Hu H. Structure basis for the modulation of CXC chemokine receptor 3 by antagonist AMG487. Cell Discov 2023; 9:119. [PMID: 38012179 PMCID: PMC10682378 DOI: 10.1038/s41421-023-00617-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/26/2023] [Indexed: 11/29/2023] Open
Affiliation(s)
- Haizhan Jiao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong, China
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong, China
| | - Bin Pang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Ying-Chih Chiang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong, China
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong, China
| | - Qiang Chen
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong, China
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong, China
| | - Qi Pan
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong, China
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong, China
| | - Ruobing Ren
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.
- Shanghai Qi Zhi Institute, Shanghai, China.
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong, China.
- School of Life and Health Sciences, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Shenzhen, Guangdong, China.
| |
Collapse
|
13
|
Xu Y, Cohen E, Johnson CN, Parent CA, Coulombe PA. Keratin 17- and PKCα-dependent transient amplification of neutrophil influx after repeated stress to the skin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.11.561954. [PMID: 37873256 PMCID: PMC10592713 DOI: 10.1101/2023.10.11.561954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Neutrophils contribute to the pathogenesis of chronic inflammatory skin diseases. Little is known about the source and identity of the signals mediating their recruitment in inflamed skin. We used the phorbol ester TPA and UVB, alone or in combination, to induce sterile inflammation in mouse skin and assess whether keratinocyte-derived signals impact neutrophil recruitment. A single TPA treatment results in a neutrophil influx in the dermis that peaks at 12h and resolves within 24h. A second TPA treatment or a UVB challenge, when applied at 24h but not 48h later, accelerates, amplifies, and prolongs neutrophil infiltration. This transient amplification response (TAR) is mediated by local signals in inflamed skin, can be recapitulated in ex vivo culture, and involves the K17-dependent sustainment of protein kinase Cα (PKCα) activity and release of neutrophil chemoattractants by stressed keratinocytes. We show that K17 binds RACK1, a scaffold essential for PKCα activity. Finally, analyses of RNAseq data reveal the presence of a transcriptomic signature consistent with TAR and PKCα activation in chronic inflammatory skin diseases. These findings uncover a novel, transient, and keratin-dependent mechanism that amplifies neutrophil recruitment to the skin under stress, with direct implications for inflammatory skin disorders.
Collapse
|
14
|
Diamond T, Lau M, Morrissette J, Chu N, Behrens EM. CXCL9 inhibition does not ameliorate disease in murine models of both primary and secondary hemophagocytic lymphohistiocytosis. Sci Rep 2023; 13:12298. [PMID: 37516815 PMCID: PMC10387083 DOI: 10.1038/s41598-023-39601-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/27/2023] [Indexed: 07/31/2023] Open
Abstract
Hemophagocytic Lymphohistiocytosis (HLH) is a group of disorders culminating in systemic inflammation and multi-organ failure with high incidence of hepatic dysfunction. Overproduction of IFN-γ is the main immunopathological driver in this disorder. Monokine induced by IFN-γ (CXCL9) serves as a biomarker for disease activity and response to treatment in this disorder. However, very little is understood about the actual functional role of CXCL9 in pathogenesis in HLH. In the current study, we sought to determine the role of CXCL9 in pathogenesis in murine models of both Familial HLH (prf1-/-) and Toll Like Receptor (TLR) 9 repeated stimulation induced Macrophage Activation Syndrome (MAS), a form of secondary HLH. FHL and MAS were induced in both CXCL9 genetically deficient mice (cxcl9-/-) and controls as well as using AMG487, a pharmacological antagonist of the CXCL9 receptor, CXCR3. Results showed that CXCL9 genetic deficiency did not improve disease parameters or hepatitis in both models. Consistent with genetic ablation of CXCL9, inhibition of its receptor, CXCR3, by AMG487 did not show any significant effects in the FHL model. Taken together, inhibition of CXCL9-CXCR3 interaction does not ameliorate HLH physiology in general, or hepatitis as a classical target organ of disease.
Collapse
Affiliation(s)
- Tamir Diamond
- Division of Gastroenterology, Hepatology and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| | - Michelle Lau
- Division of Gastroenterology, Hepatology and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeremy Morrissette
- Department of Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Niansheng Chu
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Edward M Behrens
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
15
|
Dillemans L, De Somer L, Neerinckx B, Proost P. A review of the pleiotropic actions of the IFN-inducible CXC chemokine receptor 3 ligands in the synovial microenvironment. Cell Mol Life Sci 2023; 80:78. [PMID: 36862204 PMCID: PMC11071919 DOI: 10.1007/s00018-023-04715-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/09/2023] [Accepted: 02/01/2023] [Indexed: 03/03/2023]
Abstract
Chemokines are pivotal players in instigation and perpetuation of synovitis through leukocytes egress from the blood circulation into the inflamed articulation. Multitudinous literature addressing the involvement of the dual-function interferon (IFN)-inducible chemokines CXCL9, CXCL10 and CXCL11 in diseases characterized by chronic inflammatory arthritis emphasizes the need for detangling their etiopathological relevance. Through interaction with their mutual receptor CXC chemokine receptor 3 (CXCR3), the chemokines CXCL9, CXCL10 and CXCL11 exert their hallmark function of coordinating directional trafficking of CD4+ TH1 cells, CD8+ T cells, NK cells and NKT cells towards inflammatory niches. Among other (patho)physiological processes including infection, cancer, and angiostasis, IFN-inducible CXCR3 ligands have been implicated in autoinflammatory and autoimmune diseases. This review presents a comprehensive overview of the abundant presence of IFN-induced CXCR3 ligands in bodily fluids of patients with inflammatory arthritis, the outcomes of their selective depletion in rodent models, and the attempts at developing candidate drugs targeting the CXCR3 chemokine system. We further propose that the involvement of the CXCR3 binding chemokines in synovitis and joint remodeling encompasses more than solely the directional ingress of CXCR3-expressing leukocytes. The pleotropic actions of the IFN-inducible CXCR3 ligands in the synovial niche reiteratively illustrate the extensive complexity of the CXCR3 chemokine network, which is based on the intercommunion of IFN-inducible CXCR3 ligands with distinct CXCR3 isoforms, enzymes, cytokines, and infiltrated and resident cells present in the inflamed joints.
Collapse
Affiliation(s)
- Luna Dillemans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Lien De Somer
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Barbara Neerinckx
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
16
|
Fitz-Henley JN, Rozema SD, Golden JE. Dihydropyrazinoquinazolinones via S N2 Sulfamidate Ring-Opening and a Sequential Quinazolinone-Amidine Rearrangement Strategy (SQuAReS). J Org Chem 2022; 87:14889-14898. [PMID: 36194836 PMCID: PMC9795801 DOI: 10.1021/acs.joc.2c01717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A synthesis of dihydropyrazino-[2,1-b]-quinazolinones is described using a 2-alkylaminoquinazolinone-mediated ring opening of a-/chiral sulfamidates, followed by a tandem quinazolinone-amidine rearrangement termed SQuAReS. This approach takes advantage of sulfamidates whose regioselective ring opening, after hydrolysis, appends an optimally distanced nucleophilic amine to a quinazolinone such that subsequent domino rearrangements are favored, integrating unique substitution patterns on a privileged core. This three-step protocol integrated five telescoped transformations and generated 20 pyrazinoquinazolinones in up to 74% yield with high enantiomeric fidelity and diastereoselectivity.
Collapse
Affiliation(s)
- Jhewelle N Fitz-Henley
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin─Madison, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Soren D Rozema
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin─Madison, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Jennifer E Golden
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin─Madison, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| |
Collapse
|
17
|
Hua X, Zhang J, Ge S, Liu H, Du H, Niu Q, Chen X, Yang C, Zhang L, Liang C. CXCR3 antagonist AMG487 ameliorates experimental autoimmune prostatitis by diminishing Th1 cell differentiation and inhibiting macrophage M1 phenotypic activation. Prostate 2022; 82:1223-1236. [PMID: 35700340 DOI: 10.1002/pros.24395] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 04/16/2022] [Accepted: 05/16/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Chronic prostatitis and chronic pelvic pain syndrome (CP/CPPS) is an inflammatory immune disease that is characterized by infiltrating inflammatory cells in the prostate and pelvic or by perineal pain. Receptor CXCR3modulates immune and inflammatory responses; however, the effects of CXCR3 antagonist AMG487 in the context of CP/CPPS are unknown. Therefore, we investigated the effect of AMG487 in experimental autoimmune prostatitis (EAP) mice and explored the potential functional mechanisms. METHODS The EAP model was induced by intradermally injecting a mixture of prostate antigens and complete Freund's adjuvant on Days 0 and 28. To evaluate the effect of AMG487 on EAP mice, treatment with AMG487 and vehicle solution was conducted for the indicated period. Then, procedures were performed, including behavioral test, to evaluate the pain response to stimulation before the mice were killed and a histological assessment to evaluate the inflammation after the mice were killed. Immunofluorescence, flow cytometry, and Western blot assay were used to analyze the functional phenotype and regulation mechanism of AMG487 on T helper type 1 (Th1) cells and macrophages. RESULTS We found high expression of CXCR3 in human benign prostate tissues with inflammation and EAP mice. The elevated CXCR3 in prostate tissues correlates with the severity of inflammation. CXCR3 antagonist AMG487 treatment ameliorated the inflammatory changes and the pelvic pain of EAP mice. AMG487 inhibits Th1 cell differentiation through the IL-12/STAT4pathway and inhibits pro-inflammatory M1 macrophages through the lipopolysaccharide/NF-κB p65signaling. AMG487 could inhibit the secretion of inflammatory mediators in EAP mice. CONCLUSION CXCR3 antagonist AMG487 could ameliorate the inflammatory changes and the pelvic pain of EAP mice by diminishing Th1 cell differentiation and inhibiting macrophage M1 phenotypic activation. Thus, the results imply that AMG487 has the potential as an effective therapeutic agent in the prevention and treatment of EAP.
Collapse
Affiliation(s)
- Xiaoliang Hua
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
- The Institute of Urology, Anhui Medical University, Hefei, China
| | - Jiong Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
- The Institute of Urology, Anhui Medical University, Hefei, China
| | - Shengdong Ge
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
- The Institute of Urology, Anhui Medical University, Hefei, China
| | - Haoran Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
- The Institute of Urology, Anhui Medical University, Hefei, China
| | - Hexi Du
- The Institute of Urology, Anhui Medical University, Hefei, China
| | - Qingsong Niu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- The Institute of Urology, Anhui Medical University, Hefei, China
| | - Xianguo Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
- The Institute of Urology, Anhui Medical University, Hefei, China
| | - Cheng Yang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
- The Institute of Urology, Anhui Medical University, Hefei, China
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
- The Institute of Urology, Anhui Medical University, Hefei, China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, China
- The Institute of Urology, Anhui Medical University, Hefei, China
- Anhui Institute of Translational Medicine, Hefei, China
| |
Collapse
|
18
|
Caroff E, Meyer EA, Äänismaa P, Froidevaux S, Keller M, Piali L. Design, Synthesis, and Pharmacological Evaluation of Benzimidazolo-thiazoles as Potent CXCR3 Antagonists with Therapeutic Potential in Autoimmune Diseases: Discovery of ACT-672125. J Med Chem 2022; 65:11533-11549. [PMID: 35969159 DOI: 10.1021/acs.jmedchem.2c00676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The chemokine receptor CXCR3 allows the selective recruitment of innate and adaptive inflammatory immune cells into inflamed tissue. CXCR3 ligands are secreted after exposure to pro-inflammatory cytokines. Upon binding to CXCR3 ligands, CXCR3 expressing T-lymphocytes migrate toward sites of inflammation and can promote tissue damage. Therefore, antagonizing this receptor may provide clinical benefits for patients suffering from autoimmune diseases characterized by high concentrations of CXCR3 ligands. Herein, we report the second part of our CXCR3 discovery program where we explored the benzimidazolo-thiazole core scaffold. The optimization of potency and the mitigation of an hERG liability are described. Further pharmacokinetic considerations led to the identification of the potent CXCR3 antagonist ACT-672125 (29). The compound showed good physicochemical properties and safety profile. In a proof-of-mechanism model of lung inflammation, ACT-672125 inhibited the recruitment of CXCR3 expressing T cells into the inflamed lung in a dose-dependent manner.
Collapse
Affiliation(s)
- Eva Caroff
- Drug Discovery Chemistry Immunology, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| | - Emmanuel A Meyer
- Drug Discovery Chemistry Immunology, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| | - Päivi Äänismaa
- DMPK, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| | | | - Marcel Keller
- Drug Discovery Biology Immunology, Idorsia Pharmaceuticals Ltd., Allschwil 4123, Switzerland
| | - Luca Piali
- Immunology, Infectious Diseases and Ophthalmology, pRED Roche, Basel 4070, Switzerland
| |
Collapse
|
19
|
Heng AHS, Han CW, Abbott C, McColl SR, Comerford I. Chemokine-Driven Migration of Pro-Inflammatory CD4 + T Cells in CNS Autoimmune Disease. Front Immunol 2022; 13:817473. [PMID: 35250997 PMCID: PMC8889115 DOI: 10.3389/fimmu.2022.817473] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/25/2022] [Indexed: 12/13/2022] Open
Abstract
Pro-inflammatory CD4+ T helper (Th) cells drive the pathogenesis of many autoimmune conditions. Recent advances have modified views of the phenotype of pro-inflammatory Th cells in autoimmunity, extending the breadth of known Th cell subsets that operate as drivers of these responses. Heterogeneity and plasticity within Th1 and Th17 cells, and the discovery of subsets of Th cells dedicated to production of other pro-inflammatory cytokines such as GM-CSF have led to these advances. Here, we review recent progress in this area and focus specifically upon evidence for chemokine receptors that drive recruitment of these various pro-inflammatory Th cell subsets to sites of autoimmune inflammation in the CNS. We discuss expression of specific chemokine receptors by subsets of pro-inflammatory Th cells and highlight which receptors may be tractable targets of therapeutic interventions to limit pathogenic Th cell recruitment in autoimmunity.
Collapse
Affiliation(s)
- Aaron H S Heng
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Caleb W Han
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Caitlin Abbott
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Shaun R McColl
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| | - Iain Comerford
- The Chemokine Biology Laboratory, Department of Molecular and Biomedical Science, School of Biological Sciences, Faculty of Science, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
20
|
Cxcl10 chemokine induces migration of ING4-deficient breast cancer cells via a novel crosstalk mechanism between the Cxcr3 and Egfr receptors. Mol Cell Biol 2021; 42:e0038221. [PMID: 34871062 DOI: 10.1128/mcb.00382-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The chemokine Cxcl10 has been associated with poor prognosis in breast cancer, but the mechanism is not well understood. Our previous study have shown that CXCL10 was repressed by the ING4 tumor suppressor, suggesting a potential inverse functional relationship. We thus investigated a role for Cxcl10 in the context of ING4 deficiencies in breast cancer. We first analyzed public gene expression datasets and found that patients with CXCL10-high/ING4-low expressing tumors had significantly reduced disease-free survival in breast cancer. In vitro, Cxcl10 induced migration of ING4-deleted breast cancer cells, but not of ING4-intact cells. Using inhibitors, we found that Cxcl10-induced migration of ING4-deleted cells required Cxcr3, Egfr, and the Gβγ subunits downstream of Cxcr3, but not Gαi. Immunofluorescent imaging showed that Cxcl10 induced early transient colocalization between Cxcr3 and Egfr in both ING4-intact and ING4-deleted cells, which recurred only in ING4-deleted cells. A peptide agent that binds to the internal juxtamembrane domain of Egfr inhibited Cxcr3/Egfr colocalization and cell migration. Taken together, these results presented a novel mechanism of Cxcl10 that elicits migration of ING4-deleted cells, in part by inducing a physical or proximal association between Cxcr3 and Egfr and signaling downstream via Gβγ. These results further indicated that ING4 plays a critical role in the regulation of Cxcl10 signaling that enables breast cancer progression.
Collapse
|
21
|
Li X, Golden JE. Construction of N-Boc-2-Alkylaminoquinazolin-4(3 H)-Ones via a Three-Component, One-Pot Protocol Mediated by Copper(II) Chloride that Spares Enantiomeric Purity. Adv Synth Catal 2021; 363:1638-1645. [PMID: 33867902 PMCID: PMC8048503 DOI: 10.1002/adsc.202001279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/27/2020] [Indexed: 12/21/2022]
Abstract
Chiral 2-alkylquinazolinones are key synthetic intermediates, but their preparation in high optical purity is challenging. Thus, a multicomponent procedure integrating anthranilic acids, N-Boc-amino acids, and amines in the presence of methanesulfonyl chloride, N-methylimidazole, and copper(II) chloride was developed to mildly afford N-Boc-2-alkylaminoquinazolin-4(3H)-ones with excellent preservation of enantiomeric purity (>99% ee). Copper(II) chloride was essential to retaining enantiopurity, and reaction component structural changes were well tolerated, resulting in an efficient, all-in-one procedure that promotes sequential coupling, lactonization, aminolysis, and cyclization in good yields. The method was applied to the rapid assembly of four key intermediates used in the synthesis of high profile quinazolinones, including several PI3K inhibitor drugs.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Pharmaceutical SciencesSchool of PharmacyUniversity of Wisconsin-MadisonMadisonWisconsin53705-2222USA
| | - Jennifer E. Golden
- Department of Pharmaceutical SciencesSchool of PharmacyUniversity of Wisconsin-MadisonMadisonWisconsin53705-2222USA
| |
Collapse
|
22
|
Cxcl9l and Cxcr3.2 regulate recruitment of osteoclast progenitors to bone matrix in a medaka osteoporosis model. Proc Natl Acad Sci U S A 2020; 117:19276-19286. [PMID: 32719141 PMCID: PMC7431079 DOI: 10.1073/pnas.2006093117] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bone remodeling requires a balanced interplay of osteoblasts and osteoclasts. While the intercellular signaling that triggers bone cell differentiation is well understood, it remains unclear how bone progenitor cells are recruited to remodeling sites. Various chemokines are upregulated under osteoporotic conditions. However, whether they are involved in progenitor recruitment or instead have inflammatory roles is unknown. Here we used a medaka fish osteoporosis model to identify the chemokine ligand Cxcl9l and receptor Cxcr3.2 as essential to control osteoclast progenitor recruitment and differentiation at bone resorption sites. Cxcr3.2 activity can be blocked by small-molecule inhibitors that protect bone from osteoporotic insult. Our study demonstrates the potential of fish for osteoporosis drug discovery and opens avenues for future osteoporosis therapy. Bone homeostasis requires continuous remodeling of bone matrix to maintain structural integrity. This involves extensive communication between bone-forming osteoblasts and bone-resorbing osteoclasts to orchestrate balanced progenitor cell recruitment and activation. Only a few mediators controlling progenitor activation are known to date and have been targeted for intervention of bone disorders such as osteoporosis. To identify druggable pathways, we generated a medaka (Oryzias latipes) osteoporosis model, where inducible expression of receptor-activator of nuclear factor kappa-Β ligand (Rankl) leads to ectopic formation of osteoclasts and excessive bone resorption, which can be assessed by live imaging. Here we show that upon Rankl induction, osteoblast progenitors up-regulate expression of the chemokine ligand Cxcl9l. Ectopic expression of Cxcl9l recruits mpeg1-positive macrophages to bone matrix and triggers their differentiation into osteoclasts. We also demonstrate that the chemokine receptor Cxcr3.2 is expressed in a distinct subset of macrophages in the aorta-gonad-mesonephros (AGM). Live imaging revealed that upon Rankl induction, Cxcr3.2-positive macrophages get activated, migrate to bone matrix, and differentiate into osteoclasts. Importantly, mutations in cxcr3.2 prevent macrophage recruitment and osteoclast differentiation. Furthermore, Cxcr3.2 inhibition by the chemical antagonists AMG487 and NBI-74330 also reduced osteoclast recruitment and protected bone integrity against osteoporotic insult. Our data identify a mechanism for progenitor recruitment to bone resorption sites and Cxcl9l and Cxcr3.2 as potential druggable regulators of bone homeostasis and osteoporosis.
Collapse
|
23
|
Bakheet SA, Alrwashied BS, Ansari MA, Nadeem A, Attia SM, Assiri MA, Alqahtani F, Ibrahim KE, Ahmad SF. CXCR3 antagonist AMG487 inhibits glucocorticoid-induced tumor necrosis factor-receptor-related protein and inflammatory mediators in CD45 expressing cells in collagen-induced arthritis mouse model. Int Immunopharmacol 2020; 84:106494. [PMID: 32304993 DOI: 10.1016/j.intimp.2020.106494] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease classified by uncontrolled joint inflammation leading to the destruction of both cartilage and joints. Despite progress made in RA treatment in the past decade, new drugs with high efficacy and fewer long-term adverse effects are still needed; thus, safe anti-inflammatory therapies for RA are urgently needed. Previous results demonstrated that the CXCR3 antagonist is an extremely attractive therapeutic target for the treatment of several autoimmune diseases, suggesting that it might have an inhibitory effect on RA. In this study, we investigated the effect of AMG487, a selective CXCR3 antagonist, on collagen-induced arthritis (CIA) in mice and evaluated its potential therapeutic mechanism.Following induction of CIA, mice were treated with AMG487 (5 mg/kg, intraperitoneally), to investigate their protective effects against CIA. CD4, CD25, CCR6, IL-9, NF-κB, IL-6, IL-17A, IL-21, STAT6 and Foxp3 expressing GITR+ and CD45+ cells were measured in the spleen using flow cytometry to assess anti-inflammatory effects of AMG487. The mRNA and protein expression of GITR, CCR6, IL-9, and IL-21 were measured using quantitative real-time PCR and western blot analysis in knee tissue. AMG487 significantly alleviated joint inflammation by decreasing GITR+CD25+, GITR+CD45+, GITR+IL-9+, GITR+NF-κB+ CD45+CD4+, CD45+CCR6+, CD45+IL-6+ cells, CD45+IL-17A+, and CD45+IL-21+, and increasing GITR+Foxp3+ and GITR+STAT6+ cells. There was a significant decrease in mRNA and protein expression of GITR, CD4, CCR6, IL-6, IL-9, and IL-21 in knee tissue of CIA mice. This study demonstrates that AMG487 has a potential therapeutic effect on RA and could explore novel anti-inflammatory therapies for its treatment.
Collapse
Affiliation(s)
- Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Bader S Alrwashied
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed A Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khalid E Ibrahim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
24
|
Bakheet SA, Ansari MA, Nadeem A, Attia SM, Alhoshani AR, Gul G, Al-Qahtani QH, Albekairi NA, Ibrahim KE, Ahmad SF. CXCR3 antagonist AMG487 suppresses rheumatoid arthritis pathogenesis and progression by shifting the Th17/Treg cell balance. Cell Signal 2019; 64:109395. [PMID: 31449849 DOI: 10.1016/j.cellsig.2019.109395] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 10/26/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that is characterized by uncontrolled joint inflammation and damage to bone and cartilage. Previous studies have shown that chemokine receptors have important roles in RA development, and that blocking these receptors effectively inhibits RA progression. Our study was undertaken to investigate the role of AMG487, a selective CXCR3 antagonist, in DBA/1J mice bearing collagen-induced arthritis (CIA). Following induction of CIA, animals were treated with 5 mg/kg AMG487 intraperitoneally every 48 h, starting from day 21 until day 41 and evaluated for clinical score, and histological hallmarks of arthritic inflammation. We further investigated the effect of AMG487 on Th1 (T-bet), Th17 (IL-17A, RORγt, STAT3), Th22 (IL-22), and T regulatory (Treg; Foxp3 and IL-10) cells in splenic CXCR3+ and CD4+ T cells using flow cytometry. We also assessed the effect of AMG487 on T-bet, RORγt, IL-17A, IL-22, Foxp3, and IL-10 at both mRNA and protein levels using RT-PCR and Western blot analyses of knee samples. The severity of clinical scores, and histological inflammatory damage decreased significantly in AMG487-treated compared with CIA control mice. Moreover, the percentage of Th1, Th17, and Th22 cells decreased significantly and that of Treg cells increased in AMG487-treated mice. We further observed that AMG487-treatment downregulated T-bet, IL-17A, RORγt, and IL-22, whereas it upregulated Foxp3 and IL-10 mRNA and protein levels. This study demonstrates the antiarthritic effects of AMG487 in CIA animal model and supports the development of CXCR3 antagonists as a novel strategy for the treatment of inflammatory and arthritic conditions.
Collapse
Affiliation(s)
- Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Ali R Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Gazala Gul
- Department of Pathology, College of Medicine, Yenepoya University, Mangaluru, Karnataka, India
| | - Q H Al-Qahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Norah A Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Khalid E Ibrahim
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
25
|
Blocking CXCR3 with AMG487 ameliorates the blood-retinal barrier disruption in diabetic mice through anti-oxidative. Life Sci 2019; 228:198-207. [PMID: 31039363 DOI: 10.1016/j.lfs.2019.04.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 12/16/2022]
Abstract
Oxidative stress and blood-retinal barrier (BRB) damage induced by hyperglycemia are the principal processes involved in the early stages of diabetic retinopathy (DR). CXC chemokine receptor 3 (CXCR3)-mediated inflammatory infiltration exists in many disease models. The main objective of the present study was to determine whether AMG487, a CXCR3 antagonist, can ameliorate BRB disruption and reactive oxygen species generation in the DR model. The retinal endothelial cell and ganglion cell ultrastructures were observed using a transmission electron microscope. The pericyte marker PDGFR-β, tight junction occludin, and leaking albumin were evaluated. The oxidative stress level, CCAAT-enhancer-binding protein homologous protein (CHOP), and p-p38 expression were also investigated in vivo and in vitro. The results indicated that AMG487 application might alleviate PDGFR-β and occludin loss, and decreased the residual content of retinal albumin in the streptozocin-induced DR mouse model via the inhibition of oxidative and endoplasmic reticulum stress, in which p38 activation was also involved. Thus, CXCR3 inhibition might be a target to prevent the early stage of DR injury.
Collapse
|
26
|
Regenass P, Abboud D, Daubeuf F, Lehalle C, Gizzi P, Riché S, Hachet-Haas M, Rohmer F, Gasparik V, Boeglin D, Haiech J, Knehans T, Rognan D, Heissler D, Marsol C, Villa P, Galzi JL, Hibert M, Frossard N, Bonnet D. Discovery of a Locally and Orally Active CXCL12 Neutraligand (LIT-927) with Anti-inflammatory Effect in a Murine Model of Allergic Airway Hypereosinophilia. J Med Chem 2018; 61:7671-7686. [PMID: 30106292 DOI: 10.1021/acs.jmedchem.8b00657] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We previously reported Chalcone-4 (1) that binds the chemokine CXCL12, not its cognate receptors CXCR4 or CXCR7, and neutralizes its biological activity. However, this neutraligand suffers from limitations such as poor chemical stability, solubility, and oral activity. Herein, we report on the discovery of pyrimidinone 57 (LIT-927), a novel neutraligand of CXCL12 which displays a higher solubility than 1 and is no longer a Michael acceptor. While both 1 and 57 reduce eosinophil recruitment in a murine model of allergic airway hypereosinophilia, 57 is the only one to display inhibitory activity following oral administration. Thereby, we here describe 57 as the first orally active CXCL12 neutraligand with anti-inflammatory properties. Combined with a high binding selectivity for CXCL12 over other chemokines, 57 represents a powerful pharmacological tool to investigate CXCL12 physiology in vivo and to explore the activity of chemokine neutralization in inflammatory and related diseases.
Collapse
Affiliation(s)
- Pierre Regenass
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Dayana Abboud
- Biotechnologie et Signalisation Cellulaire , Ecole Supérieure de Biotechnologie de Strasbourg, UMR 7242 CNRS/Université de Strasbourg , Bld Sébastien Brant , 67412 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - François Daubeuf
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Plate-forme de chimie biologique intégrative de Strasbourg , UMS 3286 CNRS/Université de Strasbourg , 67412 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Christine Lehalle
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Plate-forme de chimie biologique intégrative de Strasbourg , UMS 3286 CNRS/Université de Strasbourg , 67412 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Patrick Gizzi
- Plate-forme de chimie biologique intégrative de Strasbourg , UMS 3286 CNRS/Université de Strasbourg , 67412 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Stéphanie Riché
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Muriel Hachet-Haas
- Biotechnologie et Signalisation Cellulaire , Ecole Supérieure de Biotechnologie de Strasbourg, UMR 7242 CNRS/Université de Strasbourg , Bld Sébastien Brant , 67412 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - François Rohmer
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Vincent Gasparik
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Damien Boeglin
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Jacques Haiech
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Tim Knehans
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Didier Rognan
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Denis Heissler
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Claire Marsol
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Plate-forme de chimie biologique intégrative de Strasbourg , UMS 3286 CNRS/Université de Strasbourg , 67412 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Pascal Villa
- Plate-forme de chimie biologique intégrative de Strasbourg , UMS 3286 CNRS/Université de Strasbourg , 67412 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Jean-Luc Galzi
- Biotechnologie et Signalisation Cellulaire , Ecole Supérieure de Biotechnologie de Strasbourg, UMR 7242 CNRS/Université de Strasbourg , Bld Sébastien Brant , 67412 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Marcel Hibert
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| | - Dominique Bonnet
- Laboratoire d'Innovation Thérapeutique , Faculté de Pharmacie, UMR7200 CNRS/Université de Strasbourg , 74 route du Rhin , 67401 Illkirch , France.,Labex MEDALIS , Université de Strasbourg , 67000 Strasbourg , France
| |
Collapse
|
27
|
CXCR3 blockade combined with cyclosporine A alleviates acute graft-versus-host disease by inhibiting alloreactive donor T cell responses in a murine model. Mol Immunol 2017; 94:82-90. [PMID: 29288898 DOI: 10.1016/j.molimm.2017.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/20/2017] [Accepted: 12/12/2017] [Indexed: 01/28/2023]
Abstract
Chemotaxis of T cells to acute graft-versus-host disease (aGvHD) target tissues directed by chemokines and their receptors plays a key role in the pathogenesis of aGvHD. Blockade of lymphocyte migration by targeting chemokine receptors may be a viable strategy for the prevention and treatment of aGvHD, which is quite distinguishable from typical efforts to use immunosuppressive medications that have been associated with some side effects. CXCR3 and its ligands have been reported to be correlated with aGvHD pathogenesis. Using the small-molecule CXCR3 antagonist AMG487, we demonstrated that AMG487 combined with cyclosporine A (CsA) effectively alleviated aGvHD with a prolonged mean survival time and significantly inhibited the infiltration of inflammatory cells in aGvHD target tissues in a murine aGvHD model. In addition, AMG487 combined with CsA inhibited the activation, proliferation and differentiation of donor-derived T cells in the spleens. Further results showed that the concentrations of Th1 cells associated with pro-inflammatory cytokines such as IFN-γ and TNFα in serum were decreased. In addition, AMG487 treatment did not alter CXCR3 and CCR5 expression in donor-derived T cells but elevated the serum CXCL9 and CXCL10 levels. This novel and effective approach has the potential to develop a new clinical method to prevent and treat aGvHD.
Collapse
|
28
|
Vaseghi G, Jafari E, Hassanzadeh F, Haghjooy-Javanmard S, Dana N, Rafieian-Kopaei M. Cytotoxic Evaluation of Some Fused Pyridazino- and Pyrrolo-quinazolinones Derivatives on Melanoma and Prostate Cell Lines. Adv Biomed Res 2017; 6:76. [PMID: 28706884 PMCID: PMC5501066 DOI: 10.4103/2277-9175.209049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Quinazolinon as an important class of heterocycles is attractive in medicinal research areas due to their wide range of biological effects. Cytotoxic activities of the quinazolinone derivatives in various cell lines including: HeLa, L1210 (mouse lymphocytic leukemia) and HT29 (human colon adenocarcinoma) were reported. Materials and Methods: In this study, a number of newly made tricycles quinazolinone derivatives such as fused pyridazino-quinazolinones and fused pyrrolo-quinazolinones were evaluated on two cancerous cell lines, melanoma (B16F10) and prostate (PC3) using the 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide colorimetric assay. Results: The results of cytotoxicity evaluations indicated that almost all of the compounds at the concentrations of 10 and 100 μM showed significant differences in viability in comparison with negative control at 48 h exposure (P < 0.05). However, during 24 h exposure some of the compounds showed cytotoxicity activity. Conclusion: Results showed that both cell lines were sensitive to synthesized compounds and longer duration of exposure (48 h) had better results compared to that of 24 h screening.
Collapse
Affiliation(s)
- Golnaz Vaseghi
- Applied Physiology Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Jafari
- Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farshid Hassanzadeh
- Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shaghayegh Haghjooy-Javanmard
- Applied Physiology Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasim Dana
- Applied Physiology Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
29
|
Wei M, Zhang X, Wang X, Song Z, Ding J, Meng LH, Zhang A. SAR study of 5-alkynyl substituted quinazolin-4(3H)-ones as phosphoinositide 3-kinase delta (PI3Kδ) inhibitors. Eur J Med Chem 2017; 125:1156-1171. [DOI: 10.1016/j.ejmech.2016.11.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 11/01/2016] [Accepted: 11/06/2016] [Indexed: 01/21/2023]
|
30
|
Abstract
INTRODUCTION By virtue of its specificity for chemokines induced in Th1-associated pathologies, CXCR3 has attracted considerable attention as a target for therapeutic intervention. Several pharmacologically distinct small molecules with in vitro and in vivo potency have been described in the literature, although to date, none have shown efficacy in clinical trials. Areas covered: In this article, the author outlines the rationale for targeting CXCR3 and discusses the potential pitfalls in targeting receptors in poorly understood areas of chemokine biology. Furthermore, they cover emerging therapeutic areas outside of the 'traditional' Th1 arena in which CXCR3 antagonists may ultimately bear fruit. Finally, they discuss the design of recently discovered small molecules targeting CXCR3. Expert opinion: CXCR3 and its ligands appear to play roles in a multitude of diverse diseases in humans. In vitro studies suggest that CXCR3 is inherently 'druggable' and that potent, efficacious small molecules targeting CXCR3 antagonists will find a clinical niche. However, the well-trodden path to failure of small molecule chemokine receptor antagonists in clinical trials suggests that a cautious approach should be undertaken. Ideally, unequivocal evidence elucidating the precise role of CXCR3 should be obtained before targeting the receptor in a particular disease cohort.
Collapse
Affiliation(s)
- James E Pease
- a Inflammation, Repair & Development Section, National Heart & Lung Institute, Faculty of Medicine , Imperial College London , London , UK
| |
Collapse
|
31
|
Ha Y, Liu H, Zhu S, Yi P, Liu W, Nathanson J, Kayed R, Loucas B, Sun J, Frishman LJ, Motamedi M, Zhang W. Critical Role of the CXCL10/C-X-C Chemokine Receptor 3 Axis in Promoting Leukocyte Recruitment and Neuronal Injury during Traumatic Optic Neuropathy Induced by Optic Nerve Crush. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 187:352-365. [PMID: 27960090 DOI: 10.1016/j.ajpath.2016.10.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/14/2016] [Accepted: 10/13/2016] [Indexed: 12/11/2022]
Abstract
Traumatic optic neuropathy (TON) is an acute injury of the optic nerve secondary to trauma. Loss of retinal ganglion cells (RGCs) is a key pathological process in TON, yet mechanisms responsible for RGC death remain unclear. In a mouse model of TON, real-time noninvasive imaging revealed a dramatic increase in leukocyte rolling and adhesion in veins near the optic nerve (ON) head at 9 hours after ON injury. Although RGC dysfunction and loss were not detected at 24 hours after injury, massive leukocyte infiltration was observed in the superficial retina. These cells were identified as T cells, microglia/monocytes, and neutrophils but not B cells. CXCL10 is a chemokine that recruits leukocytes after binding to its receptor C-X-C chemokine receptor (CXCR) 3. The levels of CXCL10 and CXCR3 were markedly elevated in TON, and up-regulation of CXCL10 was mediated by STAT1/3. Deleting CXCR3 in leukocytes significantly reduced leukocyte recruitment, and prevented RGC death at 7 days after ON injury. Treatment with CXCR3 antagonist attenuated TON-induced RGC dysfunction and cell loss. In vitro co-culture of primary RGCs with leukocytes resulted in increased RGC apoptosis, which was exaggerated in the presence of CXCL10. These results indicate that leukocyte recruitment in retinal vessels near the ON head is an early event in TON and the CXCL10/CXCR3 axis has a critical role in recruiting leukocytes and inducing RGC death.
Collapse
Affiliation(s)
- Yonju Ha
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Hua Liu
- Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, Texas
| | - Shuang Zhu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Panpan Yi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Wei Liu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Jared Nathanson
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Rakez Kayed
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas
| | - Bradford Loucas
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, Texas
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | | | - Massoud Motamedi
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas; Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, Texas
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, Texas; Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas.
| |
Collapse
|
32
|
Patel L, Chandrasekhar J, Evarts J, Forseth K, Haran AC, Ip C, Kashishian A, Kim M, Koditek D, Koppenol S, Lad L, Lepist EI, McGrath ME, Perreault S, Puri KD, Villaseñor AG, Somoza JR, Steiner BH, Therrien J, Treiberg J, Phillips G. Discovery of Orally Efficacious Phosphoinositide 3-Kinase δ Inhibitors with Improved Metabolic Stability. J Med Chem 2016; 59:9228-9242. [DOI: 10.1021/acs.jmedchem.6b01169] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Leena Patel
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | | | - Jerry Evarts
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Kristen Forseth
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Aaron C. Haran
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Carmen Ip
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Adam Kashishian
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Musong Kim
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - David Koditek
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Sandy Koppenol
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Latesh Lad
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Eve-Irene Lepist
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Mary E. McGrath
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Stephane Perreault
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Kamal D. Puri
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Armando G. Villaseñor
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - John R. Somoza
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Bart H. Steiner
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Joseph Therrien
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Jennifer Treiberg
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Gary Phillips
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| |
Collapse
|
33
|
Pharmacological opportunities to control inflammatory diseases through inhibition of the leukocyte recruitment. Pharmacol Res 2016; 112:37-48. [DOI: 10.1016/j.phrs.2016.01.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/14/2016] [Accepted: 01/15/2016] [Indexed: 12/30/2022]
|
34
|
Patel L, Chandrasekhar J, Evarts J, Haran AC, Ip C, Kaplan JA, Kim M, Koditek D, Lad L, Lepist EI, McGrath ME, Novikov N, Perreault S, Puri KD, Somoza JR, Steiner BH, Stevens KL, Therrien J, Treiberg J, Villaseñor AG, Yeung A, Phillips G. 2,4,6-Triaminopyrimidine as a Novel Hinge Binder in a Series of PI3Kδ Selective Inhibitors. J Med Chem 2016; 59:3532-48. [DOI: 10.1021/acs.jmedchem.6b00213] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Leena Patel
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Jayaraman Chandrasekhar
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Jerry Evarts
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Aaron C. Haran
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Carmen Ip
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Joshua A. Kaplan
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Musong Kim
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - David Koditek
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Latesh Lad
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Eve-Irene Lepist
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Mary E. McGrath
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Nikolai Novikov
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Stephane Perreault
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Kamal D. Puri
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - John R. Somoza
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Bart H. Steiner
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Kirk L. Stevens
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Joseph Therrien
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Jennifer Treiberg
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Armando G. Villaseñor
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Arthur Yeung
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| | - Gary Phillips
- Gilead Sciences, Inc., 199 E
Blaine Street, Seattle, Washington 98102, United States
- Gilead Sciences, Inc., 333 Lakeside Drive, Foster City, California 94404, United States
| |
Collapse
|
35
|
Admas TH, Bernat V, Heinrich MR, Tschammer N. Development of Photoactivatable Allosteric Modulators for the Chemokine Receptor CXCR3. ChemMedChem 2016; 11:575-84. [PMID: 26880380 DOI: 10.1002/cmdc.201500573] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Indexed: 11/05/2022]
Abstract
The CXCR3 receptor, a class A G protein-coupled receptor (GPCR), is involved in the regulation and trafficking of various immune cells. CXCR3 antagonists have been proposed to be beneficial for the treatment of a wide range of disorders including but not limited to inflammatory and autoimmune diseases. The structure-based design of CXCR3 ligands remains, however, hampered by a lack of structural information describing in detail the interactions between an allosteric ligand and the receptor. We designed and synthesized photoactivatable probes for the structural and functional characterization, using photoaffinity labeling followed by mass spectrometry, of the CXCR3 allosteric binding pocket of AMG 487 and RAMX3, two potent and selective CXCR3 negative allosteric modulators. Photoaffinity labeling is a common approach to elucidate binding modes of small-molecule ligands of GPCRs through the aid of photoactivatable probes that convert to extremely reactive intermediates upon photolysis. The photolabile probe N-[({1-[3-(4-ethoxyphenyl)-4-oxo-3,4-dihydropyrido[2,3-d]pyrimidin-2-yl]ethyl}-2-[4-fluoro-3-(trifluoromethyl)phenyl]-N-{1-[4-(3-(trifluoromethyl)-3H-diazirin-3-yl]benzyl}piperidin-4-yl)methyl]acetamide (10) showed significant labeling of the CXCR3 receptor (80%) in a [(3) H]RAMX3 radioligand displacement assay. Compound 10 will serve as an important tool compound for the detailed investigation of the binding pocket of CXCR3 by mass spectrometry.
Collapse
Affiliation(s)
- Tizita Haimanot Admas
- Department of Chemistry & Pharmacy, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg, Schuhstr. 19, 91052, Erlangen, Germany
| | - Viachaslau Bernat
- Department of Chemistry & Pharmacy, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg, Schuhstr. 19, 91052, Erlangen, Germany.,Department of Chemistry, The Scripps Research Institute, 130 Scripps Way, 3A1, 33458, Jupiter, FL, USA
| | - Markus R Heinrich
- Department of Chemistry & Pharmacy, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg, Schuhstr. 19, 91052, Erlangen, Germany
| | - Nuska Tschammer
- Department of Chemistry & Pharmacy, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg, Schuhstr. 19, 91052, Erlangen, Germany. .,NanoTemper Technologies GmbH, Flößergasse 4, 81369, München, Germany.
| |
Collapse
|
36
|
Abstract
Chemokines and their receptors are known to play important roles in disease. More than 40 chemokine ligands and 20 chemokine receptors have been identified, but, to date, only two small molecule chemokine receptor antagonists have been approved by the FDA. The chemokine receptor CXCR3 was identified in 1996, and nearly 20 years later, new areas of CXCR3 disease biology continue to emerge. Several classes of small molecule CXCR3 antagonists have been developed, and two have shown efficacy in preclinical models of inflammatory disease. However, only one CXCR3 antagonist has been evaluated in clinical trials, and there remain many opportunities to further investigate known classes of CXCR3 antagonists and to identify new chemotypes. This Perspective reviews the known CXCR3 antagonists and considers future opportunities for the development of small molecules for clinical evaluation.
Collapse
Affiliation(s)
- Stephen P Andrews
- Heptares Therapeutics , BioPark, Broadwater Road, Welwyn Garden City, AL7 3AX, United Kingdom
| | - Rhona J Cox
- Respiratory, Inflammation & Autoimmunity iMed, AstraZeneca, Respiratory, Inflammation & Autoimmunity IMED , Pepparedsleden, 431 83 Mölndal, Sweden
| |
Collapse
|
37
|
Mladic M, Scholten DJ, Wijtmans M, Falck D, Leurs R, Niessen WMA, Smit MJ, Kool J. Metabolic profiling of ligands for the chemokine receptor CXCR3 by liquid chromatography-mass spectrometry coupled to bioaffinity assessment. Anal Bioanal Chem 2015; 407:7067-81. [PMID: 26164305 PMCID: PMC4551560 DOI: 10.1007/s00216-015-8867-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/16/2015] [Accepted: 06/18/2015] [Indexed: 01/08/2023]
Abstract
Chemokine receptors belong to the class of G protein-coupled receptors and are important in the host defense against infections and inflammation. However, aberrant chemokine signaling is linked to different disorders such as cancer, central nervous system and immune disorders, and viral infections [Scholten DJ et al. (2012) Br J Pharmacol 165(6):1617–1643]. Modulating the chemokine receptor function provides new ways of targeting specific diseases. Therefore, discovery and development of drugs targeting chemokine receptors have received considerable attention from the pharmaceutical industry in the past decade. Along with that, the determination of bioactivities of individual metabolites derived from lead compounds towards chemokine receptors is crucial for drug selectivity, pharmacodynamics, and potential toxicity issues. Therefore, advanced analytical methodologies are in high demand. This study is aimed at the optimization of a new analytical method for metabolic profiling with parallel bioaffinity assessment of CXCR3 ligands of the azaquinazolinone and piperazinyl-piperidine class and their metabolites. The method is based on mass spectrometric (MS) identification after liquid chromatographic (LC) separation of metabolic mixtures. The bioaffinity assessment is performed “at-line” via high-resolution nanofractionation onto 96-well plates allowing direct integration of radioligand binding assays. This new method enables identification of metabolites from lead compounds with associated estimation of their individual bioaffinity. Moreover, the identification of the metabolite structures via accurate mass measurements and MS2 allows the identification of liable metabolic “hotspots” for further lead optimization. The efficient combination of chemokine receptor ligand binding assays with analytical techniques, involving nanofractionation as linking technology, allows implementation of comprehensive metabolic profiling in an early phase of the drug discovery process.
Collapse
Affiliation(s)
- Marija Mladic
- />Division of BioAnalytical Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands
- />Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands
| | - Danny J. Scholten
- />Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands
| | - Maikel Wijtmans
- />Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands
| | - David Falck
- />Division of BioAnalytical Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands
| | - Rob Leurs
- />Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands
| | - Wilfried M. A. Niessen
- />Division of BioAnalytical Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands
- />hyphen MassSpec, de Wetstraat 8, 2332 XT Leiden, The Netherlands
| | - Martine J. Smit
- />Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands
| | - Jeroen Kool
- />Division of BioAnalytical Chemistry, Amsterdam Institute for Molecules Medicines and Systems, VU University Amsterdam, De Boelelaan 1083, 1081HV Amsterdam, The Netherlands
| |
Collapse
|
38
|
Scholten DJ, Wijtmans M, van Senten JR, Custers H, Stunnenberg A, de Esch IJP, Smit MJ, Leurs R. Pharmacological characterization of [3H]VUF11211, a novel radiolabeled small-molecule inverse agonist for the chemokine receptor CXCR3. Mol Pharmacol 2015; 87:639-48. [PMID: 25576486 DOI: 10.1124/mol.114.095265] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Chemokine receptor CXCR3 has attracted much attention, as it is thought to be associated with a wide range of immune-related diseases. As such, several small molecules with different chemical structures targeting CXCR3 have been discovered. Despite limited clinical success so far, these compounds serve as interesting tools for investigating receptor activation and antagonism. Accumulating evidence suggests that many of these compounds are allosteric modulators for CXCR3. One feature of allosteric ligands is that the magnitude of the mediated allosteric effect is dependent on the orthosteric probe that is used. Consequently, there is a risk for incorrect assessment of affinity for allosteric modulators with orthosteric radioligands, which has so far been the most applied approach for chemokine receptors. Therefore, we aimed to use a small-molecule allosteric ligand from the piperazinyl-piperidine class, also known as VUF11211 [(S)-5-chloro-6-(4-(1-(4-chlorobenzyl)piperidin-4-yl)-3-ethylpiperazin-1-yl)-N-ethylnicotinamide]. VUF11211 acts as an inverse agonist at a constitutively active mutant of CXCR3. Radiolabeling of VUF11211 gave [(3)H]VUF11211, which in radioligand binding studies shows high affinity for CXCR3 (Kd = 0.65 nM) and reasonably fast association (kon= 0.03 minute(-1)nM(-1)) and dissociation kinetics (koff = 0.02 minute(-1)). The application of the [(3)H]VUF11211 to assess CXCR3 pharmacology was validated with diverse classes of CXCR3 compounds, including both antagonists and agonists, as well as VUF11211 analogs. Interestingly, VUF11211 seems to bind to a different population of CXCR3 conformations compared with the CXCR3 agonists CXC chemokine ligand 11 (CXCL11), VUF11418 [1-((1R,5S)-6,6-dimethylbicyclo[3.1.1]hept-2-en-2-yl)-N-((2'-iodobiphenyl-4-yl)methyl)-N,N-dimethylmethanaminium Iodide], and VUF10661 [N-(6-amino-1-(2,2-diphenylethylamino)-1-oxohexan-2-yl)-2-(4-oxo-4-phenylbutanoyl)-1,2,3,4-tetrahydroisoquinoline-3-carboxamide]. These findings, taken together, indicate that this allosteric inverse agonist radioligand for CXCR3 may facilitate the discovery, characterization, and optimization of allosteric modulators for the chemokine receptor CXCR3.
Collapse
Affiliation(s)
- Danny J Scholten
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, The Netherlands
| | - Maikel Wijtmans
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, The Netherlands
| | - Jeffrey R van Senten
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, The Netherlands
| | - Hans Custers
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, The Netherlands
| | - Ailas Stunnenberg
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, The Netherlands
| | - Iwan J P de Esch
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, The Netherlands
| | - Martine J Smit
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, The Netherlands
| | - Rob Leurs
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
39
|
CXCR3 signaling in BRAFWT melanoma increases IL-8 expression and tumorigenicity. PLoS One 2015; 10:e0121140. [PMID: 25798946 PMCID: PMC4370421 DOI: 10.1371/journal.pone.0121140] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 02/12/2015] [Indexed: 01/02/2023] Open
Abstract
Patients with early stage, radial growth phase (RGP) melanoma have a 97% survival rate; however, when the melanoma progresses to the invasive vertical growth phase (VGP), survival rates decrease to 15%. The targets of many clinical trials are the known genetic and molecular mechanisms involved in melanoma progression, with the most common oncogenic mutation being the BRAFV600E. However, less than half of melanomas harbor this mutation, and consequently, do not respond to the current BRAF targeted treatments. It is therefore critical to elucidate alternative mechanisms regulating melanoma progression. Increased expression of the chemokine receptor, CXCR3, on melanoma cells is correlated with increased metastasis and poor patient outcomes, suggesting a role for CXCR3 in the RGP to VGP transition. We found that endogenous CXCR3 can be induced in two RGP cell lines, BOWES (BRAFWT) and WM35 (BRAFV600E), with in vitro environmental stress and nutrient deprivation. Signaling via induced endogenous CXCR3 is linked with IL-8 expression in BOWES cells. Ectopic overexpression of CXCR3 in BOWES cells leads to increased ligand-mediated phERK, cellular migration, and IL-8 expression in vitro, and to increased tumorigenesis and lymph node metastasis in vivo. Our results demonstrate that, in BRAFWT melanomas, CXCR3 signaling mediates significant increases in IL-8 expression, suggesting that CXCR3 expression and signaling may represent a transformative event that drives the progression of BRAFWT melanomas. Implications: Expression of CXCR3 on BRAFWT melanoma cells may be a mediator of melanoma progression.
Collapse
|
40
|
Nedjai B, Viney JM, Li H, Hull C, Anderson CA, Horie T, Horuk R, Vaidehi N, Pease JE. CXCR3 antagonist VUF10085 binds to an intrahelical site distinct from that of the broad spectrum antagonist TAK-779. Br J Pharmacol 2015; 172:1822-33. [PMID: 25425280 PMCID: PMC4376459 DOI: 10.1111/bph.13027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 11/16/2014] [Accepted: 11/20/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE The chemokine receptor CXCR3 is implicated in a variety of clinically important diseases, notably rheumatoid arthritis and atherosclerosis. Consequently, antagonists of CXCR3 are of therapeutic interest. In this study, we set out to characterize binding sites of the specific low MW CXCR3 antagonist VUF10085 and the broad spectrum antagonist TAK-779 which blocks CXCR3 along with CCR2 and CCR5. EXPERIMENTAL APPROACH Molecular modelling of CXCR3, followed by virtual ligand docking, highlighted several CXCR3 residues likely to contact either antagonist, notably a conserved aspartate in helix 2 (Asp-112(2:63) ), which was postulated to interact with the quaternary nitrogen of TAK-779. Validation of modelling was carried out by site-directed mutagenesis of CXCR3, followed by assays of cell surface expression, ligand binding and receptor activation. KEY RESULTS Mutation of Asn-132(3.33) , Phe-207 and Tyr-271(6.51) within CXCR3 severely impaired both ligand binding and chemotactic responses, suggesting that these residues are critical for maintenance of a functional CXCR3 conformation. Contrary to our hypothesis, mutation of Asp-112(2:63) had no observable effects on TAK-779 activity, but clearly decreased the antagonist potency of VUF 10085. Likewise, mutations of Phe-131(3.32) , Ile-279(6.59) and Tyr-308(7.43) were well tolerated and were critical for the antagonist activity of VUF 10085 but not for that of TAK-779. CONCLUSIONS AND IMPLICATIONS This more detailed definition of a binding pocket within CXCR3 for low MW antagonists should facilitate the rational design of newer CXCR3 antagonists, with obvious clinical potential.
Collapse
Affiliation(s)
- Belinda Nedjai
- Leukocyte Biology Section, NHLI Division, Faculty of Medicine, Imperial College, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bernat V, Brox R, Heinrich MR, Auberson YP, Tschammer N. Ligand-Biased and Probe-Dependent Modulation of Chemokine Receptor CXCR3 Signaling by Negative Allosteric Modulators. ChemMedChem 2015; 10:566-74. [DOI: 10.1002/cmdc.201402507] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/14/2015] [Indexed: 11/10/2022]
|
42
|
Bernat V, Admas TH, Brox R, Heinemann FW, Tschammer N. Boronic acids as probes for investigation of allosteric modulation of the chemokine receptor CXCR3. ACS Chem Biol 2014; 9:2664-77. [PMID: 25233453 DOI: 10.1021/cb500678c] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The chemokine receptor CXCR3 is a G protein-coupled receptor, which conveys extracellular signals into cells by changing its conformation upon agonist binding. To facilitate the mechanistic understanding of allosteric modulation of CXCR3, we combined computational modeling with the synthesis of novel chemical tools containing boronic acid moiety, site-directed mutagenesis, and detailed functional characterization. The design of boronic acid derivatives was based on the predictions from homology modeling and docking. The choice of the boronic acid moiety was dictated by its unique ability to interact with proteins in a reversible covalent way, thereby influencing conformational dynamics of target biomolecules. During the synthesis of the library we have developed a novel approach for the purification of drug-like boronic acids. To validate the predicted binding mode and to identify amino acid residues responsible for the transduction of signal through CXCR3, we conducted a site-directed mutagenesis study. With the use of allosteric radioligand RAMX3 we were able to establish the existence of a second allosteric binding pocket in CXCR3, which enables different binding modes of structurally closely related allosteric modulators of CXCR3. We have also identified residues Trp109(2.60) and Lys300(7.35) inside the transmembrane bundle of the receptor as crucial for the regulation of the G protein activation. Furthermore, we report the boronic acid 14 as the first biased negative allosteric modulator of the receptor. Overall, our data demonstrate that boronic acid derivatives represent an outstanding tool for determination of key receptor-ligand interactions and induction of ligand-biased signaling.
Collapse
Affiliation(s)
- Viachaslau Bernat
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Tizita Haimanot Admas
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Regine Brox
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Frank W. Heinemann
- Department
of Chemistry and Pharmacy, Inorganic Chemistry, Friedrich Alexander University, Egerlandstraße 1, 91058 Erlangen, Germany
| | - Nuska Tschammer
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| |
Collapse
|
43
|
Abstract
UNLABELLED The encephalitic response to viral infection requires local chemokine production and the ensuing recruitment of immune and inflammatory leukocytes. Accordingly, chemokine receptors present themselves as plausible therapeutic targets for drugs aimed at limiting encephalitic responses. However, it remains unclear which chemokines are central to this process and whether leukocyte recruitment is important for limiting viral proliferation and survival in the brain or whether it is predominantly a driver of coincident inflammatory pathogenesis. Here we examine chemokine expression and leukocyte recruitment in the context of avirulent and virulent Semliki Forest virus (SFV) as well as West Nile virus infection and demonstrate rapid and robust expression of a variety of inflammatory CC and CXC chemokines in all models. On this basis, we define a chemokine axis involved in leukocyte recruitment to the encephalitic brain during SFV infection. CXCR3 is the most active; CCR2 is also active but less so, and CCR5 plays only a modest role in leukocyte recruitment. Importantly, inhibition of each of these receptors individually and the resulting suppression of leukocyte recruitment to the infected brain have no effect on viral titer or survival following infection with a virulent SFV strain. In contrast, simultaneous blockade of CXCR3 and CCR2 results in significantly reduced mortality in response to virulent SFV infection. In summary, therefore, our data provide an unprecedented level of insight into chemokine orchestration of leukocyte recruitment in viral encephalitis. Our data also highlight CXCR3 and CCR2 as possible therapeutic targets for limiting inflammatory damage in response to viral infection of the brain. IMPORTANCE Brain inflammation (encephalitis) in response to viral infection can lead to severe illness and even death. This therefore represents an important clinical problem and one that requires the development of new therapeutic approaches. Central to the pathogenesis of encephalitis is the recruitment of inflammatory leukocytes to the infected brain, a process driven by members of the chemokine family. Here we provide an in-depth analysis of the chemokines involved in leukocyte recruitment to the virally infected brain and demonstrate that simultaneous blockade of two of these receptors, namely, CXCR3 and CCR2, does not alter viral titers within the brain but markedly reduces inflammatory leukocyte recruitment and enhances survival in a murine model of lethal viral encephalitis. Our results therefore highlight chemokine receptors as plausible therapeutic targets in treating viral encephalitis.
Collapse
|
44
|
Exploring the CXCR3 Chemokine Receptor with Small-Molecule Antagonists and Agonists. TOPICS IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1007/7355_2014_75] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
45
|
Scholten DJ, Roumen L, Wijtmans M, Verkade-Vreeker MCA, Custers H, Lai M, de Hooge D, Canals M, de Esch IJP, Smit MJ, de Graaf C, Leurs R. Identification of overlapping but differential binding sites for the high-affinity CXCR3 antagonists NBI-74330 and VUF11211. Mol Pharmacol 2014; 85:116-26. [PMID: 24174496 DOI: 10.1124/mol.113.088633] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
CXC chemokine receptor CXCR3 and/or its main three ligands CXCL9, CXCL10, and CXCL11 are highly upregulated in a variety of diseases. As such, considerable efforts have been made to develop small-molecule receptor CXCR3 antagonists, yielding distinct chemical classes of antagonists blocking binding and/or function of CXCR3 chemokines. Although it is suggested that these compounds bind in an allosteric fashion, thus far no evidence has been provided regarding the molecular details of their interaction with CXCR3. Using site-directed mutagenesis complemented with in silico homology modeling, we report the binding modes of two high-affinity CXCR3 antagonists of distinct chemotypes: VUF11211 [(S)-5-chloro-6-(4-(1-(4-chlorobenzyl)piperidin-4-yl)-3-ethylpiperazin-1-yl)-N-ethylnicotinamide] (piperazinyl-piperidine) with a rigid elongated structure containing two basic groups and NBI-74330 [(R)-N-(1-(3-(4-ethoxyphenyl)-4-oxo-3,4-dihydropyrido[2,3-d]pyrimidin-2-yl)ethyl)-2-(4-fluoro-3-(trifluoromethyl)phenyl)-N-(pyridin-3-ylmethyl)acetamide] (8-azaquinazolinone) without any basic group. Here we show that NBI-74330 is anchored in the transmembrane minor pocket lined by helices 2 (W2.60, D2.63), 3 (F3.32), and 7 (S7.39, Y7.43), whereas VUF11211 extends from the minor pocket into the major pocket of the transmembrane domains, located between residues in helices 1 (Y1.39), 2 (W2.60), 3 (F3.32), 4 (D4.60), 6 (Y6.51), and 7 (S7.39, Y7.43). Mutation of these residues did not affect CXCL11 binding significantly, confirming the allosteric nature of the interaction of these small molecules with CXCR3. Moreover, the model derived from our in silico-guided studies fits well with the already published structure-activity relationship data on these ligands. Altogether, in this study, we show overlapping, yet different binding sites for two high-affinity CXCR3 antagonists, which offer new opportunities for the structure-based design of allosteric modulators for CXCR3.
Collapse
Affiliation(s)
- Danny J Scholten
- Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines, and Systems, VU University Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
A role for the CXCR3/CXCL10 axis in Rasmussen encephalitis. Pediatr Neurol 2013; 49:451-457.e1. [PMID: 24080276 DOI: 10.1016/j.pediatrneurol.2013.07.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 07/22/2013] [Accepted: 07/30/2013] [Indexed: 01/30/2023]
Abstract
BACKGROUND Rasmussen encephalitis is a devastating pediatric syndrome of unknown etiology that is characterized by progressive loss of neurological function and intractable focal epilepsy. Cytotoxic T lymphocytes have an active role in the pathogenic process of Rasmussen encephalitis. We studied the implication of CXCL10-CXCR3, a chemotactic axis involved in the pathogenesis of several cases of immune encephalitis. METHODS We analyzed surgical specimens of children with Rasmussen encephalitis, and performed functional in vitro assays to test the implications of the pathological findings. RESULTS We found that cytotoxic T lymphocytes infiltrating the damaged areas of primary biopsies expressed CXCR3, whereas neurons and astrocytes in the same areas expressed CXCL10. The in vitro assays demonstrated we found that astrocytes upregulated the expression of CXCL10 messenger RNA and the release of CXCL10 to the supernatants on stimulation with polyinosinic-polycyticylic acid, a synthetic double-stranded RNA that mimics infections with either RNA or DNA viruses. Activated T lymphocytes responded to the production of CXCL10 by astrocytes by increasing their migration in a transwell assay. Finally, the chemotaxis induced by the stimulated astrocytes was completely abrogated in the presence of a small molecule antagonist of CXCR3. CONCLUSIONS Our results suggest that the CXCR3-CXCL10 axis has a role in recruiting pathogenic T lymphocytes into the brains of patients with Rasmussen encephalitis. This chemotactic mechanism may be targeted pharmacologically.
Collapse
|
47
|
Watts AO, van Lipzig MMH, Jaeger WC, Seeber RM, van Zwam M, Vinet J, van der Lee MMC, Siderius M, Zaman GJR, Boddeke HWGM, Smit MJ, Pfleger KDG, Leurs R, Vischer HF. Identification and profiling of CXCR3-CXCR4 chemokine receptor heteromer complexes. Br J Pharmacol 2013; 168:1662-74. [PMID: 23170857 DOI: 10.1111/bph.12064] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 09/29/2012] [Accepted: 10/29/2012] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE The C-X-C chemokine receptors 3 (CXCR3) and C-X-C chemokine receptors 4 (CXCR4) are involved in various autoimmune diseases and cancers. Small antagonists have previously been shown to cross-inhibit chemokine binding to CXCR4, CC chemokine receptors 2 (CCR2) and 5 (CCR5) heteromers. We investigated whether CXCR3 and CXCR4 can form heteromeric complexes and the binding characteristics of chemokines and small ligand compounds to these chemokine receptor heteromers. EXPERIMENTAL APPROACH CXCR3-CXCR4 heteromers were identified in HEK293T cells using co-immunoprecipitation, time-resolved fluorescence resonance energy transfer, saturation BRET and the GPCR-heteromer identification technology (HIT) approach. Equilibrium competition binding and dissociation experiments were performed to detect negative binding cooperativity. KEY RESULTS We provide evidence that chemokine receptors CXCR3 and CXCR4 form heteromeric complexes in HEK293T cells. Chemokine binding was mutually exclusive on membranes co-expressing CXCR3 and CXCR4 as revealed by equilibrium competition binding and dissociation experiments. The small CXCR3 agonist VUF10661 impaired binding of CXCL12 to CXCR4, whereas small antagonists were unable to cross-inhibit chemokine binding to the other chemokine receptor. In contrast, negative binding cooperativity between CXCR3 and CXCR4 chemokines was not observed in intact cells. However, using the GPCR-HIT approach, we have evidence for specific β-arrestin2 recruitment to CXCR3-CXCR4 heteromers in response to agonist stimulation. CONCLUSIONS AND IMPLICATIONS This study indicates that heteromeric CXCR3-CXCR4 complexes may act as functional units in living cells, which potentially open up novel therapeutic opportunities.
Collapse
Affiliation(s)
- A O Watts
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bachelerie F, Ben-Baruch A, Burkhardt AM, Combadiere C, Farber JM, Graham GJ, Horuk R, Sparre-Ulrich AH, Locati M, Luster AD, Mantovani A, Matsushima K, Murphy PM, Nibbs R, Nomiyama H, Power CA, Proudfoot AEI, Rosenkilde MM, Rot A, Sozzani S, Thelen M, Yoshie O, Zlotnik A. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol Rev 2013; 66:1-79. [PMID: 24218476 PMCID: PMC3880466 DOI: 10.1124/pr.113.007724] [Citation(s) in RCA: 691] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Sixteen years ago, the Nomenclature Committee of the International Union of Pharmacology approved a system for naming human seven-transmembrane (7TM) G protein-coupled chemokine receptors, the large family of leukocyte chemoattractant receptors that regulates immune system development and function, in large part by mediating leukocyte trafficking. This was announced in Pharmacological Reviews in a major overview of the first decade of research in this field [Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, and Power CA (2000) Pharmacol Rev 52:145-176]. Since then, several new receptors have been discovered, and major advances have been made for the others in many areas, including structural biology, signal transduction mechanisms, biology, and pharmacology. New and diverse roles have been identified in infection, immunity, inflammation, development, cancer, and other areas. The first two drugs acting at chemokine receptors have been approved by the U.S. Food and Drug Administration (FDA), maraviroc targeting CCR5 in human immunodeficiency virus (HIV)/AIDS, and plerixafor targeting CXCR4 for stem cell mobilization for transplantation in cancer, and other candidates are now undergoing pivotal clinical trials for diverse disease indications. In addition, a subfamily of atypical chemokine receptors has emerged that may signal through arrestins instead of G proteins to act as chemokine scavengers, and many microbial and invertebrate G protein-coupled chemokine receptors and soluble chemokine-binding proteins have been described. Here, we review this extended family of chemokine receptors and chemokine-binding proteins at the basic, translational, and clinical levels, including an update on drug development. We also introduce a new nomenclature for atypical chemokine receptors with the stem ACKR (atypical chemokine receptor) approved by the Nomenclature Committee of the International Union of Pharmacology and the Human Genome Nomenclature Committee.
Collapse
Affiliation(s)
- Francoise Bachelerie
- Chair, Subcommittee on Chemokine Receptors, Nomenclature Committee-International Union of Pharmacology, Bldg. 10, Room 11N113, NIH, Bethesda, MD 20892.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Vinet J, van Zwam M, Dijkstra IM, Brouwer N, van Weering HRJ, Watts A, Meijer M, Fokkens MR, Kannan V, Verzijl D, Vischer HF, Smit MJ, Leurs R, Biber K, Boddeke HWGM. Inhibition of CXCR3-mediated chemotaxis by the human chemokine receptor-like protein CCX-CKR. Br J Pharmacol 2013; 168:1375-87. [PMID: 23121557 DOI: 10.1111/bph.12042] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 09/28/2012] [Accepted: 10/01/2012] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Induction of cellular migration is the primary effect of chemokine receptor activation. However, several chemokine receptor-like proteins bind chemokines without subsequent induction of intracellular signalling and chemotaxis. It has been suggested that they act as chemokine scavengers, which may control local chemokine levels and contribute to the function of chemokines during inflammation. This has been verified for the chemokine-like receptor proteins D6 and DARC as well as CCX-CKR. Here, we provide evidence for an additional biological function of human (h)CCX-CKR. EXPERIMENTAL APPROACH We used transfection strategies in HEK293 and human T cells. KEY RESULTS Co-expression of hCCX-CKR completely inhibits hCXCR3-induced chemotaxis. We found that hCCX-CKR forms complexes with hCXCR3, suggesting a relationship between CCX-CKR heteromerization and inhibition of chemotaxis. Moreover, negative binding cooperativity induced by ligands both for hCXCR3 and hCCX-CKR was observed in cells expressing both receptors. This negative cooperativity may also explain the hCCX-CKR-induced inhibition of chemotaxis. CONCLUSIONS AND IMPLICATIONS These findings suggest that hCCX-CKR prevents hCXCR3-induced chemotaxis by heteromerization thus representing a novel mechanism of regulation of immune cell migration.
Collapse
Affiliation(s)
- J Vinet
- Department of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Laragione T, Brenner M, Sherry B, Gulko PS. CXCL10 and its receptor CXCR3 regulate synovial fibroblast invasion in rheumatoid arthritis. ACTA ACUST UNITED AC 2013; 63:3274-83. [PMID: 21811993 DOI: 10.1002/art.30573] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE CXCL10 is expressed in increased levels in highly invasive fibroblast-like synoviocytes (FLS) from arthritic DA rats and from patients with rheumatoid arthritis (RA). This study was undertaken to analyze the role of CXCL10 and its receptor CXCR3 in regulation of the invasive properties of FLS. METHODS FLS were isolated from synovial tissue of RA patients and from DA rats and arthritis-resistant DA.F344(Cia5d) rats with pristane-induced arthritis. We used an in vitro model of invasion through Matrigel, which has been shown to correlate with articular damage in RA and in rat arthritis. FLS were cultured in the presence or absence of CXCL10, anti-CXCR3 antibody, or the CXCR3 inhibitor AMG487 and then studied for invasion, matrix metalloproteinase (MMP) production (MMPs 1-3), intracellular calcium influx, and cell morphology. RESULTS DA rat FLS produced higher levels of CXCL10 compared with minimally invasive FLS from DA.F344(Cia5d) rats. CXCL10 treatment increased the invasiveness of FLS from DA.F344(Cia5d) rats by 2-fold, and this increase was blocked by anti-CXCR3. Both anti-CXCR3 and AMG487 reduced invasiveness of FLS from DA rats, by as much as 77%. AMG487 significantly reduced invasiveness of RA FLS (by 58%). CXCR3 blockade reduced levels of MMP-1 by 65%, inhibited receptor signaling (64-100% reduction in intracellular calcium influx), and interfered with actin cytoskeleton reorganization and lamellipodia formation in FLS from rats and RA patients. CONCLUSION We describe and characterize a new autocrine/paracrine role of CXCL10/CXCR3 in the regulation of FLS invasion in rats with arthritis and in RA patients. These observations suggest that the CXCL10/CXCR3 axis is a potential new target for therapies aimed at reducing FLS invasion and its associated joint damage and pannus invasion and destruction in RA.
Collapse
|