1
|
Cao C, Xu M, Wei Y, Peng T, Lin S, Liu X, Xu Y, Chu T, Liu S, Wu P, Hu B, Ding W, Li L, Ma D, Wu P. CXCR4 orchestrates the TOX-programmed exhausted phenotype of CD8 + T cells via JAK2/STAT3 pathway. CELL GENOMICS 2024; 4:100659. [PMID: 39317187 DOI: 10.1016/j.xgen.2024.100659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/17/2024] [Accepted: 08/22/2024] [Indexed: 09/26/2024]
Abstract
Evidence from clinical trials suggests that CXCR4 antagonists enhance immunotherapy effectiveness in several cancers. However, the specific mechanisms through which CXCR4 contributes to immune cell phenotypes are not fully understood. Here, we employed single-cell transcriptomic analysis and identified CXCR4 as a marker gene in T cells, with CD8+PD-1high exhausted T (Tex) cells exhibiting high CXCR4 expression. By blocking CXCR4, the Tex phenotype was attenuated in vivo. Mechanistically, CXCR4-blocking T cells mitigated the Tex phenotype by regulating the JAK2-STAT3 pathway. Single-cell RNA/TCR/ATAC-seq confirmed that Cxcr4-deficient CD8+ T cells epigenetically mitigated the transition from functional to exhausted phenotypes. Notably, clinical sample analysis revealed that CXCR4+CD8+ T cells showed higher expression in patients with a non-complete pathological response. Collectively, these findings demonstrate the mechanism by which CXCR4 orchestrates CD8+ Tex cells and provide a rationale for combining CXCR4 antagonists with immunotherapy in clinical trials.
Collapse
Affiliation(s)
- Canhui Cao
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Miaochun Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ye Wei
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ting Peng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shitong Lin
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaojie Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yashi Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Tian Chu
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shiyi Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ping Wu
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Bai Hu
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wencheng Ding
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Li Li
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ding Ma
- Department of Gynecologic Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Peng Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430199, China; National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
2
|
G SK, N K, Elumalai E, Gupta KK. Identification of CXCR4 inhibitors as a key therapeutic small molecule in renal fibrosis. J Biomol Struct Dyn 2024; 42:8441-8453. [PMID: 37592737 DOI: 10.1080/07391102.2023.2246575] [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: 01/11/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
The final stage of almost all chronic kidney diseases is renal fibrosis. Simple wounds or persistent inflammation can cause tissue inflammation, which, in the case of the kidney, results in scarring. Vascular sclerosis, tubulointerstitial fibrosis and glomerular fibrosis are all types of kidney fibrosis. Renal damage and fibrosis are caused by elevated expression of CXCR4. This study aimed to identify possible pharmacological agents which could bind to and inhibit isoform I of CXCR4 and determine their strength of interactions. The I-TASSER, Galaxyweb and Robetta were used to predict and refine the structure of the CXCR4 protein. ModBase was used to improve the loops, and then the quality was evaluated by using the ERRAT value (92.15) and Ramachandran plot. The improved 3D structure was subjected to small molecule database docking using Maestro (from Schrodinger) and the glide module. GROMACS was used to simulate molecules with the three top low glide scores and the best ADME properties. The best glide score was achieved by ligand ID 4990 (-11.5). Simulations, free energy landscape and residue decomposition analysis revealed that 4990 interacted more consistently with CXCR4 than the other two small molecules.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Senthil Kumar G
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Kishore N
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Elakkiya Elumalai
- Centre for Bioinformatics, Pondicherry University, Pondicherry, India
| | - Krishna Kant Gupta
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| |
Collapse
|
3
|
Pattarachotanant N, Sukjamnong S, Rangsinth P, Chaikhong K, Sillapachaiyaporn C, Leung GPH, Hu VW, Sarachana T, Chuchawankul S, Tencomnao T, Prasansuklab A. Aquilaria crassna Extract Exerts Neuroprotective Effect against Benzo[a]pyrene-Induced Toxicity in Human SH-SY5Y Cells: An RNA-Seq-Based Transcriptome Analysis. Nutrients 2024; 16:2727. [PMID: 39203863 PMCID: PMC11357018 DOI: 10.3390/nu16162727] [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: 07/27/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
Benzo[a]pyrene (B[a]P) is known to inhibit neurodifferentiation and induce neurodegeneration. Agarwood or Aquilaria crassna (AC), a plant with health-promoting properties, may counteract the neurotoxic effects of B[a]P by promoting neuronal growth and survival. This study investigated the protective effect of AC leaf ethanolic extract (ACEE) on the B[a]P-induced impairment of neuronal differentiation. A transcriptomic analysis identified the canonical pathway, the biological network, and the differentially expressed genes (DEGs) that are changed in response to neuronal differentiation and neurogenesis. Several genes, including CXCR4, ENPP2, GAP43, GFRA2, NELL2, NFASC, NSG2, NGB, BASP1, and NEUROD1, in B[a]P-treated SH-SY5Y cells were up-regulated after treatment with ACEE. Notably, a Western blot analysis further confirmed that ACEE increased the protein levels of GAP43 and neuroglobin. B[a]P treatment led to decreased phosphorylation of Akt and increased phosphorylation of ERK in SH-SY5Y cells; however, ACEE was able to reverse these effects. Clionasterol and lupenone were identified in ACEE. Molecular docking showed that these two phytochemicals had significant interactions with CXCR4, GDNF family receptor alpha (GFRA), and retinoid X receptors (RXRs). In conclusion, ACEE may be a potential alternative medicine for the prevention of impaired neuronal differentiation and neurodegenerative diseases.
Collapse
Affiliation(s)
- Nattaporn Pattarachotanant
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura), Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Suporn Sukjamnong
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Chulalongkorn Autism Research and Innovation Center of Excellence (Chula ACE), Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panthakarn Rangsinth
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kamonwan Chaikhong
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura), Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chanin Sillapachaiyaporn
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura), Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Valerie W Hu
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Tewarit Sarachana
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Chulalongkorn Autism Research and Innovation Center of Excellence (Chula ACE), Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Siriporn Chuchawankul
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Tewin Tencomnao
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura), Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Anchalee Prasansuklab
- Center of Excellence on Natural Products for Neuroprotection and Anti-Ageing (Neur-Age Natura), Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- College of Public Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
4
|
Ruminski PG, Rettig MP, DiPersio JF. Development of VLA4 and CXCR4 Antagonists for the Mobilization of Hematopoietic Stem and Progenitor Cells. Biomolecules 2024; 14:1003. [PMID: 39199390 PMCID: PMC11353233 DOI: 10.3390/biom14081003] [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: 06/28/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 09/01/2024] Open
Abstract
The treatment of patients diagnosed with hematologic malignancies typically includes hematopoietic stem cell transplantation (HSCT) as part of a therapeutic standard of care. The primary graft source of hematopoietic stem and progenitor cells (HSPCs) for HSCT is mobilized from the bone marrow into the peripheral blood of allogeneic donors or patients. More recently, these mobilized HSPCs have also been the source for gene editing strategies to treat diseases such as sickle-cell anemia. For a HSCT to be successful, it requires the infusion of a sufficient number of HSPCs that are capable of adequate homing to the bone marrow niche and the subsequent regeneration of stable trilineage hematopoiesis in a timely manner. Granulocyte-colony-stimulating factor (G-CSF) is currently the most frequently used agent for HSPC mobilization. However, it requires five or more daily infusions to produce an adequate number of HSPCs and the use of G-CSF alone often results in suboptimal stem cell yields in a significant number of patients. Furthermore, there are several undesirable side effects associated with G-CSF, and it is contraindicated for use in sickle-cell anemia patients, where it has been linked to serious vaso-occlusive and thrombotic events. The chemokine receptor CXCR4 and the cell surface integrin α4β1 (very late antigen 4 (VLA4)) are both involved in the homing and retention of HSPCs within the bone marrow microenvironment. Preclinical and/or clinical studies have shown that targeted disruption of the interaction of the CXCR4 or VLA4 receptors with their endogenous ligands within the bone marrow niche results in the rapid and reversible mobilization of HSPCs into the peripheral circulation and is synergistic when combined with G-CSF. In this review, we discuss the roles CXCR4 and VLA4 play in bone marrow homing and retention and will summarize more recent development of small-molecule CXCR4 and VLA4 inhibitors that, when combined, can synergistically improve the magnitude, quality and convenience of HSPC mobilization for stem cell transplantation and ex vivo gene therapy after the administration of just a single dose. This optimized regimen has the potential to afford a superior alternative to G-CSF for HSPC mobilization.
Collapse
Affiliation(s)
| | | | - John F. DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave., St Louis, MO 63105, USA
| |
Collapse
|
5
|
Abdoul-Azize S, Hami R, Riou G, Derambure C, Charbonnier C, Vannier JP, Guzman ML, Schneider P, Boyer O. Glucocorticoids paradoxically promote steroid resistance in B cell acute lymphoblastic leukemia through CXCR4/PLC signaling. Nat Commun 2024; 15:4557. [PMID: 38811530 PMCID: PMC11136999 DOI: 10.1038/s41467-024-48818-9] [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: 02/15/2023] [Accepted: 05/15/2024] [Indexed: 05/31/2024] Open
Abstract
Glucocorticoid (GC) resistance in childhood relapsed B-cell acute lymphoblastic leukemia (B-ALL) represents an important challenge. Despite decades of clinical use, the mechanisms underlying resistance remain poorly understood. Here, we report that in B-ALL, GC paradoxically induce their own resistance by activating a phospholipase C (PLC)-mediated cell survival pathway through the chemokine receptor, CXCR4. We identify PLC as aberrantly activated in GC-resistant B-ALL and its inhibition is able to induce cell death by compromising several transcriptional programs. Mechanistically, dexamethasone (Dex) provokes CXCR4 signaling, resulting in the activation of PLC-dependent Ca2+ and protein kinase C signaling pathways, which curtail anticancer activity. Treatment with a CXCR4 antagonist or a PLC inhibitor improves survival of Dex-treated NSG mice in vivo. CXCR4/PLC axis inhibition significantly reverses Dex resistance in B-ALL cell lines (in vitro and in vivo) and cells from Dex resistant ALL patients. Our study identifies how activation of the PLC signalosome in B-ALL by Dex limits the upfront efficacy of this chemotherapeutic agent.
Collapse
Affiliation(s)
| | - Rihab Hami
- Univ Brest, Inserm, UMR 1101, F-29200, Brest, France
| | - Gaetan Riou
- Univ Rouen Normandie, Inserm, UMR 1234, F-76000, Rouen, France
| | | | | | | | - Monica L Guzman
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Pascale Schneider
- Univ Rouen Normandie, Inserm, UMR 1234, F-76000, Rouen, France
- Rouen University Hospital, Department of Pediatric Immuno-Hemato-Oncology, F-76000, Rouen, France
| | - Olivier Boyer
- Univ Rouen Normandie, Inserm, UMR 1234, F-76000, Rouen, France
- Rouen University Hospital, Department of Immunology and Biotherapy, F-76000, Rouen, France
| |
Collapse
|
6
|
Stazi M, Megighian A, D'Este G, Negro S, Ivanušec A, Lonati D, Pirazzini M, Križaj I, Montecucco C. An agonist of CXCR4 induces a rapid recovery from the neurotoxic effects of Vipera ammodytes and Vipera aspis venoms. J Neurochem 2024; 168:428-440. [PMID: 36912731 DOI: 10.1111/jnc.15803] [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: 12/05/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 03/14/2023]
Abstract
People bitten by Alpine vipers are usually treated with antivenom antisera to prevent the noxious consequences caused by the injected venom. However, this treatment suffers from a number of drawbacks and additional therapies are necessary. The venoms of Vipera ammodytes and of Vipera aspis are neurotoxic and cause muscle paralysis by inducing neurodegeneration of motor axon terminals because they contain a presynaptic acting sPLA2 neurotoxin. We have recently found that any type of damage to motor axons is followed by the expression and activation of the intercellular signaling axis consisting of the CXCR4 receptor present on the membrane of the axon stump and of its ligand, the chemokine CXCL12 released by activated terminal Schwann cells. We show here that also V. ammodytes and V. aspis venoms cause the expression of the CXCL12-CXCR4 axis. We also show that a small molecule agonist of CXCR4, dubbed NUCC-390, induces a rapid regeneration of the motor axon terminal with functional recovery of the neuromuscular junction. These findings qualify NUCC-390 as a promising novel therapeutics capable of improving the recovery from the paralysis caused by the snakebite of the two neurotoxic Alpine vipers.
Collapse
Affiliation(s)
- M Stazi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - A Megighian
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - G D'Este
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - S Negro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - A Ivanušec
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
- Doctoral School, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - D Lonati
- Pavia Poison Control Centre - National Toxicology Information Centre - Clinical and Experimental Lab, Toxicology Unit, Istituti Clinici Scientifici Maugeri SpA SB IRCCS, Pavia, Italy
| | - M Pirazzini
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - I Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - C Montecucco
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- National Research Council, Institute of Neuroscience, Padova, Italy
| |
Collapse
|
7
|
Theme 07 - Pre-Clinical Therapeutic Strategies. Amyotroph Lateral Scler Frontotemporal Degener 2023; 24:173-191. [PMID: 37966323 DOI: 10.1080/21678421.2023.2260198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
|
8
|
Peddibhotla S, Caples K, Mehta A, Chen QY, Hu J, Idlett-Ali S, Zhang L, Zgheib C, Xu J, Liechty KW, Malany S. Triazolothiadiazine derivative positively modulates CXCR4 signaling and improves diabetic wound healing. Biochem Pharmacol 2023; 216:115764. [PMID: 37634595 PMCID: PMC11115308 DOI: 10.1016/j.bcp.2023.115764] [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: 07/02/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 08/29/2023]
Abstract
Development of specific therapies that target and accelerate diabetic wound repair is an urgent need to alleviate pain and suffering and the huge socioeconomic burden of this debilitating disease. C-X-C Motif Chemokine Ligand 12 (CXCL12) also know an stromal cell-derived factor 1α (SDF-1α) is a chemokine that binds the CXC chemokine receptor type 4 (CXCR4) and activates downstream signaling resulting in recruitment of hematopoietic cells to locations of tissue injury and promotes tissue repair. In diabetes, low expression of CXCL12 correlates with impaired wound healing. Activation of CXCR4 receptor signaling with agonists or positive allosteric modulators (PAMs) provides a potential for small molecule therapeutic discovery and development. We recently reported high throughput screening and identification of the CXCR4 partial agonist UCUF-728, characterization of in vitro activity and reduced wound closure time in diabetic mice at 100 μM as a proof-of-concept study. We report here, the discovery of a second chemical scaffold demonstrating increased agonist potency and represented by thiadiazine derivative, UCUF-965. UCUF-965 is a potent partial agonist of β-arrestin recruitment in CXCR4 receptor overexpressing cell line. Furthermore, UCUF-965 potentiates the CXCL12 maximal response in cAMP signaling pathway, activates CXCL12 stimulated migration in lymphoblast cells and modulates the levels of specific microRNA involved in the complex wound repair process, specifically in mouse fibroblasts. Our results indicate that UCUF-965 acts as a PAM agonist of the CXCR4 receptor. Furthermore, UCUF-965 enhanced angiogenesis markers and reduced wound healing time by 36% at 10.0 μM in diabetic mice models compared to untreated control.
Collapse
Affiliation(s)
| | - Karly Caples
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Alka Mehta
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Qi-Yin Chen
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Junyi Hu
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Shaquia Idlett-Ali
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver - Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Liping Zhang
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Carlos Zgheib
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver - Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA
| | - Junwang Xu
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver - Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA; Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Kenneth W Liechty
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver - Anschutz Medical Campus and Children's Hospital Colorado, Aurora, CO 80045, USA.
| | - Siobhan Malany
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
9
|
Yamane H, Ishida T. Helix encoder: a compound-protein interaction prediction model specifically designed for class A GPCRs. FRONTIERS IN BIOINFORMATICS 2023; 3:1193025. [PMID: 37304403 PMCID: PMC10250622 DOI: 10.3389/fbinf.2023.1193025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Class A G protein-coupled receptors (GPCRs) represent the largest class of GPCRs. They are essential targets of drug discovery and thus various computational approaches have been applied to predict their ligands. However, there are a large number of orphan receptors in class A GPCRs and it is difficult to use a general protein-specific supervised prediction scheme. Therefore, the compound-protein interaction (CPI) prediction approach has been considered one of the most suitable for class A GPCRs. However, the accuracy of CPI prediction is still insufficient. The current CPI prediction model generally employs the whole protein sequence as the input because it is difficult to identify the important regions in general proteins. In contrast, it is well-known that only a few transmembrane helices of class A GPCRs play a critical role in ligand binding. Therefore, using such domain knowledge, the CPI prediction performance could be improved by developing an encoding method that is specifically designed for this family. In this study, we developed a protein sequence encoder called the Helix encoder, which takes only a protein sequence of transmembrane regions of class A GPCRs as input. The performance evaluation showed that the proposed model achieved a higher prediction accuracy compared to a prediction model using the entire protein sequence. Additionally, our analysis indicated that several extracellular loops are also important for the prediction as mentioned in several biological researches.
Collapse
|
10
|
Furumiya T, Itokazu T, Nakanishi T, Yamashita T. CXCR4 signaling regulates repair Schwann cell infiltration into the spinal cord after spinal cord injury in mice. Neurosci Res 2022; 191:38-47. [PMID: 36592826 DOI: 10.1016/j.neures.2022.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/12/2022] [Accepted: 12/25/2022] [Indexed: 12/31/2022]
Abstract
Schwann cells are glial cells that myelinate neuronal axons in the peripheral nervous system (PNS). When the PNS is damaged, Schwann cells de-differentiate into p75-positive "repair Schwann cells," which contribute to neural circuit regeneration. Interestingly, Schwann cells in the dorsal roots are known to be reprogrammed to repair Schwann cells even after spinal cord injury (SCI) and then migrate into the injured spinal cord. However, the molecular mechanism underlying the migration of repair Schwann cells remains unknown. Since a recent in vitro study revealed the importance of CXCR4 signaling in Schwann cell migration, we investigated whether CXCR4 signaling is involved in the PNS-to-central nervous system (CNS) migration of repair Schwann cells after SCI. We revealed that repair Schwann cells express CXCR4, and its ligand CXCL12 is upregulated in the injured spinal cord. We also found that the pharmacological inhibition of CXCR4 signaling decreased the infiltration of repair Schwann cells. Moreover, CXCR4 agonist administration effectively increased the infiltration of repair Schwann cells along with improved motor function. These findings strongly suggest the involvement of CXCR4 signaling in the PNS-to-CNS migration of repair Schwann cells after SCI.
Collapse
Affiliation(s)
- Takeru Furumiya
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Toru Nakanishi
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka, Japan; Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| |
Collapse
|
11
|
Kumar N, Acharya V. Machine intelligence-driven framework for optimized hit selection in virtual screening. J Cheminform 2022; 14:48. [PMID: 35869511 PMCID: PMC9306080 DOI: 10.1186/s13321-022-00630-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractVirtual screening (VS) aids in prioritizing unknown bio-interactions between compounds and protein targets for empirical drug discovery. In standard VS exercise, roughly 10% of top-ranked molecules exhibit activity when examined in biochemical assays, which accounts for many false positive hits, making it an arduous task. Attempts for conquering false-hit rates were developed through either ligand-based or structure-based VS separately; however, nonetheless performed remarkably well. Here, we present an advanced VS framework—automated hit identification and optimization tool (A-HIOT)—comprises chemical space-driven stacked ensemble for identification and protein space-driven deep learning architectures for optimization of an array of specific hits for fixed protein receptors. A-HIOT implements numerous open-source algorithms intending to integrate chemical and protein space leading to a high-quality prediction. The optimized hits are the selective molecules which we retrieve after extreme refinement implying chemical space and protein space modules of A-HIOT. Using CXC chemokine receptor 4, we demonstrated the superior performance of A-HIOT for hit molecule identification and optimization with tenfold cross-validation accuracies of 94.8% and 81.9%, respectively. In comparison with other machine learning algorithms, A-HIOT achieved higher accuracies of 96.2% for hit identification and 89.9% for hit optimization on independent benchmark datasets for CXCR4 and 86.8% for hit identification and 90.2% for hit optimization on independent test dataset for androgen receptor (AR), thus, shows its generalizability and robustness. In conclusion, advantageous features impeded in A-HIOT is making a reliable approach for bridging the long-standing gap between ligand-based and structure-based VS in finding the optimized hits for the desired receptor. The complete resource (framework) code is available at https://gitlab.com/neeraj-24/A-HIOT.
Graphical Abstract
Collapse
|
12
|
Recovery from the Neuroparalysis Caused by the Micrurus nigrocinctus Venom Is Accelerated by an Agonist of the CXCR4 Receptor. Toxins (Basel) 2022; 14:toxins14080531. [PMID: 36006193 PMCID: PMC9412299 DOI: 10.3390/toxins14080531] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/30/2022] [Indexed: 12/10/2022] Open
Abstract
Snake envenoming is a major but neglected human disease in tropical and subtropical regions. Among venomous snakes in the Americas, coral snakes of the genus Micrurus are particularly dangerous because they cause a peripheral neuroparalysis that can persist for many days or, in severe cases, progress to death. Ventilatory support and the use of snake species-specific antivenoms may prevent death from respiratory paralysis in most cases. However, there is a general consensus that additional and non-expensive treatments that can be delivered even long after the snake bite are needed. Neurotoxic degeneration of peripheral motor neurons activates pro-regenerative intercellular signaling programs, the greatest of which consist of the chemokine CXCL12α, produced by perisynaptic Schwann cells, which act on the CXCR4 receptor expressed on damaged neuronal axons. We recently found that the CXCR4 agonist NUCC-390 promotes axonal growth. Here, we show that the venom of the highly neurotoxic snake Micrurus nigrocinctus causes a complete degeneration of motor axon terminals of the soleus muscle, followed by functional regeneration whose time course is greatly accelerated by NUCC-390. These results suggest that NUCC-390 is a potential candidate for treating human patients envenomed by Micrurus nigrocinctus as well as other neurotoxic Micrurus spp. in order to improve the recovery of normal neuromuscular physiology, thus reducing the mortality and hospital costs of envenoming.
Collapse
|
13
|
Hopkins BE, Masuho I, Ren D, Iyamu ID, Lv W, Malik N, Martemyanov KA, Schiltz GE, Miller RJ. Effects of Small Molecule Ligands on ACKR3 Receptors. Mol Pharmacol 2022; 102:128-138. [PMID: 35809897 PMCID: PMC9393849 DOI: 10.1124/molpharm.121.000295] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/31/2022] [Indexed: 11/30/2022] Open
Abstract
Chemokines such as stromal derived factor 1 and their G protein coupled receptors are well-known regulators of the development and functions of numerous tissues. C-X-C motif chemokine ligand 12 (CXCL12) has two receptors: C-X-C chemokine motif receptor 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3). ACKR3 has been described as an atypical “biased” receptor because it does not appear to signal through G proteins and, instead, signals solely through the β-arrestin pathway. In support of this conclusion, we have shown that ACKR3 is unable to signal through any of the known mammalian Gα isoforms and have generated a comprehensive map of the Gα activation by CXCL12/CXCR4. We also synthesized a series of small molecule ligands which acted as selective agonists for ACKR3 as assessed by their ability to recruit β-arrestin to the receptor. Using select point mutations, we studied the molecular characteristics that determine the ability of small molecules to activate ACKR3 receptors, revealing a key role for the deeper binding pocket composed of residues in the transmembrane domains of ACKR3. The development of more selective ACKR3 ligands should allow us to better appreciate the unique roles of ACKR3 in the CXCL12/CXCR4/ACKR3-signaling axis and better understand the structural determinants for ACKR3 activation.
Collapse
Affiliation(s)
| | - Ikuo Masuho
- Department of Neuroscience, The Scripps Research Institute Florida, United States
| | - Dongjun Ren
- Department of Pharmacology, Northwestern University, United States
| | - Iredia D Iyamu
- Center for Molecular Innovation and Drug Discovery, Northwestern University, United States
| | - Wei Lv
- Center for Molecular Innovation and Drug Discovery, Northwestern University, United States
| | - Neha Malik
- Center for Molecular Innovation and Drug Discovery, Northwestern University, United States
| | | | - Gary E Schiltz
- Center for Molecular Innovation and Drug Discovery, Department of Pharmacology, Department of Chemistry, and Robert H. Lurie Comprehensive Cancer Center, Northwestern University, United States
| | - Richard J Miller
- Department of Pharmacology, Northwestern University, United States
| |
Collapse
|
14
|
Stazi M, Fabris F, Tan KY, Megighian A, Rubini A, Mattarei A, Negro S, D'Este G, Lista F, Rossetto O, Tan CH, Montecucco C. An agonist of the CXCR4 receptor is therapeutic for the neuroparalysis induced by
Bungarus
snakes envenoming. Clin Transl Med 2022; 12:e651. [PMID: 35075797 PMCID: PMC8787102 DOI: 10.1002/ctm2.651] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Marco Stazi
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Federico Fabris
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Kae Yi Tan
- Department of Molecular Medicine Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| | - Aram Megighian
- Department of Biomedical Sciences University of Padova Padova Italy
| | | | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences University of Padova Padova Italy
| | - Samuele Negro
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Giorgia D'Este
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Florigio Lista
- Center of Medical and Veterinary Research of the Ministry of Defense Policlinico Militare Rome Italy
| | - Ornella Rossetto
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Choo Hock Tan
- Department of Pharmacology Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| | - Cesare Montecucco
- Department of Biomedical Sciences University of Padova Padova Italy
- CNR Institute of Neuroscience Padova Italy
| |
Collapse
|
15
|
Latrotoxin-Induced Neuromuscular Junction Degeneration Reveals Urocortin 2 as a Critical Contributor to Motor Axon Terminal Regeneration. Int J Mol Sci 2022; 23:ijms23031186. [PMID: 35163106 PMCID: PMC8835473 DOI: 10.3390/ijms23031186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 11/24/2022] Open
Abstract
We used α-Latrotoxin (α-LTx), the main neurotoxic component of the black widow spider venom, which causes degeneration of the neuromuscular junction (NMJ) followed by a rapid and complete regeneration, as a molecular tool to identify by RNA transcriptomics factors contributing to the structural and functional recovery of the NMJ. We found that Urocortin 2 (UCN2), a neuropeptide involved in the stress response, is rapidly expressed at the NMJ after acute damage and that inhibition of CRHR2, the specific receptor of UCN2, delays neuromuscular transmission rescue. Experiments in neuronal cultures show that CRHR2 localises at the axonal tips of growing spinal motor neurons and that its expression inversely correlates with synaptic maturation. Moreover, exogenous UCN2 enhances the growth of axonal sprouts in cultured neurons in a CRHR2-dependent manner, pointing to a role of the UCN2-CRHR2 axis in the regulation of axonal growth and synaptogenesis. Consistently, exogenous administration of UCN2 strongly accelerates the regrowth of motor axon terminals degenerated by α-LTx, thereby contributing to the functional recovery of neuromuscular transmission after damage. Taken together, our results posit a novel role for UCN2 and CRHR2 as a signalling axis involved in NMJ regeneration.
Collapse
|
16
|
Jacobsen D, Bushara O, Mishra RK, Sun L, Liao J, Yang GY. Druggable sites/pockets of the p53-DNAJA1 protein–protein interaction: In silico modeling and in vitro/in vivo validation. Methods Enzymol 2022; 675:83-107. [DOI: 10.1016/bs.mie.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
17
|
Ballante F, Kooistra AJ, Kampen S, de Graaf C, Carlsson J. Structure-Based Virtual Screening for Ligands of G Protein-Coupled Receptors: What Can Molecular Docking Do for You? Pharmacol Rev 2021; 73:527-565. [PMID: 34907092 DOI: 10.1124/pharmrev.120.000246] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
G protein-coupled receptors (GPCRs) constitute the largest family of membrane proteins in the human genome and are important therapeutic targets. During the last decade, the number of atomic-resolution structures of GPCRs has increased rapidly, providing insights into drug binding at the molecular level. These breakthroughs have created excitement regarding the potential of using structural information in ligand design and initiated a new era of rational drug discovery for GPCRs. The molecular docking method is now widely applied to model the three-dimensional structures of GPCR-ligand complexes and screen for chemical probes in large compound libraries. In this review article, we first summarize the current structural coverage of the GPCR superfamily and the understanding of receptor-ligand interactions at atomic resolution. We then present the general workflow of structure-based virtual screening and strategies to discover GPCR ligands in chemical libraries. We assess the state of the art of this research field by summarizing prospective applications of virtual screening based on experimental structures. Strategies to identify compounds with specific efficacy and selectivity profiles are discussed, illustrating the opportunities and limitations of the molecular docking method. Our overview shows that structure-based virtual screening can discover novel leads and will be essential in pursuing the next generation of GPCR drugs. SIGNIFICANCE STATEMENT: Extraordinary advances in the structural biology of G protein-coupled receptors have revealed the molecular details of ligand recognition by this large family of therapeutic targets, providing novel avenues for rational drug design. Structure-based docking is an efficient computational approach to identify novel chemical probes from large compound libraries, which has the potential to accelerate the development of drug candidates.
Collapse
Affiliation(s)
- Flavio Ballante
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden (F.B., S.K., J.C.); Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark (A.J.K.); and Sosei Heptares, Steinmetz Building, Granta Park, Great Abington, Cambridge, United Kingdom (C.d.G.)
| | - Albert J Kooistra
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden (F.B., S.K., J.C.); Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark (A.J.K.); and Sosei Heptares, Steinmetz Building, Granta Park, Great Abington, Cambridge, United Kingdom (C.d.G.)
| | - Stefanie Kampen
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden (F.B., S.K., J.C.); Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark (A.J.K.); and Sosei Heptares, Steinmetz Building, Granta Park, Great Abington, Cambridge, United Kingdom (C.d.G.)
| | - Chris de Graaf
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden (F.B., S.K., J.C.); Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark (A.J.K.); and Sosei Heptares, Steinmetz Building, Granta Park, Great Abington, Cambridge, United Kingdom (C.d.G.)
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden (F.B., S.K., J.C.); Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark (A.J.K.); and Sosei Heptares, Steinmetz Building, Granta Park, Great Abington, Cambridge, United Kingdom (C.d.G.)
| |
Collapse
|
18
|
Jia H, Neptune E, Cui H. Targeting ACE2 for COVID-19 Therapy: Opportunities and Challenges. Am J Respir Cell Mol Biol 2021; 64:416-425. [PMID: 33296619 PMCID: PMC8008810 DOI: 10.1165/rcmb.2020-0322ps] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/09/2020] [Indexed: 12/23/2022] Open
Abstract
The coronavirus disease (COVID-19) pandemic is sweeping the globe. Even with a number of effective vaccines being approved and available to the public, new cases and escalating mortality are climbing every day. ACE2 (angiotensin-converting enzyme 2) is the primary receptor for the COVID-19 causative virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and its complexation with spike proteins plays a crucial role in viral entry into host cells and the subsequent infection. Blocking this binding event or reducing the accessibility of the virus to the ACE2 receptor, represents an alternative strategy to prevent COVID-19. In addition, the biological significance of ACE2 in modulating the innate immune system and tissue repair cascades and anchors its therapeutic potential for treating the infected patients. In this viewpoint article, we review the current efforts of exploiting ACE2 as a therapeutic target to address this dire medical need. We also provide a holistic view of the pros and cons of each treatment strategy. We highlight the fundamental and translational challenges in moving these research endeavors to clinical applications.
Collapse
Affiliation(s)
- Hongpeng Jia
- Division of Pediatric Surgery, Department of Surgery
| | - Enid Neptune
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, and
- Institute for Nano Biotechnology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
19
|
Cadima-Couto I, Tauzin A, Freire JM, Figueira TN, Silva RDM, Pérez-Peinado C, Cunha-Santos C, Bártolo I, Taveira N, Gano L, Correia JDG, Goncalves J, Mammano F, Andreu D, Castanho MARB, Veiga AS. Anti-HIV-1 Activity of pepRF1, a Proteolysis-Resistant CXCR4 Antagonist Derived from Dengue Virus Capsid Protein. ACS Infect Dis 2021; 7:6-22. [PMID: 33319557 DOI: 10.1021/acsinfecdis.9b00507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
There is an urgent need for the development of new anti-HIV drugs that can complement existing medicines to be used against resistant strains. Here, we report the anti-HIV-1 peptide pepRF1, a human serum-resistant peptide derived from the Dengue virus capsid protein. In vitro, pepRF1 shows a 50% inhibitory concentration of 1.5 nM with a potential therapeutic window higher than 53 000. This peptide is specific for CXCR4-tropic strains, preventing viral entry into target cells by binding to the viral coreceptor CXCR4, acting as an antagonist of this receptor. pepRF1 is more effective than T20, the only peptide-based HIV-1 entry inhibitor approved, and excels in inhibiting a HIV-1 strain resistant to T20. Potentially, pepRF1 can be used alone or in combination with other anti-HIV drugs. Furthermore, one can also envisage its use as a novel therapeutic strategy for other CXCR4-related diseases.
Collapse
Affiliation(s)
- Iris Cadima-Couto
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Alexandra Tauzin
- INSERM UMR 1124, Université de Paris, 45 rue des Saints Pères, F-75006 Paris, France
| | - João M. Freire
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Tiago N. Figueira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Rúben D. M. Silva
- Centro de Ciências e Tecnologias Nucleares and Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal
| | - Clara Pérez-Peinado
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Catarina Cunha-Santos
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Inês Bártolo
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Nuno Taveira
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Universitário Egas Moniz, 2829-511 Monte de Caparica, Portugal
| | - Lurdes Gano
- Centro de Ciências e Tecnologias Nucleares and Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal
| | - João D. G. Correia
- Centro de Ciências e Tecnologias Nucleares and Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066 Bobadela LRS, Portugal
| | - Joao Goncalves
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Fabrizio Mammano
- INSERM UMR 1124, Université de Paris, 45 rue des Saints Pères, F-75006 Paris, France
| | - David Andreu
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, 08003 Barcelona, Spain
| | - Miguel A. R. B. Castanho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Ana Salomé Veiga
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
20
|
Tong X, Xu D, Mishra RK, Jones RD, Sun L, Schiltz GE, Liao J, Yang GY. Identification of a druggable protein-protein interaction site between mutant p53 and its stabilizing chaperone DNAJA1. J Biol Chem 2021; 296:100098. [PMID: 33208462 PMCID: PMC7948449 DOI: 10.1074/jbc.ra120.014749] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/26/2020] [Accepted: 11/18/2020] [Indexed: 12/15/2022] Open
Abstract
The TP53 gene is the most frequently mutated gene in human cancers, and the majority of TP53 mutations are missense mutations. As a result, these mutant p53 (mutp53) either directly lose wildtype p53 (wtp53) tumor suppressor function or exhibit a dominant negative effect over wtp53. In addition, some mutp53 have acquired new oncogenic function (gain of function). Therefore, targeting mutp53 for its degradation may serve as a promising strategy for cancer prevention and therapy. Based on our previous finding that farnesylated DNAJA1 is a crucial chaperone in maintaining mutp53 stabilization, and by using an in silico approach, we built 3D homology models of human DNAJA1 and mutp53R175H proteins, identified the interacting pocket in the DNAJA1-mutp53R175H complex, and found one critical druggable small molecule binding site in the DNAJA1 glycine/phenylalanine-rich region. We confirmed that the interacting pocket in the DNAJA1-mutp53R175H complex was crucial for stabilizing mutp53R175H using a site-directed mutagenesis approach. We further screened a drug-like library to identify a promising small molecule hit (GY1-22) against the interacting pocket in the DNAJA1-mutp53R175H complex. The GY1-22 compound displayed an effective activity against the DNAJA1-mutp53R175H complex. Treatment with GY1-22 significantly reduced mutp53 protein levels, enhanced Waf1p21 expression, suppressed cyclin D1 expression, and inhibited mutp53-driven pancreatic cancer growth both in vitro and in vivo. Together, our results indicate that the interacting pocket in the DNAJA1-mutp53R175H complex is critical for mutp53's stability and oncogenic function, and DNAJA1 is a robust therapeutic target for developing the efficient small molecule inhibitors against oncogenic mutp53.
Collapse
Affiliation(s)
- Xin Tong
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Dandan Xu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rama K Mishra
- Center for Molecular Innovation and Drug Discovery (CMIDD), Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ryan D Jones
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leyu Sun
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Gary E Schiltz
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Center for Molecular Innovation and Drug Discovery (CMIDD), Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jie Liao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Guang-Yu Yang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
21
|
Stazi M, D’Este G, Mattarei A, Negro S, Lista F, Rigoni M, Megighian A, Montecucco C. An agonist of the CXCR4 receptor accelerates the recovery from the peripheral neuroparalysis induced by Taipan snake envenomation. PLoS Negl Trop Dis 2020; 14:e0008547. [PMID: 32898186 PMCID: PMC7537909 DOI: 10.1371/journal.pntd.0008547] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/06/2020] [Accepted: 06/19/2020] [Indexed: 01/15/2023] Open
Abstract
Envenomation by snakes is a major neglected human disease. Hospitalization and use of animal-derived antivenom are the primary therapeutic supports currently available. There is consensus that additional, not expensive, treatments that can be delivered even long after the snake bite are needed. We recently showed that the drug dubbed NUCC-390 shortens the time of recovery from the neuroparalysis caused by traumatic or toxic degeneration of peripheral motor neurons. These syndromes are characterized by the activation of a pro-regenerative molecular axis, consisting of the CXCR4 receptor expressed at the damaged site in neuronal axons and by the release of its ligand CXCL12α, produced by surrounding Schwann cells. This intercellular signaling axis promotes axonal growth and functional recovery from paralysis. NUCC-390 is an agonist of CXCR4 acting similarly to CXCL12α. Here, we have tested its efficacy in a murine model of neuroparalytic envenoming by a Papuan Taipan (Oxyuranus scutellatus) where a degeneration of the motor axon terminals caused by the presynaptic PLA2 toxin Taipoxin, contained in the venom, occurs. Using imaging of the neuromuscular junction and electrophysiological analysis, we found that NUCC-390 administration after injection of either the purified neuroparalytic Taipoxin or the whole Taipan venom, significantly accelerates the recovery from paralysis. These results indicate that NUCC-390, which is non-toxic in mice, should be considered for trials in humans to test its efficacy in accelerating the recovery from the peripheral neuroparalysis induced by Taipans. NUCC-390 should be tested as well in the envenomation by other snakes that cause neuroparalytic syndromes in humans. NUCC-390 could become an additional treatment, common to many snake envenomings, that can be delivered after the bite to reduce death by respiratory deficits and to shorten and improve functional recovery.
Collapse
Affiliation(s)
- Marco Stazi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Giorgia D’Este
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Samuele Negro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Florigio Lista
- Department of Medical and Veterinary Research, the Ministry of Defense, Rome, Italy
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- CNR Institute of Neuroscience, Department of Biomedical Sciences, Padua, Italy
| |
Collapse
|
22
|
Bianchi ME, Mezzapelle R. The Chemokine Receptor CXCR4 in Cell Proliferation and Tissue Regeneration. Front Immunol 2020; 11:2109. [PMID: 32983169 PMCID: PMC7484992 DOI: 10.3389/fimmu.2020.02109] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
The CXCR4 receptor upon binding its ligands triggers multiple signaling pathways that orchestrate cell migration, hematopoiesis and cell homing, and retention in the bone marrow. However, CXCR4 also directly controls cell proliferation of non-hematopoietic cells. This review focuses on recent reports pointing to its pivotal role in tissue regeneration and stem cell activation, and discusses the connection to the known role of CXCR4 in promoting tumor growth. The mechanisms may be similar in all cases, since regeneration often recapitulates developmental processes, and cancer often exploits developmental pathways. Moreover, cell migration and cell proliferation appear to be downstream of the same signaling pathways. A deeper understanding of the complex signaling originating from CXCR4 is needed to exploit the opportunities to repair damaged organs safely and effectively.
Collapse
Affiliation(s)
- Marco E Bianchi
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Rosanna Mezzapelle
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
23
|
Murphy JW, Rajasekaran D, Merkel J, Skeens E, Keeler C, Hodsdon ME, Lisi GP, Lolis E. High-Throughput Screening of a Functional Human CXCL12-CXCR4 Signaling Axis in a Genetically Modified S. cerevisiae: Discovery of a Novel Up-Regulator of CXCR4 Activity. Front Mol Biosci 2020; 7:164. [PMID: 32766282 PMCID: PMC7378375 DOI: 10.3389/fmolb.2020.00164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/25/2020] [Indexed: 12/03/2022] Open
Abstract
CXCL12 activates CXCR4 and is involved in embryogenesis, hematopoiesis, and angiogenesis. It has pathological roles in HIV-1, WHIM disease, cancer, and autoimmune diseases. An antagonist, AMD3100, is used for the release of CD34+ hematopoietic stem cells from the bone marrow for autologous transplantation for lymphoma or multiple myeloma patients. Adverse effects are tolerated due to its short-term treatment, but AMD3100 is cardiotoxic in clinical studies for HIV-1. In an effort to determine whether Saccharomyces cerevisiae expressing a functional human CXCR4 could be used as a platform for identifying a ligand from a library of less ∼1,000 compounds, a high-throughput screening was developed. We report that 2-carboxyphenyl phosphate (fosfosal) up-regulates CXCR4 activation only in the presence of CXCL12. This is the first identification of a compound that increases CXCR4 activity by any mechanism. We mapped the fosfosal binding site on CXCL12, described its mechanism of action, and studied its chemical components, salicylate and phosphate, to conclude that they synergize to achieve the functional effect.
Collapse
Affiliation(s)
- James W Murphy
- Department of Pharmacology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Deepa Rajasekaran
- Department of Pharmacology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Janie Merkel
- Yale Center for Molecular Discovery, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Erin Skeens
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States
| | - Camille Keeler
- Department of Laboratory Medicine, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Michael E Hodsdon
- Department of Laboratory Medicine, Yale School of Medicine, Yale University, New Haven, CT, United States.,Yale Cancer Center, New Haven, CT, United States
| | - George P Lisi
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States
| | - Elias Lolis
- Department of Pharmacology, Yale School of Medicine, Yale University, New Haven, CT, United States.,Yale Cancer Center, New Haven, CT, United States
| |
Collapse
|
24
|
Tomassi S, Trotta AM, Ieranò C, Merlino F, Messere A, Rea G, Santoro F, Brancaccio D, Carotenuto A, D'Amore VM, Di Leva FS, Novellino E, Cosconati S, Marinelli L, Scala S, Di Maro S. Disulfide Bond Replacement with 1,4‐ and 1,5‐Disubstituted [1,2,3]‐Triazole on C‐X‐C Chemokine Receptor Type 4 (CXCR4) Peptide Ligands: Small Changes that Make Big Differences. Chemistry 2020; 26:10113-10125. [DOI: 10.1002/chem.202002468] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/29/2020] [Indexed: 01/10/2023]
Affiliation(s)
- Stefano Tomassi
- Department of Pharmacy University of Naples “Federico II” Via Domenico Montesano 49 80131 Naples Italy
| | - Anna Maria Trotta
- U.O.C. “Bersagli molecolari del microambiente” Istituto Nazionale Tumori—IRCCS—Fondazione “G. Pascale” Via M. Semmola 80131 Naples Italy
| | - Caterina Ieranò
- U.O.C. “Bersagli molecolari del microambiente” Istituto Nazionale Tumori—IRCCS—Fondazione “G. Pascale” Via M. Semmola 80131 Naples Italy
| | - Francesco Merlino
- Department of Pharmacy University of Naples “Federico II” Via Domenico Montesano 49 80131 Naples Italy
| | - Anna Messere
- DiSTABiF University of Campania “Luigi Vanvitelli” Via Vivaldi 43 81100 Caserta Italy
| | - Giuseppina Rea
- U.O.C. “Bersagli molecolari del microambiente” Istituto Nazionale Tumori—IRCCS—Fondazione “G. Pascale” Via M. Semmola 80131 Naples Italy
| | - Federica Santoro
- Department of Pharmacy University of Naples “Federico II” Via Domenico Montesano 49 80131 Naples Italy
| | - Diego Brancaccio
- Department of Pharmacy University of Naples “Federico II” Via Domenico Montesano 49 80131 Naples Italy
| | - Alfonso Carotenuto
- Department of Pharmacy University of Naples “Federico II” Via Domenico Montesano 49 80131 Naples Italy
| | - Vincenzo Maria D'Amore
- Department of Pharmacy University of Naples “Federico II” Via Domenico Montesano 49 80131 Naples Italy
| | - Francesco Saverio Di Leva
- Department of Pharmacy University of Naples “Federico II” Via Domenico Montesano 49 80131 Naples Italy
| | - Ettore Novellino
- Department of Pharmacy University of Naples “Federico II” Via Domenico Montesano 49 80131 Naples Italy
| | - Sandro Cosconati
- DiSTABiF University of Campania “Luigi Vanvitelli” Via Vivaldi 43 81100 Caserta Italy
| | - Luciana Marinelli
- Department of Pharmacy University of Naples “Federico II” Via Domenico Montesano 49 80131 Naples Italy
| | - Stefania Scala
- U.O.C. “Bersagli molecolari del microambiente” Istituto Nazionale Tumori—IRCCS—Fondazione “G. Pascale” Via M. Semmola 80131 Naples Italy
| | - Salvatore Di Maro
- DiSTABiF University of Campania “Luigi Vanvitelli” Via Vivaldi 43 81100 Caserta Italy
| |
Collapse
|
25
|
Discovery of novel aminopiperidinyl amide CXCR4 modulators through virtual screening and rational drug design. Eur J Med Chem 2020; 201:112479. [PMID: 32534343 DOI: 10.1016/j.ejmech.2020.112479] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/08/2020] [Accepted: 05/16/2020] [Indexed: 12/29/2022]
Abstract
The C-X-C chemokine receptor type 4 (CXCR4) is a potential therapeutic target for HIV infection, metastatic cancer, and inflammatory autoimmune diseases. In this study, we screened the ZINC chemical database for novel CXCR4 modulators through a series of in silico guided processes. After evaluating the screened compounds for their binding affinities to CXCR4 and inhibitory activities against the chemoattractant CXCL12, we identified a hit compound (ZINC 72372983) showing 100 nM affinity and 69% chemotaxis inhibition at the same concentration (100 nM). To increase the potency of our hit compound, we explored the protein-ligand interactions at an atomic level using molecular dynamics simulation which enabled us to design and synthesize a novel compound (Z7R) with nanomolar affinity (IC50 = 1.25 nM) and improved chemotaxis inhibition (78.5%). Z7R displays promising anti-inflammatory activity (50%) in a mouse edema model by blocking CXCR4-expressed leukocytes, being supported by our immunohistochemistry study.
Collapse
|
26
|
Perpiñá-Viciano C, Işbilir A, Zarca A, Caspar B, Kilpatrick LE, Hill SJ, Smit MJ, Lohse MJ, Hoffmann C. Kinetic Analysis of the Early Signaling Steps of the Human Chemokine Receptor CXCR4. Mol Pharmacol 2020; 98:72-87. [PMID: 32474443 PMCID: PMC7330677 DOI: 10.1124/mol.119.118448] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 05/06/2020] [Indexed: 01/14/2023] Open
Abstract
G protein–coupled receptors (GPCRs) are biologic switches that transduce extracellular stimuli into intracellular responses in the cell. Temporally resolving GPCR transduction pathways is key to understanding how cell signaling occurs. Here, we investigate the kinetics and dynamics of the activation and early signaling steps of the CXC chemokine receptor (CXCR) 4 in response to its natural ligands CXC chemokine ligand (CXCL) 12 and macrophage migration inhibitory factor (MIF), using Förster resonance energy transfer–based approaches. We show that CXCR4 presents a multifaceted response to CXCL12, with receptor activation (≈0.6 seconds) followed by a rearrangement in the receptor/G protein complex (≈1 seconds), a slower dimer rearrangement (≈1.7 seconds), and prolonged G protein activation (≈4 seconds). In comparison, MIF distinctly modulates every step of the transduction pathway, indicating distinct activation mechanisms and reflecting the different pharmacological properties of these two ligands. Our study also indicates that CXCR4 exhibits some degree of ligand-independent activity, a relevant feature for drug development.
Collapse
Affiliation(s)
- Cristina Perpiñá-Viciano
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Ali Işbilir
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Aurélien Zarca
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Birgit Caspar
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Laura E Kilpatrick
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Stephen J Hill
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Martine J Smit
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Martin J Lohse
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| | - Carsten Hoffmann
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), University Hospital Jena, University of Jena, Jena, Germany (C.P.-V., C.H.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.-V., A.I., M.J.L., C.H.); Max-Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Amsterdam Institute for Molecules Medicines and Systems (AIMMS), Division of Medicinal Chemistry, Vrije Universiteit, Amsterdam, The Netherlands (A.Z., M.J.S.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, United Kingdom (B.C., L.E.K., S.J.H.); and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., L.E.K., S.J.H.)
| |
Collapse
|
27
|
Dahlgren C, Holdfeldt A, Lind S, Mårtensson J, Gabl M, Björkman L, Sundqvist M, Forsman H. Neutrophil Signaling That Challenges Dogmata of G Protein-Coupled Receptor Regulated Functions. ACS Pharmacol Transl Sci 2020; 3:203-220. [PMID: 32296763 DOI: 10.1021/acsptsci.0c00004] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Indexed: 12/24/2022]
Abstract
Activation as well as recruitment of neutrophils, the most abundant leukocyte in human blood, to sites of infection/inflammation largely rely on surface-exposed chemoattractant receptors. These receptors belong to the family of 7-transmembrane domain receptors also known as G protein-coupled receptors (GPCRs) due to the fact that part of the downstream signaling relies on an activation of heterotrimeric G proteins. The neutrophil GPCRs share significant sequence homologies but bind many structurally diverse activating (agonistic) and inhibiting (antagonistic) ligands, ranging from fatty acids to purines, peptides, and lipopeptides. Recent structural and functional studies of neutrophil receptors have generated important information on GPCR biology in general; this knowledge aids in the overall understanding of general pharmacological principles, governing regulation of neutrophil function and inflammatory processes, including novel leukocyte receptor activities related to ligand recognition, biased/functional selective signaling, allosteric modulation, desensitization mechanisms and reactivation, and communication (cross-talk) between GPCRs. This review summarizes the recent discoveries and pharmacological hallmarks with focus on neutrophil GPCRs. In addition, unmet challenges are dealt with, including recognition by the receptors of diverse ligands and how biased signaling mediates different biological effects.
Collapse
Affiliation(s)
- Claes Dahlgren
- Department of Rheumatology and Inflammation Research, University of Göteborg, Göteborg 405 30, Sweden
| | - André Holdfeldt
- Department of Rheumatology and Inflammation Research, University of Göteborg, Göteborg 405 30, Sweden
| | - Simon Lind
- Department of Rheumatology and Inflammation Research, University of Göteborg, Göteborg 405 30, Sweden
| | - Jonas Mårtensson
- Department of Rheumatology and Inflammation Research, University of Göteborg, Göteborg 405 30, Sweden
| | - Michael Gabl
- Department of Rheumatology and Inflammation Research, University of Göteborg, Göteborg 405 30, Sweden
| | - Lena Björkman
- Department of Rheumatology and Inflammation Research, University of Göteborg, Göteborg 405 30, Sweden
| | - Martina Sundqvist
- Department of Rheumatology and Inflammation Research, University of Göteborg, Göteborg 405 30, Sweden
| | - Huamei Forsman
- Department of Rheumatology and Inflammation Research, University of Göteborg, Göteborg 405 30, Sweden
| |
Collapse
|
28
|
Eckardt V, Miller MC, Blanchet X, Duan R, Leberzammer J, Duchene J, Soehnlein O, Megens RT, Ludwig AK, Dregni A, Faussner A, Wichapong K, Ippel H, Dijkgraaf I, Kaltner H, Döring Y, Bidzhekov K, Hackeng TM, Weber C, Gabius HJ, von Hundelshausen P, Mayo KH. Chemokines and galectins form heterodimers to modulate inflammation. EMBO Rep 2020; 21:e47852. [PMID: 32080959 PMCID: PMC7132340 DOI: 10.15252/embr.201947852] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 01/14/2023] Open
Abstract
Chemokines and galectins are simultaneously upregulated and mediate leukocyte recruitment during inflammation. Until now, these effector molecules have been considered to function independently. Here, we tested the hypothesis that they form molecular hybrids. By systematically screening chemokines for their ability to bind galectin‐1 and galectin‐3, we identified several interacting pairs, such as CXCL12 and galectin‐3. Based on NMR and MD studies of the CXCL12/galectin‐3 heterodimer, we identified contact sites between CXCL12 β‐strand 1 and Gal‐3 F‐face residues. Mutagenesis of galectin‐3 residues involved in heterodimer formation resulted in reduced binding to CXCL12, enabling testing of functional activity comparatively. Galectin‐3, but not its mutants, inhibited CXCL12‐induced chemotaxis of leukocytes and their recruitment into the mouse peritoneum. Moreover, galectin‐3 attenuated CXCL12‐stimulated signaling via its receptor CXCR4 in a ternary complex with the chemokine and receptor, consistent with our structural model. This first report of heterodimerization between chemokines and galectins reveals a new type of interaction between inflammatory mediators that can underlie a novel immunoregulatory mechanism in inflammation. Thus, further exploration of the chemokine/galectin interactome is warranted.
Collapse
Affiliation(s)
- Veit Eckardt
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Michelle C Miller
- Department of Biochemistry, Molecular Biology & Biophysics, Health Sciences Center, University of Minnesota, Minneapolis, MN, USA
| | - Xavier Blanchet
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Rundan Duan
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Julian Leberzammer
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Johan Duchene
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Oliver Soehnlein
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Remco Ta Megens
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Anna-Kristin Ludwig
- Faculty of Veterinary Medicine, Institute of Physiological Chemistry, Ludwig-Maximilians-University, Munich, Germany
| | - Aurelio Dregni
- Department of Biochemistry, Molecular Biology & Biophysics, Health Sciences Center, University of Minnesota, Minneapolis, MN, USA
| | - Alexander Faussner
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Kanin Wichapong
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Hans Ippel
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Ingrid Dijkgraaf
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Herbert Kaltner
- Faculty of Veterinary Medicine, Institute of Physiological Chemistry, Ludwig-Maximilians-University, Munich, Germany
| | - Yvonne Döring
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Kiril Bidzhekov
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Tilman M Hackeng
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Christian Weber
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany.,Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands.,German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Hans-Joachim Gabius
- Faculty of Veterinary Medicine, Institute of Physiological Chemistry, Ludwig-Maximilians-University, Munich, Germany
| | - Philipp von Hundelshausen
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany.,German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany
| | - Kevin H Mayo
- Department of Biochemistry, Molecular Biology & Biophysics, Health Sciences Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
29
|
Festa LK, Irollo E, Platt BJ, Tian Y, Floresco S, Meucci O. CXCL12-induced rescue of cortical dendritic spines and cognitive flexibility. eLife 2020; 9:e49717. [PMID: 31971513 PMCID: PMC7007222 DOI: 10.7554/elife.49717] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 01/21/2020] [Indexed: 01/05/2023] Open
Abstract
Synaptodendritic pruning is a common cause of cognitive decline in neurological disorders, including HIV-associated neurocognitive disorders (HAND). HAND persists in treated patients as a result of chronic inflammation and low-level expression of viral proteins, though the mechanisms involved in synaptic damage are unclear. Here, we report that the chemokine CXCL12 recoups both cognitive performance and synaptodendritic health in a rodent model of HAND, which recapitulates the neuroinflammatory state of virally controlled individuals and the associated structural/functional deficiencies. CXCL12 preferentially regulates plastic thin spines on layer II/III pyramidal neurons of the medial prefrontal cortex via CXCR4-dependent stimulation of the Rac1/PAK actin polymerization pathway, leading to increased spine density and improved flexible behavior. Our studies unveil a critical role of CXCL12/CXCR4 signaling in spine dynamics and cognitive flexibility, suggesting that HAND - or other diseases driven by spine loss - may be reversible and upturned by targeting Rac1-dependent processes in cortical neurons.
Collapse
Affiliation(s)
- Lindsay K Festa
- Department of Pharmacology and PhysiologyDrexel University College of MedicinePhiladelphiaUnited States
- Center of Neuroimmunology and CNS Therapeutics, Institute of Molecular Medicine and Infectious DiseasesDrexel University College of MedicinePhiladelphiaUnited States
| | - Elena Irollo
- Department of Pharmacology and PhysiologyDrexel University College of MedicinePhiladelphiaUnited States
| | - Brian J Platt
- Department of Pharmacology and PhysiologyDrexel University College of MedicinePhiladelphiaUnited States
| | - Yuzen Tian
- Department of Pharmacology and PhysiologyDrexel University College of MedicinePhiladelphiaUnited States
| | - Stan Floresco
- Department of PsychologyUniversity of British ColumbiaVancouverCanada
| | - Olimpia Meucci
- Department of Pharmacology and PhysiologyDrexel University College of MedicinePhiladelphiaUnited States
- Center of Neuroimmunology and CNS Therapeutics, Institute of Molecular Medicine and Infectious DiseasesDrexel University College of MedicinePhiladelphiaUnited States
- Department of Microbiology and ImmunologyDrexel University College of MedicinePhiladelphiaUnited States
| |
Collapse
|
30
|
Festa LK, Irollo E, Platt BJ, Tian Y, Floresco S, Meucci O. CXCL12-induced rescue of cortical dendritic spines and cognitive flexibility. eLife 2020. [PMID: 31971513 DOI: 10.7554/elife.49717.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Synaptodendritic pruning is a common cause of cognitive decline in neurological disorders, including HIV-associated neurocognitive disorders (HAND). HAND persists in treated patients as a result of chronic inflammation and low-level expression of viral proteins, though the mechanisms involved in synaptic damage are unclear. Here, we report that the chemokine CXCL12 recoups both cognitive performance and synaptodendritic health in a rodent model of HAND, which recapitulates the neuroinflammatory state of virally controlled individuals and the associated structural/functional deficiencies. CXCL12 preferentially regulates plastic thin spines on layer II/III pyramidal neurons of the medial prefrontal cortex via CXCR4-dependent stimulation of the Rac1/PAK actin polymerization pathway, leading to increased spine density and improved flexible behavior. Our studies unveil a critical role of CXCL12/CXCR4 signaling in spine dynamics and cognitive flexibility, suggesting that HAND - or other diseases driven by spine loss - may be reversible and upturned by targeting Rac1-dependent processes in cortical neurons.
Collapse
Affiliation(s)
- Lindsay K Festa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, United States.,Center of Neuroimmunology and CNS Therapeutics, Institute of Molecular Medicine and Infectious Diseases, Drexel University College of Medicine, Philadelphia, United States
| | - Elena Irollo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, United States
| | - Brian J Platt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, United States
| | - Yuzen Tian
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, United States
| | - Stan Floresco
- Department of Psychology, University of British Columbia, Vancouver, Canada
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, United States.,Center of Neuroimmunology and CNS Therapeutics, Institute of Molecular Medicine and Infectious Diseases, Drexel University College of Medicine, Philadelphia, United States.,Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, United States
| |
Collapse
|
31
|
Zanetti G, Negro S, Megighian A, Mattarei A, Lista F, Fillo S, Rigoni M, Pirazzini M, Montecucco C. A CXCR4 receptor agonist strongly stimulates axonal regeneration after damage. Ann Clin Transl Neurol 2019; 6:2395-2402. [PMID: 31725979 PMCID: PMC6917312 DOI: 10.1002/acn3.50926] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/30/2019] [Accepted: 10/02/2019] [Indexed: 12/21/2022] Open
Abstract
Objective To test whether the signaling axis CXCL12α‐CXCR4 is activated upon crush/cut of the sciatic nerve and to test the activity of NUCC‐390, a new CXCR4 agonist, in promoting nerve recovery from damage. Methods The sciatic nerve was either crushed or cut. Expression and localization of CXCL12α and CXCR4 were evaluated by imaging with specific antibodies. Their functional involvement in nerve regeneration was determined by antibody‐neutralization of CXCL12α, and by the CXCR4 specific antagonist AMD3100, using as quantitative read‐out the compound muscle action potential (CMAP). NUCC‐390 activity on nerve regeneration was determined by imaging and CMAP recordings. Results CXCR4 is expressed at the injury site within the axonal compartment, whilst its ligand CXCL12α is expressed in Schwann cells. The CXCL12α‐CXCR4 axis is involved in the recovery of neurotransmission of the injured nerve. More importantly, the small molecule NUCC‐390 is a strong promoter of the functional and anatomical recovery of the nerve, by acting very similarly to CXCL12α. This pharmacological action is due to the capability of NUCC‐390 to foster elongation of motor neuron axons both in vitro and in vivo. Interpretation Imaging and electrophysiological data provide novel and compelling evidence that the CXCL12α‐CXCR4 axis is involved in sciatic nerve repair after crush/cut. This makes NUCC‐390 a strong candidate molecule to stimulate nerve repair by promoting axonal elongation. We propose this molecule to be tested in other models of neuronal damage, to lay the basis for clinical trials on the efficacy of NUCC‐390 in peripheral nerve repair in humans.
Collapse
Affiliation(s)
- Giulia Zanetti
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Samuele Negro
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Padua Neuroscience Center, University of Padua, Padua, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | | | - Silvia Fillo
- Scientific Department, Army Medical Center, Roma, Italy
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Marco Pirazzini
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,CNR Institute of Neuroscience, Padua, Italy
| |
Collapse
|
32
|
Negro S, Zanetti G, Mattarei A, Valentini A, Megighian A, Tombesi G, Zugno A, Dianin V, Pirazzini M, Fillo S, Lista F, Rigoni M, Montecucco C. An Agonist of the CXCR4 Receptor Strongly Promotes Regeneration of Degenerated Motor Axon Terminals. Cells 2019; 8:E1183. [PMID: 31575088 PMCID: PMC6829515 DOI: 10.3390/cells8101183] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/24/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022] Open
Abstract
The activation of the G-protein coupled receptor CXCR4 by its ligand CXCL12α is involved in a large variety of physiological and pathological processes, including the growth of B cells precursors and of motor axons, autoimmune diseases, stem cell migration, inflammation, and several neurodegenerative conditions. Recently, we demonstrated that CXCL12α potently stimulates the functional recovery of damaged neuromuscular junctions via interaction with CXCR4. This result prompted us to test the neuroregeneration activity of small molecules acting as CXCR4 agonists, endowed with better pharmacokinetics with respect to the natural ligand. We focused on NUCC-390, recently shown to activate CXCR4 in a cellular system. We designed a novel and convenient chemical synthesis of NUCC-390, which is reported here. NUCC-390 was tested for its capability to induce the regeneration of motor axon terminals completely degenerated by the presynaptic neurotoxin α-Latrotoxin. NUCC-390 was found to strongly promote the functional recovery of the neuromuscular junction, as assayed by electrophysiology and imaging. This action is CXCR4 dependent, as it is completely prevented by AMD3100, a well-characterized CXCR4 antagonist. These data make NUCC-390 a strong candidate to be tested in human therapy to promote nerve recovery of function after different forms of neurodegeneration.
Collapse
Affiliation(s)
- Samuele Negro
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
| | - Giulia Zanetti
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua 35131, Italy.
| | - Alice Valentini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua 35131, Italy.
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
- Padua Neuroscience Institute, Padua 35131, Italy.
| | - Giulia Tombesi
- Department of Biology, University of Padua, Padua 35131, Italy.
| | - Alessandro Zugno
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua 35131, Italy.
| | - Valentina Dianin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua 35131, Italy.
| | - Marco Pirazzini
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
| | - Silvia Fillo
- Center of Medical and Veterinary Research of the Ministry of Defence, Rome 00184, Italy.
| | - Florigio Lista
- Center of Medical and Veterinary Research of the Ministry of Defence, Rome 00184, Italy.
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
- CNR Institute of Neuroscience, Padua 35131, Italy.
| |
Collapse
|
33
|
Reyes AWB, Arayan LT, Huy TXN, Vu SH, Kang CK, Min W, Lee HJ, Lee JH, Kim S. Chemokine receptor 4 (CXCR4) blockade enhances resistance to bacterial internalization in RAW264.7 cells and AMD3100, a CXCR4 antagonist, attenuates susceptibility to Brucella abortus 544 infection in a murine model. Vet Microbiol 2019; 237:108402. [PMID: 31585647 DOI: 10.1016/j.vetmic.2019.108402] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 02/06/2023]
Abstract
We investigated the involvement of chemokine receptor type 4 (CXCR4) signaling on the outcome of Brucella (B.) abortus 544 infection in murine macrophages and in a mouse model. CXCR4 manipulation were first evaluated for Brucella invasion and intracellular survival efficiency, mitogen-activated protein kinases (ERK1/2, JNK, p38α) activation and generation of nitric oxide (NO), and then in the splenic bacterial proliferation and cytokine production in BALB/c mice. CXCR4 blockade is involved in the successful control of Brucella invasion, reduction of ERK1/2 phosphorylation and inhibition of nitric oxide release from macrophages. Furthermore, using a reported CXCR4-specific antagonist AMD3100 resulted in splenomegaly but attenuated Brucella proliferation in these organs with elevated serum levels of MCP-1, TNF and IL-12. These findings provide insights on the contribution of CXCR4 signaling in the phagocytic pathway and immune modulation during B. abortus infection.
Collapse
Affiliation(s)
| | - Lauren Togonon Arayan
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Tran Xuan Ngoc Huy
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Son Hai Vu
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Chang Keun Kang
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Wongi Min
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Hu Jang Lee
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - John Hwa Lee
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Suk Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
34
|
Mishra RK, Clutter MR, Blyth GT, Kosciuczuk EM, Blackburn AZ, Beauchamp EM, Schiltz GE, Platanias LC. Discovery of novel Mnk inhibitors using mutation-based induced-fit virtual high-throughput screening. Chem Biol Drug Des 2019; 94:1813-1823. [PMID: 31260185 DOI: 10.1111/cbdd.13585] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/17/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022]
Abstract
Mnk kinases (Mnk1 and 2) are downstream effectors of Map kinase pathways and regulate phosphorylation of eukaryotic initiation factor 4E. Engagement of the Mnk pathway is critical in acute myeloid leukemia (AML) leukemogenesis and Mnk inhibitors have potent antileukemic properties in vitro and in vivo, suggesting that targeting Mnk kinases may provide a novel approach for treating AML. Here, we report the development and application of a mutation-based induced-fit in silico screen to identify novel Mnk inhibitors. The Mnk1 structure was modeled by temporarily mutating an amino acid that obstructs the ATP-binding site in the Mnk1 crystal structure while carrying out docking simulations of known inhibitors. The hit compounds display activity in Mnk biochemical and cellular assays, including acute myeloid leukemia progenitors. This approach will enable further rational structure-based drug design of new Mnk inhibitors and potentially novel ways of therapeutically targeting this kinase.
Collapse
Affiliation(s)
- Rama K Mishra
- The Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, USA.,Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Matthew R Clutter
- Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Gavin T Blyth
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ewa M Kosciuczuk
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Division of Hematology-Oncology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Amy Z Blackburn
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elspeth M Beauchamp
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Division of Hematology-Oncology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Gary E Schiltz
- The Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, USA.,Department of Pharmacology, Northwestern University, Chicago, IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Leonidas C Platanias
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.,Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Division of Hematology-Oncology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
35
|
Iyamu ID, Lv W, Malik N, Mishra RK, Schiltz GE. Development of Tetrahydroindazole-Based Potent and Selective Sigma-2 Receptor Ligands. ChemMedChem 2019; 14:1248-1256. [PMID: 31071238 PMCID: PMC6613831 DOI: 10.1002/cmdc.201900203] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/08/2019] [Indexed: 11/08/2022]
Abstract
The sigma-2 receptor has been shown to play important roles in a number of important diseases, including central nervous system (CNS) disorders and cancer. However, mechanisms by which sigma-2 contributes to these diseases remain unclear. The development of new sigma-2 ligands that can be used to probe the function of this protein and potentially as drug discovery leads is therefore of great importance. Herein we report the development of a series of tetrahydroindazole compounds that are highly potent and selective for sigma-2. Structure-activity relationship data were used to generate a pharmacophore model that summarizes the common features present in the potent ligands. Assays for solubility and microsomal stability showed that several members of this compound series possess promising characteristics for further development of useful chemical probes or drug discovery leads.
Collapse
Affiliation(s)
- Iredia D Iyamu
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, 60208, USA
| | - Wei Lv
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, 60208, USA
| | - Neha Malik
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, 60208, USA
| | - Rama K Mishra
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, 60208, USA
- Department of Pharmacology, Northwestern University, Chicago, IL, 60611, USA
| | - Gary E Schiltz
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, 60208, USA
- Department of Pharmacology, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| |
Collapse
|
36
|
Arimont M, Hoffmann C, de Graaf C, Leurs R. Chemokine Receptor Crystal Structures: What Can Be Learned from Them? Mol Pharmacol 2019; 96:765-777. [PMID: 31266800 DOI: 10.1124/mol.119.117168] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/21/2019] [Indexed: 12/18/2022] Open
Abstract
Chemokine receptors belong to the class A of G protein-coupled receptors (GPCRs) and are implicated in a wide variety of physiologic functions, mostly related to the homeostasis of the immune system. Chemokine receptors are also involved in multiple pathologic processes, including immune and autoimmune diseases, as well as cancer. Hence, several members of this GPCR subfamily are considered to be very relevant therapeutic targets. Since drug discovery efforts can be significantly reinforced by the availability of crystal structures, substantial efforts in the area of chemokine receptor structural biology could dramatically increase the outcome of drug discovery campaigns. This short review summarizes the available data on chemokine receptor crystal structures, discusses the numerous applications from chemokine receptor structures that can enhance the daily work of molecular pharmacologists, and describes the challenges and pitfalls to consider when relying on crystal structures for further research applications. SIGNIFICANCE STATEMENT: This short review summarizes the available data on chemokine receptor crystal structures, discusses the numerous applications from chemokine receptor structures that can enhance the daily work of molecular pharmacologists, and describes the challenges and pitfalls to consider when relying on crystal structures for further research applications.
Collapse
Affiliation(s)
- Marta Arimont
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., R.L.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University, Jena, Germany (C.H.); and Sosei Heptares, Great Abington, Cambridge, United Kingdom (C.d.G.)
| | - Carsten Hoffmann
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., R.L.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University, Jena, Germany (C.H.); and Sosei Heptares, Great Abington, Cambridge, United Kingdom (C.d.G.)
| | - Chris de Graaf
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., R.L.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University, Jena, Germany (C.H.); and Sosei Heptares, Great Abington, Cambridge, United Kingdom (C.d.G.)
| | - Rob Leurs
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., R.L.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University, Jena, Germany (C.H.); and Sosei Heptares, Great Abington, Cambridge, United Kingdom (C.d.G.)
| |
Collapse
|
37
|
Smith N, Rodero MP, Bekaddour N, Bondet V, Ruiz-Blanco YB, Harms M, Mayer B, Bader-Meunier B, Quartier P, Bodemer C, Baudouin V, Dieudonné Y, Kirchhoff F, Sanchez Garcia E, Charbit B, Leboulanger N, Jahrsdörfer B, Richard Y, Korganow AS, Münch J, Nisole S, Duffy D, Herbeuval JP. Control of TLR7-mediated type I IFN signaling in pDCs through CXCR4 engagement-A new target for lupus treatment. SCIENCE ADVANCES 2019; 5:eaav9019. [PMID: 31309143 PMCID: PMC6620093 DOI: 10.1126/sciadv.aav9019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 06/06/2019] [Indexed: 06/10/2023]
Abstract
Type I interferons are highly potent cytokines essential for self-protection against tumors and infections. Deregulations of type I interferon signaling are associated with multiple diseases that require novel therapeutic options. Here, we identified the small molecule, IT1t, a previously described CXCR4 ligand, as a highly potent inhibitor of Toll-like receptor 7 (TLR7)-mediated inflammation. IT1t inhibits chemical (R848) and natural (HIV) TLR7-mediated inflammation in purified human plasmacytoid dendritic cells from blood and human tonsils. In a TLR7-dependent lupus-like model, in vivo treatment of mice with IT1t drives drastic reduction of both systemic inflammation and anti-double-stranded DNA autoantibodies and prevents glomerulonephritis. Furthermore, IT1t controls inflammation, including interferon α secretion, in resting and stimulated cells from patients with systemic lupus erythematosus. Our findings highlight a groundbreaking immunoregulatory property of CXCR4 signaling that opens new therapeutic perspectives in inflammatory settings and autoimmune diseases.
Collapse
Affiliation(s)
- Nikaïa Smith
- CNRS UMR-8601, CICB, 45 rue des Saints-Pères, 75006 Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy, CBMIT, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Mathieu P. Rodero
- CNRS UMR-8601, CICB, 45 rue des Saints-Pères, 75006 Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy, CBMIT, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nassima Bekaddour
- CNRS UMR-8601, CICB, 45 rue des Saints-Pères, 75006 Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy, CBMIT, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Vincent Bondet
- Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Yasser B. Ruiz-Blanco
- Computational Biochemistry and Center of Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Mirja Harms
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Benjamin Mayer
- Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Brigitte Bader-Meunier
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Imagine Institute, Paris, France
- Paediatric Haematology-Immunology and Rheumatology Department, Hôpital Universitaire Necker, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM UMR 1163, Laboratory of Immunogenetics of Paediatric Autoimmunity, Paris, France
| | - Pierre Quartier
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Imagine Institute, Paris, France
- Paediatric Haematology-Immunology and Rheumatology Department, Hôpital Universitaire Necker, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Christine Bodemer
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Imagine Institute, Paris, France
- Department of Paediatric Dermatology, Reference Centre for Rare Skin Disorders (MAGEC), Hôpital Universitaire Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Véronique Baudouin
- Hôpital Universitaire Robert Debré, Néphrologie pédiatrique, Paris, France
| | - Yannick Dieudonné
- CNRS UPR 3572 “Immunopathology and Therapeutic Chemistry”/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Rare Autoimmune Diseases, University Hospital, Strasbourg, France
- UFR Medicine, University of Strasbourg, Strasbourg, France
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Elsa Sanchez Garcia
- Computational Biochemistry and Center of Medical Biotechnology, University of Duisburg-Essen, 45141 Essen, Germany
| | - Bruno Charbit
- Centre for Translational Research, Institut Pasteur, Paris, France
| | - Nicolas Leboulanger
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Pediatric Otolaryngology Department, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Bernd Jahrsdörfer
- Institute of Transfusion Medicine and Immunogenetics (IKT) Ulm, Helmholtzstr. 10, 89081 Ulm, Germany
| | - Yolande Richard
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- INSERM U1016, Institut Cochin, Paris, France
- CNRS UMR 8104, Paris, France
| | - Anne-Sophie Korganow
- CNRS UPR 3572 “Immunopathology and Therapeutic Chemistry”/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Rare Autoimmune Diseases, University Hospital, Strasbourg, France
- UFR Medicine, University of Strasbourg, Strasbourg, France
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Sébastien Nisole
- IRIM, Université de Montpellier, CNRS UMR, 9004 Montpellier, France
| | - Darragh Duffy
- Immunobiology of Dendritic Cells, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
- Centre for Translational Research, Institut Pasteur, Paris, France
| | - Jean-Philippe Herbeuval
- CNRS UMR-8601, CICB, 45 rue des Saints-Pères, 75006 Paris, France
- Team Chemistry & Biology, Modeling & Immunology for Therapy, CBMIT, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
38
|
Diao PC, Hu MJ, Yang HK, You WW, Zhao PL. Facile one-pot synthesis, antiproliferative evaluation and structure-activity relationships of 3-amino-1H-indoles and 3-amino-1H-7-azaindoles. Bioorg Chem 2019; 88:102914. [DOI: 10.1016/j.bioorg.2019.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/26/2019] [Accepted: 04/06/2019] [Indexed: 12/31/2022]
|
39
|
Iyamu ID, Lv W, Malik N, Mishra RK, Schiltz GE. Discovery of a novel class of potent and selective tetrahydroindazole-based sigma-1 receptor ligands. Bioorg Med Chem 2019; 27:1824-1835. [PMID: 30904383 PMCID: PMC6548570 DOI: 10.1016/j.bmc.2019.03.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 03/14/2019] [Accepted: 03/16/2019] [Indexed: 11/20/2022]
Abstract
The sigma-1 and sigma-2 receptors have been shown to play important roles in CNS diseases, cancer, and other disorders. These findings suggest that targeting these proteins with small-molecule modulators may be of important therapeutic value. Here we report the development of a new class of tetrahydroindazoles that are highly potent and selective ligands for sigma-1. Molecular modeling was used to rationalize the observed structure-activity relationships and identify key interactions responsible for increased potency of the optimized compounds. Assays for solubility and microsomal stability showed this series possesses favorable characteristics and is amenable to further therapeutic development. The compounds described herein will be useful in the development of new chemical probes for sigma-1 and to aid in future work therapeutically targeting this protein.
Collapse
Affiliation(s)
- Iredia D Iyamu
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL 60208, United States
| | - Wei Lv
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL 60208, United States
| | - Neha Malik
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL 60208, United States
| | - Rama K Mishra
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL 60208, United States; Department of Pharmacology, Northwestern University, Chicago, IL 60611, United States
| | - Gary E Schiltz
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL 60208, United States; Department of Pharmacology, Northwestern University, Chicago, IL 60611, United States; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States.
| |
Collapse
|
40
|
De Clercq E. Mozobil® (Plerixafor, AMD3100), 10 years after its approval by the US Food and Drug Administration. Antivir Chem Chemother 2019; 27:2040206619829382. [PMID: 30776910 PMCID: PMC6379795 DOI: 10.1177/2040206619829382] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AMD3100 (plerixafor, Mozobil®) was first identified as an anti-HIV agent
specifically active against the T4-lymphotropic HIV strains, as it selectively
blocked the CXCR4 receptor. Through interference with the interaction of CXCR4
with its natural ligand, SDF-1 (also named CXCL12), it also mobilized the
CD34+stem cells from the bone marrow into the peripheral blood
stream. In December 2008, AMD3100 was formally approved by the US FDA for
autologous transplantation in patients with Non-Hodgkin’s Lymphoma or multiple
myeloma. It may be beneficially used in various other malignant diseases as well
as hereditary immunological disorders such as WHIM syndrome, and
physiopathological processes such as hepatopulmonary syndrome.
Collapse
|
41
|
Kamali A, Oryan A, Hosseini S, Ghanian MH, Alizadeh M, Baghaban Eslaminejad M, Baharvand H. Cannabidiol-loaded microspheres incorporated into osteoconductive scaffold enhance mesenchymal stem cell recruitment and regeneration of critical-sized bone defects. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 101:64-75. [PMID: 31029357 DOI: 10.1016/j.msec.2019.03.070] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/05/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023]
Abstract
Recruitment of mesenchymal stem cells (MSCs) to an injury site and their differentiation into the desired cell lineage are implicated in deficient bone regeneration. To date, there is no ideal structure that provides these conditions for bone regeneration. In the current study, we aim to develop a novel scaffold that induces MSC migration towards the defect site, followed by their differentiation into an osteogenic lineage. We have fabricated a gelatin/nano-hydroxyapatite (G/nHAp) scaffold that delivered cannabidiol (CBD)-loaded poly (lactic-co-glycolic acid) (PLGA) microspheres to critical size radial bone defects in a rat model. The fabricated scaffolds were characterized by X-ray diffraction (XRD) and scanning electron microscopy (SEM), and then analyzed for porosity and degradation rate. The release profile of CBD from the PLGA microsphere and CBD-PLGA-G/nHAp scaffold was analyzed by fluorescence spectroscopy. We performed an in vitro assessment of the effects of CBD on cellular behaviors of viability and osteogenic differentiation. Radiological evaluation, histomorphometry, and immunohistochemistry (IHC) analysis of all defects in the scaffold and control groups were conducted following transplantation into the radial bone defects. An in vitro migration assay showed that CBD considerably increased MSCs migration. qRT-PCR results showed upregulated expression of osteogenic markers in the presence of CBD. Histological and immunohistochemical findings confirmed new bone formation and reconstruction of the defect at 4 and 12 week post-surgery (WPS) in the CBD-PLGA-G/nHAp group. Immunofluorescent analysis revealed enhanced migration of MSCs into the defect areas in the CBD-PLGA-G/nHAp group in vivo. Based on the results of the current study, we concluded that CBD improved bone healing and showed a critical role for MSC migration in the bone regeneration process.
Collapse
Affiliation(s)
- Amir Kamali
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ahmad Oryan
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Samaneh Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Alizadeh
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
42
|
Izquierdo J, Jain AD, Abdulkadir SA, Schiltz GE. Palladium-catalyzed coupling reactions on functionalized 2-trifluoromethyl-4-chromenone scaffolds. Synthesis of highly functionalized trifluoromethyl-heterocycles. SYNTHESIS-STUTTGART 2019; 51:1342-1352. [PMID: 31274934 PMCID: PMC6605783 DOI: 10.1055/s-0037-1610669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The chromenone core is a ubiquitous group in biologically-active natural products and has been extensively used in organic synthesis. Fluorine derived compounds, including those with a trifluoromethyl group (-CF3), have shown enhanced biological activities in numerous pharmaceuticals compared with their non-fluorinated analogs. We have found that 2-trifluoromethyl chromenones can be readily functionalized in the 8- and 7-positions, providing chromenones cores of high structural complexity which are excellent precursors for numerous trifluoromethyl-heterocycles.
Collapse
Affiliation(s)
- Javier Izquierdo
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois, USA
| | - Atul D Jain
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois, USA
| | - Sarki A Abdulkadir
- Department of Urology, Northwestern University, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Gary E Schiltz
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Department of Pharmacology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
43
|
Therapeutic peptides: Historical perspectives, current development trends, and future directions. Bioorg Med Chem 2018; 26:2700-2707. [DOI: 10.1016/j.bmc.2017.06.052] [Citation(s) in RCA: 957] [Impact Index Per Article: 159.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/13/2017] [Accepted: 06/30/2017] [Indexed: 01/04/2023]
|
44
|
Berg C, Daugvilaite V, Steen A, Jørgensen AS, Våbenø J, Rosenkilde MM. Inhibition of HIV Fusion by Small Molecule Agonists through Efficacy-Engineering of CXCR4. ACS Chem Biol 2018; 13:881-886. [PMID: 29461034 DOI: 10.1021/acschembio.8b00061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
CXC chemokine receptor 4 (CXCR4) is involved in multiple physiological and pathological processes, notably as a coreceptor for human immunodeficiency virus (HIV) cell entry. Its broad expression pattern and vital biological importance make CXCR4 a troublesome drug target, as disruption of the interaction with its endogenous ligand, CXC chemokine ligand 12 (CXCL12), has severe consequences. In fact, only one CXCR4 drug, the bicyclam antagonist and HIV entry inhibitor AMD3100 (Plerixafor/Mozobil), has been approved for clinical use, however only for stem cell mobilization-a consequence of CXCR4 antagonism. Here, we report the engineering of an efficacy switch mutation in CXCR4-F292A7.43 in the middle of transmembrane helix 7-that converted the antagonists AMD3100 and AMD11070 into partial agonists. As agonists on F292A CXCR4, AMD3100 and AMD11070 were less disruptive to CXCR4 signaling while they remained efficient inhibitors of HIV fusion. This demonstrates that small molecule CXCR4 agonists can have a therapeutic potential as HIV entry inhibitors.
Collapse
Affiliation(s)
- Christian Berg
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
- Infectious Diseases Unit, Department of Medicine, Herlev−Gentofte Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Viktorija Daugvilaite
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Anne Steen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Astrid Sissel Jørgensen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Jon Våbenø
- Helgeland Hospital Trust, Prestmarkveien 1, 8800 Sandnessjøen, Norway
| | - Mette Marie Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| |
Collapse
|
45
|
Wu CH, Song JS, Kuan HH, Wu SH, Chou MC, Jan JJ, Tsou LK, Ke YY, Chen CT, Yeh KC, Wang SY, Yeh TK, Tseng CT, Huang CL, Wu MH, Kuo PC, Lee CJ, Shia KS. Development of Stem-Cell-Mobilizing Agents Targeting CXCR4 Receptor for Peripheral Blood Stem Cell Transplantation and Beyond. J Med Chem 2018; 61:818-833. [PMID: 29314840 DOI: 10.1021/acs.jmedchem.7b01322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The function of the CXCR4/CXCL12 axis accounts for many disease indications, including tissue/nerve regeneration, cancer metastasis, and inflammation. Blocking CXCR4 signaling with its antagonists may lead to moving out CXCR4+ cell types from bone marrow to peripheral circulation. We have discovered a novel series of pyrimidine-based CXCR4 antagonists, a representative (i.e., 16) of which was tolerated at a higher dose and showed better HSC-mobilizing ability at the maximal response dose relative to the approved drug 1 (AMD3100), and thus considered a potential drug candidate for PBSCT indication. Docking compound 16 into the X-ray crystal structure of CXCR4 receptor revealed that it adopted a spider-like conformation striding over both major and minor subpockets. This putative binding mode provides a new insight into CXCR4 receptor-ligand interactions for further structural modifications.
Collapse
Affiliation(s)
- Chien-Huang Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Jen-Shin Song
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Hsuan-Hao Kuan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Szu-Huei Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Ming-Chen Chou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Jiing-Jyh Jan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Lun K Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Yi-Yu Ke
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Kai-Chia Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Sing-Yi Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Chen-Tso Tseng
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Chen-Lung Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Mine-Hsine Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Po-Chu Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Chia-Jui Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| | - Kak-Shan Shia
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli County 35053, Taiwan, R.O.C
| |
Collapse
|
46
|
Signaling properties of the human chemokine receptors CXCR4 and CXCR7 by cellular electric impedance measurements. PLoS One 2017; 12:e0185354. [PMID: 28945785 PMCID: PMC5612718 DOI: 10.1371/journal.pone.0185354] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 09/11/2017] [Indexed: 01/05/2023] Open
Abstract
The chemokine receptor 4 (CXCR4) and 7 (CXCR7) are G-protein-coupled receptors involved in various diseases including human cancer. As such, they have become important targets for therapeutic intervention. Cell-based receptor assays, able to detect agents that modulate receptor activity, are of key importance for drug discovery. We evaluated the potential of cellular electric impedance for this purpose. Dose-dependent and specific stimulation of CXCR4 was detected upon addition of its unique chemokine ligand CXCL12. The response magnitude correlated with the CXCR4 expression level. Gαi coupling and signaling contributed extensively to the impedance response, whereas Gαq- and Gβγ-related events had only minor effects on the impedance profile. CXCR7 signaling could not be detected using impedance measurements. However, increasing levels of CXCR7 expression significantly reduced the CXCR4-mediated impedance readout, suggesting a regulatory role for CXCR7 on CXCR4-mediated signaling. Taken together, cellular electric impedance spectroscopy can represent a valuable alternative pharmacological cell-based assay for the identification of molecules targeting CXCR4, but not for CXCR7 in the absence of CXCR4.
Collapse
|
47
|
Structure and Function of Peptide-Binding G Protein-Coupled Receptors. J Mol Biol 2017; 429:2726-2745. [PMID: 28705763 DOI: 10.1016/j.jmb.2017.06.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors and are important human drug targets. Of the 826 human GPCRs, 118 of them recognize endogenous peptide or protein ligands, and 30 of the 118 are targeted by approved drug molecules, including the very high-profile class B glucagon-like peptide 1 receptor. In this review, we analyze the 21 experimentally determined three-dimensional structures of the known peptide-binding GPCRs in relation to the endogenous peptides and drug molecules that modulate their cell signaling processes. Our integrated analyses reveal that half of the marketed drugs and most of the drugs in clinical trials that interact with peptide GPCRs are small molecules with a wide range of binding modes distinct from those of large peptide ligands. As we continue to collect additional data on these receptors from orthogonal approaches, including nuclear magnetic resonance and electron microscopy, we are beginning to understand how these receptors interact with their ligands at the molecular level and how improving the pharmacology of GPCR signal transduction requires us to study these receptors using multiple biophysical techniques.
Collapse
|
48
|
Arimont M, Sun SL, Leurs R, Smit M, de Esch IJP, de Graaf C. Structural Analysis of Chemokine Receptor-Ligand Interactions. J Med Chem 2017; 60:4735-4779. [PMID: 28165741 PMCID: PMC5483895 DOI: 10.1021/acs.jmedchem.6b01309] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
This
review focuses on the construction and application of structural chemokine
receptor models for the elucidation of molecular determinants of chemokine
receptor modulation and the structure-based discovery and design of
chemokine receptor ligands. A comparative analysis of ligand binding
pockets in chemokine receptors is presented, including a detailed
description of the CXCR4, CCR2, CCR5, CCR9, and US28 X-ray structures,
and their implication for modeling molecular interactions of chemokine
receptors with small-molecule ligands, peptide ligands, and large
antibodies and chemokines. These studies demonstrate how the integration
of new structural information on chemokine receptors with extensive
structure–activity relationship and site-directed mutagenesis
data facilitates the prediction of the structure of chemokine receptor–ligand
complexes that have not been crystallized. Finally, a review of structure-based
ligand discovery and design studies based on chemokine receptor crystal
structures and homology models illustrates the possibilities and challenges
to find novel ligands for chemokine receptors.
Collapse
Affiliation(s)
- Marta Arimont
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Shan-Liang Sun
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Rob Leurs
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Martine Smit
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Chris de Graaf
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|