1
|
Syed M, Dishman AF, Volkman BF, Walker TL. The multifaceted role of XCL1 in health and disease. Protein Sci 2025; 34:e70032. [PMID: 39840812 PMCID: PMC11751857 DOI: 10.1002/pro.70032] [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: 09/26/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 01/23/2025]
Abstract
The chemokine XC motif chemokine ligand 1 (XCL1) is an unusually specialized member of a conserved family of around 50 small, secreted proteins that are best known for their ability to stimulate the directional migration of cells. All chemokines adopt a very similar folded structure that binds a specific G protein-coupled receptor (GPCR), and most chemokines bind extracellular matrix glycosaminoglycans, often in a dimeric or oligomeric form. Owing in part to the lack of a disulfide bond that is conserved in all other chemokines, XCL1 interconverts between two distinct structures with distinct functions. One XCL1 fold resembles the structure of all other chemokines (chemokine fold), while the other does not (alternate fold). The chemokine fold of XCL1 displays high affinity for the GPCR XCR1, while the alternative fold binds GAGs and exhibits antimicrobial activity. Although the canonical role of XCL1 as a CD8+ dendritic cell chemoattractant was defined more than a decade ago, the misconception that XCL1 is a lymphocyte-specific chemoattractant still prevails in the recent literature. This review aims to highlight the structure-guided functions of XCL1 and reclarify its immunological role. In addition, the implications of this metamorphic chemokine in vaccine development and emerging functions in the nervous system will be explored.
Collapse
Affiliation(s)
- Muhammed Syed
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQueenslandAustralia
| | - Acacia F. Dishman
- Department of BiochemistryMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Brian F. Volkman
- Department of BiochemistryMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Tara L. Walker
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain InstituteThe University of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
2
|
Zhang Y, Li X, Peng P, Qiu Z, Di C, Chen X, Wang N, Chen F, He Y, Liu Z, Zhao F, Zhu D, Dong S, Hu S, Yang Z, Li Y, Guo Y, Yang T. RUNX2 Phase Separation Mediates Long-Range Regulation Between Osteoporosis-Susceptibility Variant and XCR1 to Promote Osteoblast Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413561. [PMID: 39704037 PMCID: PMC11809430 DOI: 10.1002/advs.202413561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Indexed: 12/21/2024]
Abstract
GWASs have identified many loci associated with osteoporosis, but the underlying genetic regulatory mechanisms and the potential drug target need to be explored. Here, a new regulatory mechanism is found that a GWAS intergenic SNP (rs4683184) functions as an enhancer to influence the binding affinity of transcription factor RUNX2, whose phase separation can mediate the long-range chromatin interaction between enhancer and target gene XCR1 (a member of the GPCR family), leading to changes of XCR1 expression and osteoblast differentiation. Bone-targeting AAV of Xcr1 can improve bone formation in osteoporosis mice, suggesting that XCR1 can be a new susceptibility gene for osteoporosis. This study is the first to link non-coding SNP with phase separation, providing a new insight into long-range chromatin regulation mechanisms with susceptibility to complex diseases, and finding a potential target for the development of osteoporosis drugs and corresponding translational research.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Xin‐Hao Li
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Pai Peng
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Zi‐Han Qiu
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Chen‐Xi Di
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Xiao‐Feng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Nai‐Ning Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| | - Fei Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Yin‐Wei He
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Zhong‐Bo Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Fan Zhao
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Dong‐Li Zhu
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Shan‐Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Shou‐Ye Hu
- Department of Joint SurgeryHonghui HospitalXi'an Jiaotong UniversityXi'anShaanxi710054China
| | - Zhi Yang
- Department of Joint SurgeryHonghui HospitalXi'an Jiaotong UniversityXi'anShaanxi710054China
| | - Yi‐Ping Li
- Division in Cellular and Molecular MedicineDepartment of Pathology and Laboratory MedicineTulane University School of MedicineTulane UniversityNew OrleansLA70112USA
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
| | - Tie‐Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of EducationKey Laboratory of Biology Multiomics and Diseases in Shaanxi Province Higher Education Institutionsand Biomedical Informatics & Genomics CenterSchool of Life Science and TechnologyXi'an Jiaotong UniversityXi'anShaanxi710049China
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi710061China
| |
Collapse
|
3
|
Zhang X, Schlimgen RR, Singh S, Tomani MP, Volkman BF, Zhang C. Molecular basis for chemokine recognition and activation of XCR1. Proc Natl Acad Sci U S A 2024; 121:e2405732121. [PMID: 39565315 PMCID: PMC11621518 DOI: 10.1073/pnas.2405732121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 10/15/2024] [Indexed: 11/21/2024] Open
Abstract
The X-C motif chemokine receptor XCR1, which selectively binds to the chemokine XCL1, is highly expressed in conventional dendritic cells subtype 1 (cDC1s) and crucial for their activation. Modulating XCR1 signaling in cDC1s could offer novel opportunities in cancer immunotherapy and vaccine development by enhancing the antigen presentation function of cDC1s. To investigate the molecular mechanism of XCL-induced XCR1 signaling, we determined a high-resolution structure of the human XCR1 and Gi complex with an engineered form of XCL1, XCL1 CC3, by cryoelectron microscopy. Through mutagenesis and structural analysis, we elucidated the molecular details for the binding of the N-terminal segment of XCL1 CC3, which is vital for activating XCR1. The unique arrangement within the XCL1 CC3 binding site confers specificity for XCL1 in XCR1. We propose an activation mechanism for XCR1 involving structural alterations of key residues at the bottom of the XCL1 binding pocket. These detailed insights into XCL1 CC3-XCR1 interaction and XCR1 activation pave the way for developing novel XCR1-targeted therapeutics.
Collapse
MESH Headings
- Humans
- Chemokines, C/metabolism
- Chemokines, C/genetics
- Chemokines, C/chemistry
- Cryoelectron Microscopy
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/chemistry
- Receptors, G-Protein-Coupled/genetics
- Binding Sites
- Protein Binding
- Signal Transduction
- Models, Molecular
- Dendritic Cells/metabolism
- Dendritic Cells/immunology
- Receptors, Chemokine/metabolism
- Receptors, Chemokine/chemistry
- Receptors, Chemokine/genetics
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
| | - Roman R. Schlimgen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI53226
| | - Stephanie Singh
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
| | - Michael P. Tomani
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
| | - Brian F. Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI53226
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI53226
| | - Cheng Zhang
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA15261
| |
Collapse
|
4
|
Singh P, Taborsky M, Peichel CL, Sturmbauer C. Genomic basis of Y-linked dwarfism in cichlids pursuing alternative reproductive tactics. Mol Ecol 2023; 32:1592-1607. [PMID: 36588349 DOI: 10.1111/mec.16839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 01/03/2023]
Abstract
Sexually antagonistic selection, which favours different optima in males and females, is predicted to play an important role in the evolution of sex chromosomes. Body size is a sexually antagonistic trait in the shell-brooding cichlid fish Lamprologous callipterus, as "bourgeois" males must be large enough to carry empty snail shells to build nests whereas females must be small enough to fit into shells for breeding. In this species, there is also a second male morph: smaller "dwarf" males employ an alternative reproductive strategy by wriggling past spawning females into shells to fertilize eggs. L. callipterus male morphology is passed strictly from father to son, suggesting Y-linkage. However, sex chromosomes had not been previously identified in this species, and the genomic basis of size dimorphism was unknown. Here we used whole-genome sequencing to identify a 2.4-Mb sex-linked region on scaffold_23 with reduced coverage and single nucleotide polymorphism density in both male morphs compared to females. Within this sex region, distinct Y-haplotypes delineate the two male morphs, and candidate genes for body size (GHRHR, a known dwarfism gene) and sex determination (ADCYAP1R1) are in high linkage disequilibrium. Because differences in body size between females and males are under strong selection in L. callipterus, we hypothesize that sexual antagonism over body size initiated early events in sex chromosome evolution, followed by Y divergence to give rise to bourgeois and dwarf male reproductive strategies. Our results are consistent with the hypothesis that sexually antagonistic traits should be linked to young sex chromosomes.
Collapse
Affiliation(s)
- Pooja Singh
- Institute of Biology, University of Graz, Graz, Austria
- Aquatic Ecology Division, Institute of Ecology and Evolution, University of Bern, Bern, Switzerland
- Swiss Federal Institute of Aquatic Science and Technology (EAWAG), Kastanienbaum, Switzerland
| | - Michael Taborsky
- Behavioural Ecology Division, Institute of Ecology and Evolution, University of Bern, Switzerland
- Max Planck Institute of Animal Behavior, Constance, Germany
- Institute for Advanced Study (Wissenschaftskolleg) Berlin, Berlin, Germany
| | - Catherine L Peichel
- Evolutionary Ecology Division, Institute of Ecology and Evolution, University of Bern, Bern, Switzerland
| | | |
Collapse
|
5
|
Identification of circRNA expression profiles and the potential role of hsa_circ_0006916 in silicosis and pulmonary fibrosis. Toxicology 2023; 483:153384. [PMID: 36403901 DOI: 10.1016/j.tox.2022.153384] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/21/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
Circular RNAs (circRNAs) are emerging as novel regulators in the biological development of various diseases, but their expression profiles, functions and mechanisms in silicosis and pulmonary fibrosis remain largely unexplored. In this study, we constructed a mouse model of pulmonary fibrosis by intratracheal injection of silica particles and then performed transcriptome RNA sequencing of lung tissues. The results showed that 78 circRNAs, 39 miRNAs and 262 mRNAs were differentially expressed. Among them, five circRNAs, three miRNAs and four mRNAs were further selected, and their abnormal expression was verified in mouse fibrotic lung tissues by RT-qPCR assay. The circRNA-associated ceRNA network including 206 ceRNA triplets was constructed based on abnormally expressed circRNAs, miRNAs and mRNAs, and miR-199b-5p, miR-296-5p and miR-708-5p were identified as hub miRNAs connected to circRNAs and mRNAs. Subsequently, GO and KEGG pathway enrichment analyses were performed to detect the potential roles of differentially expressed mRNAs in pulmonary fibrosis, which were mainly involved in immune response, Th17 cell differentiation, NF-κB signaling pathway and PI3K-Akt signaling pathway. Furthermore, we identified that hsa_circ_0006916 was up-regulated in pulmonary fibrosis. To characterize the potential role of hsa_circ_0006916, we transfected siRNA targeting hsa_circ_0006916 into alveolar macrophages and found that knockdown of hsa_circ_0006916 significantly increased the expression levels of M1 molecules IL-1β and TNF-α and reduced the expression level of M2 molecule TGF-β1, indicating that hsa_circ_0006916 may play an important role in the activation of M1-M2 polarization effect in macrophages. Our results provided important evidence on the possible contribution of these abnormal circRNAs to the development of silicosis and pulmonary fibrosis.
Collapse
|
6
|
Drouillard D, Craig BT, Dwinell MB. Physiology of chemokines in the cancer microenvironment. Am J Physiol Cell Physiol 2023; 324:C167-C182. [PMID: 36317799 PMCID: PMC9829481 DOI: 10.1152/ajpcell.00151.2022] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 01/07/2023]
Abstract
Chemokines are chemotactic cytokines whose canonical functions govern movement of receptor-expressing cells along chemical gradients. Chemokines are a physiological system that is finely tuned by ligand and receptor expression, ligand or receptor oligomerization, redundancy, expression of atypical receptors, and non-GPCR binding partners that cumulatively influence discrete pharmacological signaling responses and cellular functions. In cancer, chemokines play paradoxical roles in both the directed emigration of metastatic, receptor-expressing cancer cells out of the tumor as well as immigration of tumor-infiltrating immune cells that culminate in a tumor-unique immune microenvironment. In the age of precision oncology, strategies to effectively harness the power of immunotherapy requires consideration of chemokine gradients within the unique spatial topography and temporal influences with heterogeneous tumors. In this article, we review current literature on the diversity of chemokine ligands and their cellular receptors that detect and process chemotactic gradients and illustrate how differences between ligand recognition and receptor activation influence the signaling machinery that drives cellular movement into and out of the tumor microenvironment. Facets of chemokine physiology across discrete cancer immune phenotypes are contrasted to existing chemokine-centered therapies in cancer.
Collapse
Affiliation(s)
- Donovan Drouillard
- Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Brian T Craig
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael B Dwinell
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Center for Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
7
|
Xi K, Zhu L. Automated Path Searching Reveals the Mechanism of Hydrolysis Enhancement by T4 Lysozyme Mutants. Int J Mol Sci 2022; 23:ijms232314628. [PMID: 36498954 PMCID: PMC9736071 DOI: 10.3390/ijms232314628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
Bacteriophage T4 lysozyme (T4L) is a glycosidase that is widely applied as a natural antimicrobial agent in the food industry. Due to its wide applications and small size, T4L has been regarded as a model system for understanding protein dynamics and for large-scale protein engineering. Through structural insights from the single conformation of T4L, a series of mutations (L99A,G113A,R119P) have been introduced, which have successfully raised the fractional population of its only hydrolysis-competent excited state to 96%. However, the actual impact of these substitutions on its dynamics remains unclear, largely due to the lack of highly efficient sampling algorithms. Here, using our recently developed travelling-salesman-based automated path searching (TAPS), we located the minimum-free-energy path (MFEP) for the transition of three T4L mutants from their ground states to their excited states. All three mutants share a three-step transition: the flipping of F114, the rearrangement of α0/α1 helices, and final refinement. Remarkably, the MFEP revealed that the effects of the mutations are drastically beyond the expectations of their original design: (a) the G113A substitution not only enhances helicity but also fills the hydrophobic Cavity I and reduces the free energy barrier for flipping F114; (b) R119P barely changes the stability of the ground state but stabilizes the excited state through rarely reported polar contacts S117OG:N132ND2, E11OE1:R145NH1, and E11OE2:Q105NE2; (c) the residue W138 flips into Cavity I and further stabilizes the excited state for the triple mutant L99A,G113A,R119P. These novel insights that were unexpected in the original mutant design indicated the necessity of incorporating path searching into the workflow of rational protein engineering.
Collapse
|
8
|
Dragan P, Atzei A, Sanmukh SG, Latek D. Computational and experimental approaches to probe GPCR activation and signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 193:1-36. [PMID: 36357073 DOI: 10.1016/bs.pmbts.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
G protein-coupled receptors (GPCRs) regulate different physiological functions, e.g., sensation, growth, digestion, reproductivity, nervous and immune systems response, and many others. In eukaryotes, they are also responsible for intercellular communication in response to pathogens. The major primary messengers binding to these cell-surface receptors constitute small-molecule or peptide hormones and neurotransmitters, nucleotides, lipids as well as small proteins. The simplicity of the way how GPCR signaling can be regulated by their endogenous agonists prompted the usage of GPCRs as major drug targets in modern pharmacology. Drugs targeting GPCRs inhibit pathological processes at the very beginning. This enables to significantly reduce the occurrence of morphological changes caused by diseases. Until recently, X-ray crystallography was the method of the first choice to obtain high-resolution structural information about GPCRs. Following X-ray crystallography, cryo-EM gained attention in GPCR studies as a quick and low-cost alternative. FRET microscopy is also widely used for GPCRs in the analysis of protein-protein interactions (PPIs) in intact cells as well as for screening purposes. Regarding computational methods, molecular dynamics (MD) for many years has proven its usefulness in studying the GPCR activation. MODELLER and Rosetta were widely used to generate preliminary homology models of GPCRs for MD simulation systems. Apart from the conventional all-atom approach with explicitly defined solvent, also other techniques have been applied to GPCRs, e.g., MARTINI or hybrid methods involving the coarse-grained representation, less demanding regarding computational resources, and thus offering much larger simulation timescales.
Collapse
Affiliation(s)
- Paulina Dragan
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
| | | | | | - Dorota Latek
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
9
|
Sonawani A, Kharche S, Dasgupta D, Sengupta D. Insights into the dynamic interactions at chemokine-receptor interfaces and mechanistic models of chemokine binding. J Struct Biol 2022; 214:107877. [PMID: 35750237 DOI: 10.1016/j.jsb.2022.107877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/20/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022]
Abstract
Chemokine receptors are the central signaling hubs of several processes such as cell migration, chemotaxis and cell positioning. In this graphical review, we provide an overview of the structural and mechanistic principles governing chemokine recognition that are currently emerging. Structural models of chemokine-receptor co-complexes with endogenous chemokines, viral chemokines and therapeutics have been resolved that highlight multiple interaction sites, termed as CRS1, CRS1.5 etc. The first site of interaction has been shown to be the N-terminal domain of the receptors (CRS1 site). A large structural flexibility of the N-terminal domain has been reported that was supported by both experimental and simulation studies. Upon chemokine binding, the N-terminal domain appears to show constricted dynamics and opens up to interact with the chemokine via a large interface. The subsequent sites such as CRS1.5 and CRS2 sites have been structurally well resolved although differences arise such as the localization of the N-terminus of the ligand to a major or minor pocket of the orthosteric binding site. Several computational studies have highlighted the dynamic protein-protein interface at the CRS1 site that seemingly appears to resolve the differences in NMR and mutagenesis studies. Interestingly, the differential dynamics at the CRS1 site suggests a mixed model of binding with complex signatures of both conformational selection and induced fit models. Integrative experimental and computational approaches could help unravel the structural basis of promiscuity and specificity in chemokine-receptor binding and open up new avenues of therapeutic design.
Collapse
Affiliation(s)
- Archana Sonawani
- School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, CBD Belapur, Navi Mumbai 400614, India
| | - Shalmali Kharche
- CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411 008, India
| | - Debjani Dasgupta
- School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, CBD Belapur, Navi Mumbai 400614, India
| | - Durba Sengupta
- CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411 008, India.
| |
Collapse
|
10
|
Li H, Wu M, Zhao X. Role of chemokine systems in cancer and inflammatory diseases. MedComm (Beijing) 2022; 3:e147. [PMID: 35702353 PMCID: PMC9175564 DOI: 10.1002/mco2.147] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
Chemokines are a large family of small secreted proteins that have fundamental roles in organ development, normal physiology, and immune responses upon binding to their corresponding receptors. The primary functions of chemokines are to coordinate and recruit immune cells to and from tissues and to participate in regulating interactions between immune cells. In addition to the generally recognized antimicrobial immunity, the chemokine/chemokine receptor axis also exerts a tumorigenic function in many different cancer models and is involved in the formation of immunosuppressive and protective tumor microenvironment (TME), making them potential prognostic markers for various hematologic and solid tumors. In fact, apart from its vital role in tumors, almost all inflammatory diseases involve chemokines and their receptors in one way or another. Modulating the expression of chemokines and/or their corresponding receptors on tumor cells or immune cells provides the basis for the exploitation of new drugs for clinical evaluation in the treatment of related diseases. Here, we summarize recent advances of chemokine systems in protumor and antitumor immune responses and discuss the prevailing understanding of how the chemokine system operates in inflammatory diseases. In this review, we also emphatically highlight the complexity of the chemokine system and explore its potential to guide the treatment of cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Hongyi Li
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of EducationWest China Second HospitalSichuan UniversityChengduChina
| |
Collapse
|
11
|
Le Gall C, Cammarata A, de Haas L, Ramos-Tomillero I, Cuenca-Escalona J, Schouren K, Wijfjes Z, Becker AMD, Bödder J, Dölen Y, de Vries IJM, Figdor CG, Flórez-Grau G, Verdoes M. Efficient targeting of NY-ESO-1 tumor antigen to human cDC1s by lymphotactin results in cross-presentation and antigen-specific T cell expansion. J Immunother Cancer 2022; 10:jitc-2021-004309. [PMID: 35428705 PMCID: PMC9014073 DOI: 10.1136/jitc-2021-004309] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2022] [Indexed: 12/20/2022] Open
Abstract
Background Type 1 conventional dendritic cells (cDC1s) are characterized by their ability to induce potent CD8+ T cell responses. In efforts to generate novel vaccination strategies, notably against cancer, human cDC1s emerge as an ideal target to deliver antigens. cDC1s uniquely express XCR1, a seven transmembrane G protein-coupled receptor. Due to its restricted expression and endocytic nature, XCR1 represents an attractive receptor to mediate antigen-delivery to human cDC1s. Methods To explore tumor antigen delivery to human cDC1s, we used an engineered version of XCR1-binding lymphotactin (XCL1), XCL1(CC3). Site-specific sortase-mediated transpeptidation was performed to conjugate XCL1(CC3) to an analog of the HLA-A*02:01 epitope of the cancer testis antigen New York Esophageal Squamous Cell Carcinoma-1 (NY-ESO-1). While poor epitope solubility prevented isolation of stable XCL1-antigen conjugates, incorporation of a single polyethylene glycol (PEG) chain upstream of the epitope-containing peptide enabled generation of soluble XCL1(CC3)-antigen fusion constructs. Binding and chemotactic characteristics of the XCL1-antigen conjugate, as well as its ability to induce antigen-specific CD8+ T cell activation by cDC1s, was assessed. Results PEGylated XCL1(CC3)-antigen conjugates retained binding to XCR1, and induced cDC1 chemoattraction in vitro. The model epitope was efficiently cross-presented by human cDC1s to activate NY-ESO-1-specific CD8+ T cells. Importantly, vaccine activity was increased by targeting XCR1 at the surface of cDC1s. Conclusion Our results present a novel strategy for the generation of targeted vaccines fused to insoluble antigens. Moreover, our data emphasize the potential of targeting XCR1 at the surface of primary human cDC1s to induce potent CD8+ T cell responses.
Collapse
Affiliation(s)
- Camille Le Gall
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Tumor Immunology, Oncode Institute, Nijmegen, The Netherlands
| | - Anna Cammarata
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Lukas de Haas
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Iván Ramos-Tomillero
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Jorge Cuenca-Escalona
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Kayleigh Schouren
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Zacharias Wijfjes
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| | - Anouk M D Becker
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Johanna Bödder
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Yusuf Dölen
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Tumor Immunology, Oncode Institute, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Department of Tumor Immunology, Oncode Institute, Nijmegen, The Netherlands
| | - Georgina Flórez-Grau
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboudumc Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- Institute for Chemical Immunology, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Transcriptome Profiling Reveals a Divergent Adaptive Response to Hyper- and Hypo-Salinity in the Yellow Drum, Nibea albiflora. Animals (Basel) 2021; 11:ani11082201. [PMID: 34438658 PMCID: PMC8388402 DOI: 10.3390/ani11082201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Global warming and certain climate disasters (typhoon, tsunami, etc.) can lead to fluctuation in seawater salinity that causes salinity stress in fish. The aim of this study was to investigate the functional genes and relevant pathways in response to salinity stress in the yellow drum. Genes and pathways related to signal transduction, osmoregulation, and metabolism may be involved in the adaptive regulation to salinity in the yellow drum. Additionally, the genes under salinity stress were mainly divided into three expression trends. Our results provided novel insights into further study of the salinity adaptability of euryhaline fishes. Abstract The yellow drum (Nibea albiflora) is an important marine economic fish that is widely distributed in the coastal waters of the Northwest Pacific. In order to understand the molecular regulatory mechanism of the yellow drum under salinity stress, in the present study, transcriptome analysis was performed under gradients with six salinities (10, 15, 20, 25, 30, and 35 psu). Compared to 25 psu, 907, 1109, 1309, 18, and 243 differentially expressed genes (DEGs) were obtained under 10, 15, 20, 30, and 35 psu salinities, respectively. The differential gene expression was further validated by quantitative real-time PCR (qPCR). The results of the tendency analysis showed that all DEGs of the yellow drum under salinity fluctuation were mainly divided into three expression trends. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed that the PI3K-Akt signaling pathway, Jak-STAT signaling pathway as well as the glutathione metabolism and steroid biosynthesis pathways may be the key pathways for the salinity adaptive regulation mechanism of the yellow drum. G protein-coupled receptors (GPCRs), the solute carrier family (SLC), the transient receptor potential cation channel subfamily V member 6 (TRPV6), isocitrate dehydrogenase (IDH1), and fructose-bisphosphate aldolase C-B (ALDOCB) may be the key genes in the response of the yellow drum to salinity stress. This study explored the transcriptional patterns of the yellow drum under salinity stress and provided fundamental information for the study of salinity adaptability in this species.
Collapse
|
13
|
Xi K, Hu Z, Wu Q, Wei M, Qian R, Zhu L. Assessing the Performance of Traveling-salesman based Automated Path Searching (TAPS) on Complex Biomolecular Systems. J Chem Theory Comput 2021; 17:5301-5311. [PMID: 34270241 DOI: 10.1021/acs.jctc.1c00182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Though crucial for understanding the function of large biomolecular systems, locating the minimum free energy paths (MFEPs) between their key conformational states is far from trivial due to their high-dimensional nature. Most existing path-searching methods require a static collective variable space as input, encoding intuition or prior knowledge of the transition mechanism. Such information is, however, hardly available a priori and expensive to validate. To alleviate this issue, we have previously introduced a Traveling-salesman based Automated Path Searching method (TAPS) and demonstrated its efficiency on simple peptide systems. Having implemented a parallel version of this method, here we assess the performance of TAPS on three realistic systems (tens to hundreds of residues) in explicit solvents. We show that TAPS successfully located the MFEP for the ground/excited state transition of the T4 lysozyme L99A variant, consistent with previous findings. TAPS also helped identifying the important role of the two polar contacts in directing the loop-in/loop-out transition of the mitogen-activated protein kinase kinase (MEK1), which explained previous mutant experiments. Remarkably, at a minimal cost of 126 ns sampling, TAPS revealed that the Ltn40/Ltn10 transition of lymphotactin needs no complete unfolding/refolding of its β-sheets and that five polar contacts are sufficient to stabilize the various partially unfolded intermediates along the MFEP. These results present TAPS as a general and promising tool for studying the functional dynamics of complex biomolecular systems.
Collapse
Affiliation(s)
- Kun Xi
- Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong 518172, P. R. China.,School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Zhenquan Hu
- Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong 518172, P. R. China.,School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Qiang Wu
- School of Science and Engineering, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong 518172, P. R. China
| | - Meihan Wei
- Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong 518172, P. R. China
| | - Runtong Qian
- Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong 518172, P. R. China
| | - Lizhe Zhu
- Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong 518172, P. R. China
| |
Collapse
|
14
|
Recent Progress in Dendritic Cell-Based Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13102495. [PMID: 34065346 PMCID: PMC8161242 DOI: 10.3390/cancers13102495] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/10/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Cancer immunotherapy has now attracted much attention because of the recent success of immune checkpoint inhibitors. However, they are only beneficial in a limited fraction of patients most probably due to lack of sufficient CD8+ cytotoxic T-lymphocytes against tumor antigens in the host. In this regard, dendritic cells are useful tools to induce host immune responses against exogenous antigens. In particular, recently characterized cross-presenting dendritic cells are capable of inducing CD8+ cytotoxic T-lymphocytes against exogenous antigens such as tumor antigens and uniquely express the chemokine receptor XCR1. Here we focus on the recent progress in DC-based cancer vaccines and especially the use of the XCR1 and its ligand XCL1 axis for the targeted delivery of cancer vaccines to cross-presenting dendritic cells. Abstract Cancer immunotherapy aims to treat cancer by enhancing cancer-specific host immune responses. Recently, cancer immunotherapy has been attracting much attention because of the successful clinical application of immune checkpoint inhibitors targeting the CTLA-4 and PD-1/PD-L1 pathways. However, although highly effective in some patients, immune checkpoint inhibitors are beneficial only in a limited fraction of patients, possibly because of the lack of enough cancer-specific immune cells, especially CD8+ cytotoxic T-lymphocytes (CTLs), in the host. On the other hand, studies on cancer vaccines, especially DC-based ones, have made significant progress in recent years. In particular, the identification and characterization of cross-presenting DCs have greatly advanced the strategy for the development of effective DC-based vaccines. In this review, we first summarize the surface markers and functional properties of the five major DC subsets. We then describe new approaches to induce antigen-specific CTLs by targeted delivery of antigens to cross-presenting DCs. In this context, the chemokine receptor XCR1 and its ligand XCL1, being selectively expressed by cross-presenting DCs and mainly produced by activated CD8+ T cells, respectively, provide highly promising molecular tools for this purpose. In the near future, CTL-inducing DC-based cancer vaccines may provide a new breakthrough in cancer immunotherapy alone or in combination with immune checkpoint inhibitors.
Collapse
|
15
|
Koch C, Engele J. Functions of the CXCL12 Receptor ACKR3/CXCR7-What Has Been Perceived and What Has Been Overlooked. Mol Pharmacol 2020; 98:577-585. [PMID: 32883765 DOI: 10.1124/molpharm.120.000056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022] Open
Abstract
The CXCL12 system is central to the development of many organs and is further crucially engaged in pathophysiological processes underlying cancer, inflammation, and cardiovascular disorders. This disease-associated role presently focuses major interest on the two CXCL12 receptors, CXCR4 and atypical chemokine receptor 3 (ACKR3)/CXCR7, as promising therapeutic targets. Major obstacles in these ongoing efforts are confusing reports on the differential use of either ACKR3/CXCR7 and/or CXCR4 across various cells as well as on the specific function(s) of ACKR3/CXCR7. Although basically no doubts remain that CXCR4 represents a classic chemokine receptor, functions assigned to ACKR3/CXCR7 range from those of a strictly silent scavenger receptor eventually modulating CXCR4 signaling to an active and independent signaling receptor. In this review, we depict a thorough analysis of our present knowledge on different modes of organization and functions of the cellular CXCL12 system. We further highlight the potential role of ACKR3/CXCR7 as a "crosslinker" of different receptor systems. Finally, we discuss mechanisms with the potency to impinge on the cellular organization of the CXCL12 system and hence might represent additional future therapeutic targets. SIGNIFICANCE STATEMENT: Delineating the recognized functions of atypical chemokine receptor 3 and CXCR4 in CXCL12 signaling is central to the more detailed understanding of the role of the CXCL12 system in health and disease and will help to guide future research efforts.
Collapse
Affiliation(s)
- Christian Koch
- Institute of Anatomy, University of Leipzig, Medical Faculty, Leipzig, Germany
| | - Jürgen Engele
- Institute of Anatomy, University of Leipzig, Medical Faculty, Leipzig, Germany
| |
Collapse
|
16
|
Jiang BC, Liu T, Gao YJ. Chemokines in chronic pain: cellular and molecular mechanisms and therapeutic potential. Pharmacol Ther 2020; 212:107581. [DOI: 10.1016/j.pharmthera.2020.107581] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/15/2020] [Indexed: 02/08/2023]
|
17
|
Transcutaneous immunization with a highly active form of XCL1 as a vaccine adjuvant using a hydrophilic gel patch elicits long-term CD8 + T cell responses. J Pharmacol Sci 2020; 143:182-187. [PMID: 32386904 DOI: 10.1016/j.jphs.2020.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/30/2020] [Accepted: 04/06/2020] [Indexed: 01/22/2023] Open
Abstract
Memory CD8+ cytotoxic T-lymphocytes (CTLs) play a key role in protective immunity against infection and cancer. However, the induction of memory CTLs with currently available vaccines remains difficult. The chemokine receptor XCR1 is predominantly expressed on CD103+ cross-presenting dendritic cells (DCs). Recently, we have demonstrated that a high activity form of murine lymphotactin/XCL1 (mXCL1-V21C/A59C), a ligand of XCR1, can induce antigen-specific memory CTLs by increasing the accumulation of CD103+ DCs in the vaccination site and the regional lymph nodes. Here, we combined a hydrophilic gel patch as a transcutaneous delivery device and mXCL1-V21C/A59C as an adjuvant to further enhance memory CTL responses. The transcutaneous delivery of ovalbumin (OVA) and mXCL1-V21C/A59C by the hydrophilic gel patch increased CD103+ DCs in the vaccination site and the regional lymph nodes for a prolonged period of time compared with the intradermal injection of OVA and mXCL1-V21C/A59C. Furthermore, the hydrophilic gel patch containing OVA and mXCL1-V21C/A59C strongly induced OVA-specific memory CTLs and efficiently inhibited the growth of OVA-expressing tumors more than the intradermal injection of OVA and mXCL1-V21C/A59C. Collectively, this type of hydrophilic gel patch and a high activity form of XCL1 may provide a useful tool for the induction of memory CTL responses.
Collapse
|
18
|
Kharche SA, Sengupta D. Dynamic protein interfaces and conformational landscapes of membrane protein complexes. Curr Opin Struct Biol 2020; 61:191-197. [DOI: 10.1016/j.sbi.2020.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/30/2019] [Accepted: 01/01/2020] [Indexed: 12/15/2022]
|