Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Mol Immunol ; 166: 39-49, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38219401

ABSTRACT

Butyrophilin-like 2 (BTNL2) is a T cell inhibitory molecule that interacts with unknown binding partners to modulate the immune response in a number of inflammatory and autoimmune diseases. In this study, we found that the inhibitory effects of BTNL2 on T cell activation and effector functions can be executed by its N-terminal IgV domain (BTNL2 IgV1) alone. Structure-guided mutation of key residues on BTNL2 IgV1 based on known receptor-ligand interfaces involving immunoglobulin superfamily members revealed that BTNL2 uses a non-canonical binding interface with its putative receptor. A high avidity BTNL2 IgV1 probe revealed that in an inducible model of ulcerative colitis, severe colitis was accompanied by a selective enrichment of BTNL2-receptor expressing effector-memory CD4+ and CD8+ T cells in the Peyer's patches. Intraperitoneal administration of BTNL2 IgV1 resulted in a significant delay in the progression of DSS-induced colitis and also showed reduced activation of the BTNL2-receptor-expressing T cells in the Peyer's patches. Thus, this study demonstrates that the BTNL2-receptor-expressing T cells in the Peyer's patches participate in the disease pathogenesis and can serve as a novel therapeutic target in ulcerative colitis, which can be modulated by BTNL2 IgV1.


Subject(s)
Colitis, Ulcerative , Colitis , Butyrophilins/metabolism , CD8-Positive T-Lymphocytes , Colitis, Ulcerative/chemically induced , Peyer's Patches/metabolism , Animals
2.
Biochem Soc Trans ; 51(6): 2103-2115, 2023 12 20.
Article in English | MEDLINE | ID: mdl-37970977

ABSTRACT

Cadherins are type-I membrane glycoproteins that primarily participate in calcium-dependent cell adhesion and homotypic cell sorting in various stages of embryonic development. Besides their crucial role in cellular and physiological processes, increasing studies highlight their involvement in pathophysiological functions ranging from cancer progression and metastasis to being entry receptors for pathogens. Cadherins mediate these cellular processes through homophilic, as well as heterophilic interactions (within and outside the superfamily) by their membrane distal ectodomains. This review provides an in-depth structural perspective of molecular recognition among type-I and type-II classical cadherins. Furthermore, this review offers structural insights into different dimeric assemblies like the 'strand-swap dimer' and 'X-dimer' as well as mechanisms relating these dimer forms like 'two-step adhesion' and 'encounter complex'. Alongside providing structural details, this review connects structural studies to bond mechanics merging crystallographic and single-molecule force spectroscopic findings. Finally, the review discusses the recent discoveries on dimeric intermediates that uncover prospects of further research beyond two-step adhesion.


Subject(s)
Cadherins , Nanotechnology , Cell Adhesion/physiology , Cadherins/metabolism
3.
Biosens Bioelectron ; 242: 115733, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37820555

ABSTRACT

A soluble isoform of cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) has been found in the serum of healthy individuals and alterations in its expression level have been linked with the development and progression of various cancers. Conventionally, soluble CTLA-4 (sCTLA-4) has been quantified by techniques such as ELISA, western blot, and flow cytometry, which however are time-consuming, highly expensive and require large sample volumes. Therefore, rapid, cost-effective and real-time monitoring of soluble CTLA-4 levels is much needed to facilitate timely diagnosis of a worsening disease and help patient selection for immunotherapeutic interventions in cancer. Here, for the first time, we report an ultrasensitive, highly selective electrochemical nanobody (NAb) based biosensor for the quantitative detection of soluble CTLA-4 employing poly(3,4-ethylenedioxythiophene) polystyrene sulfonate (PEDOT:PSS) and gold nanoparticles modified electrode with attomole sensitivity. Incorporating nanomaterials with conductive polymers enhances the sensitivity of the electrochemical biosensor, while the nanobody's stability, specificity and ease of production make it a suitable choice as a bioreceptor. The proposed NAb-based sensor can detect sCTLA-4 from pure recombinant protein in a wide concentration range of 100 ag mL-1- 500 µg mL-1, with a limit of detection of 1.19 ag mL-1 (+3σ of the blank signal). The sensor's relative standard deviation for reproducibility is less than 0.4% and has effective real sample analytics for cell culture supernatant with no significant difference with pure recombinant protein (p < 0.05). Our proposed nanobody based sensor exhibits stability for up to 2 weeks (<3% variation). Moreover, this nanobody-based sensor presents a future opportunity for quantitative, ultrasensitive, and economical biosensor development that can be adapted to monitor the immune landscape of cancer patients to provide a larger therapeutic window.


Subject(s)
Biosensing Techniques , Metal Nanoparticles , Neoplasms , Humans , CTLA-4 Antigen , Gold , Reproducibility of Results , Biosensing Techniques/methods , Neoplasms/diagnosis , Recombinant Proteins , Electrochemical Techniques/methods , Limit of Detection
4.
Eur J Immunol ; 53(12): e2350528, 2023 12.
Article in English | MEDLINE | ID: mdl-37698527

ABSTRACT

Immunotherapeutic modulation of antigen-specific T-cell responses instead of the whole repertoire helps avoid immune-related adverse events. We have developed an artificial antigen-presenting system (aAPS) where multiple copies of a multimeric peptide-MHC class I complex presenting a murine class I MHC restricted ovalbumin-derived peptide (signal 1), along with a costimulatory ligand (signal 2) are chemically conjugated to a dextran backbone. Cognate naive CD8+ T cells, when treated with this aAPS underwent significant expansion and showed an activated phenotype. Furthermore, elevated expression of effector cytokines led to the differentiation of these cells to cytotoxic T lymphocytes which resulted in target cell lysis, indicative of the functional efficacy of the aAPS. CD8+ T cells with decreased proliferative potential due to repeated antigenic stimulation could also be re-expanded by the developed aAPS. Thus, the developed aAPS warrants further engineering for future application as a rapidly customizable personalized immunotherapeutic agent, incorporating patient-specific MHC-restricted tumor antigens and different costimulatory signals to modulate both naive and antigen-experienced but exhausted tumor-specific T cells in cancer.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Mice , Animals , Dextrans/metabolism , Lymphocyte Activation , Immunotherapy , Peptides/metabolism , Antigen-Presenting Cells , Neoplasms/therapy , Neoplasms/metabolism
5.
Biochem Biophys Res Commun ; 677: 31-37, 2023 10 15.
Article in English | MEDLINE | ID: mdl-37542773

ABSTRACT

TIGIT (T cell immunoglobulin and ITIM domain) is an inhibitory receptor expressed on T and NK cells that interact with cell surface glycoprotein belonging to the nectin and nectin-like family of cell adhesion molecules, particularly nectin-2 and nectin-like 5 (PVR). Nectin-4 has been recently identified as a novel ligand for TIGIT and the interaction among them inhibits NK cell cytotoxicity. In this study, biophysical experiments were conducted to decipher the mechanism of this novel interaction, followed by structure-guided mutagenesis studies to map the nectin-4 binding interface on TIGIT. Using surface plasmon resonance, we deduced that TIGIT recognizes the membrane distal ectodomain of nectin-4 and the interaction is weaker than the well-characterized TIGIT: nectin-2 interaction. Deciphering the molecular basis of this newly identified interaction between TIGIT and nectin-4 will provide us important insight into the manipulation of this inhibitory signaling pathway, especially targeting cancer cells overexpressing nectin-4 that evade the immune surveillance of the body.


Subject(s)
Cell Adhesion Molecules , Neoplasms , Nectins/genetics , Nectins/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Receptors, Immunologic , Killer Cells, Natural , Immunotherapy , Neoplasms/genetics , Neoplasms/therapy , Neoplasms/metabolism
6.
Mol Biol Rep ; 50(5): 4665-4673, 2023 May.
Article in English | MEDLINE | ID: mdl-37016039

ABSTRACT

Viruses are obligate intracellular parasites that depend on host cellular machinery for performing even basic biological functions. One of the many ways they achieve this is through molecular mimicry, wherein the virus mimics a host sequence or structure, thereby being able to hijack the host's physiological interactions for its pathogenesis. Such adaptations are specific recognitions that often confer tissue and species-specific tropisms to the virus, and enable the virus to utilise previously existing host signalling networks, which ultimately aid in further steps of viral infection, such as entry, immune evasion and spread. A common form of sequence mimicry utilises short linear motifs (SLiMs). SLiMs are short-peptide sequences that mediate transient interactions and are major elements in host protein interaction networks. This work is aimed at providing a comprehensive review of current literature of some well-characterised SLiMs that play a role in the attachment and entry of viruses into host cells, which mimic physiological receptor-ligand interactions already present in the host. Considering recent trends in emerging diseases, further research on such motifs involved in viral entry can help in the discovery of previously unknown cellular receptors utilised by viruses, as well as help in the designing of targeted therapeutics such as vaccines or inhibitors directed towards these interactions.


Subject(s)
Virus Diseases , Viruses , Humans , Molecular Mimicry , Protein Interaction Maps , Receptors, Cell Surface/metabolism , Host-Pathogen Interactions
7.
Arch Biochem Biophys ; 727: 109329, 2022 09 30.
Article in English | MEDLINE | ID: mdl-35738425

ABSTRACT

Cadherins are a family of cell surface glycoproteins that mediate Ca2+-dependent cell to cell adhesion. They organize to form large macromolecular assemblies at the junctions of cells in order to form and maintain the integrity of tissue structures, thereby playing an indispensable role in the multicellular organization. Notably, a large body of research on E- and N-cadherin, the two most widely studied members of the cadherin superfamily, suggest for homophilic associations among them to drive cell adhesion. Interestingly, latest studies also highlight for direct crosstalk among these two classical cadherins to form heterotypic connections in physiological as well as in disease environment. However, the molecular details for the heterophilic association of E-cadherin and N-cadherin has not been investigated yet, which we aimed to address in this work. Using surface plasmon resonance and flow cytometry based biophysical studies we observed heterophilic interaction between E- and N-cadherin mediated through the membrane distal ectodomains. Further, the heterodimeric interface of E-cadherin and N-cadherin was mapped using structure-guided mutational studies followed by complementary biophysical analyses to identify the important interface residues involved in the interaction. The results obtained imply significant resemblance in the interface residues of E-cadherin that are crucial for homophilic recognition of E-cadherin and heterophilic recognition of N-cadherin as well.


Subject(s)
Cadherins , Cadherins/metabolism , Cell Adhesion/physiology , Dimerization , Mutation , Protein Binding
8.
Int J Biol Macromol ; 210: 494-503, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35504420

ABSTRACT

Tuberculosis, caused by Mycobacterium tuberculosis, is predominantly a disease of the lungs acquired by inhaling mycobacteria from infected individuals via airborne droplets. In order to facilitate their entry into the alveolar macrophages, mycobacteria have a collection of pathogen-associated molecular patterns (PAMPs) on their surface that are known to detect certain pattern recognition receptors present on the surface of host cells. A major group of these PAMPs includes mycobacterial lipoproteins, of which, the 19 kDa surface antigen LpqH, has been reported to play a critical role in both host-pathogen interactions as well as pleiotropic immune regulation. Despite its crucial involvement in tuberculosis, the detailed structure-function relationship of this protein remains to be explored. Here, we report the high-resolution crystal structure of the non-acylated LpqH (LpqH48-159) at a resolution of 1.26 Å, which adopts a unique fold. Flow cytometry-based experiments show that the protein can bind and induce apoptosis in PMA-activated human monocytic cell line THP-1, indicative of the preservation of functionality of the protein. Furthermore, analysis of conservation of LpqH sequences from Mycobacterium species reveals a patch of conserved residues on the surface which may play a role in its binding partner recognition and hence in host-pathogen interaction.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Lipoproteins/metabolism , Monocytes/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Tuberculosis/microbiology
9.
Biochim Biophys Acta Rev Cancer ; 1876(2): 188589, 2021 12.
Article in English | MEDLINE | ID: mdl-34237351

ABSTRACT

Tumour cells achieve maximum survival by modifying cellular machineries associated with processes such as cell division, migration, survival, and apoptosis, resulting in genetically complex and heterogeneous populations. While nectin and nectin-like cell adhesion molecules control development and maintenance of multicellular organisation in higher vertebrates by mediating cell-cell adhesion and related signalling processes, recent studies indicate that they also critically regulate growth and development of different types of cancers. In this review, we detail current knowledge about the role of nectin family members in various tumours. Furthermore, we also analyse the seemingly opposing roles of some members of nectin family in tumour-associated pathways, as they function as both tumour suppressors and oncogenes. Understanding this functional duality of nectin family in tumours will further our knowledge of molecular mechanisms regulating tumour development and progression, and contribute to the advancement of tumour diagnosis and therapy.


Subject(s)
Cell Adhesion Molecules/metabolism , Nectins/metabolism , Neoplasms/genetics
10.
Protein Sci ; 30(9): 1958-1973, 2021 09.
Article in English | MEDLINE | ID: mdl-34191384

ABSTRACT

T-cell co-stimulation through CD28/CTLA4:B7-1/B7-2 axis is one of the extensively studied pathways that resulted in the discovery of several FDA-approved drugs for autoimmunity and cancer. However, many aspects of the signaling mechanism remain elusive, including oligomeric association and clustering of B7-2 on the cell surface. Here, we describe the structure of the IgV domain of B7-2 and its cryptic association into 1D arrays that appear to represent the pre-signaling state of B7-2 on the cell membrane. Super-resolution microscopy experiments on heterologous cells expressing B7-2 and B7-1 suggest, B7-2 form relatively elongated and larger clusters compared to B7-1. The sequence and structural comparison of other B7 family members, B7-1:CTLA4 and B7-2:CTLA-4 complex structures, support our view that the observed B7-2 1D zipper array is physiologically important. This observed 1D zipper-like array also provides an explanation for its clustering, and upright orientation on the cell surface, and avoidance of spurious signaling.


Subject(s)
B7-1 Antigen/chemistry , B7-2 Antigen/chemistry , CD28 Antigens/chemistry , CTLA-4 Antigen/chemistry , Amino Acid Sequence , Animals , B7-1 Antigen/genetics , B7-1 Antigen/metabolism , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Binding Sites , CD28 Antigens/genetics , CD28 Antigens/metabolism , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Gene Expression , Humans , Mice , Models, Molecular , Neurons/cytology , Neurons/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
11.
Cell Microbiol ; 23(5): e13316, 2021 05.
Article in English | MEDLINE | ID: mdl-33543826

ABSTRACT

Cell adhesion molecules mediate cell-to-cell and cell-to-matrix adhesions and play an immense role in a myriad of physiological processes during the growth and development of a multicellular organism. Cadherins belong to a major group of membrane-bound cell surface proteins that, in coordination with nectins, drive the formation and maintenance of adherens junctions for mediating cell to cell adhesion, cellular communication and signalling. Alongside adhesive function, the involvement of cadherins in mediating host-pathogen interactions has been extensively explored in recent years. In this review, we provide an in-depth understanding of microbial pathogens and their virulence factors that exploit cadherins for their strategical invasion into the host cell. Furthermore, macromolecular interactions involving cadherins and various microbial factors such as secretory toxins and adhesins lead to the disintegration of host cell junctions followed by the entry of the pathogen or triggering downstream signalling pathways responsible for successful invasion of the pathogenic microbes are discussed. Besides providing a comprehensive insight into some of the structural complexes involving cadherins and microbial factors to offer the mechanistic details of host-pathogen interactions, the current review also highlights novel constituents of various cell signalling events such as endocytosis machinery elicited upon microbial infections.


Subject(s)
Bacteria/pathogenicity , Cadherins/metabolism , Fungi/pathogenicity , Host-Pathogen Interactions , Viruses/pathogenicity , Animals , Bacteria/metabolism , Bacterial Infections/microbiology , Endocytosis , Fungi/metabolism , Humans , Mycoses/microbiology , Signal Transduction , Virulence , Virulence Factors/metabolism , Virus Diseases/virology
12.
FEMS Microbiol Lett ; 368(2)2021 02 04.
Article in English | MEDLINE | ID: mdl-33355339

ABSTRACT

Immunoglobulin (Ig) domains are one of the most widespread protein domains encoded by the human genome and are present in a large array of proteins with diverse biological functions. These Ig domains possess a central structure, the immunoglobulin-fold, which is a sandwich of two ß sheets, each made up of anti-parallel ß strands, surrounding a central hydrophobic core. Apart from humans, proteins containing Ig-like domains are also distributed in a vast selection of organisms including vertebrates, invertebrates, plants, viruses and bacteria where they execute a wide array of discrete cellular functions. In this review, we have described the key structural deviations of bacterial Ig-folds when compared to the classical eukaryotic Ig-fold. Further, we have comprehensively grouped all the Ig-domain containing adhesins present in both Gram-negative and Gram-positive bacteria. Additionally, we describe the role of these particular adhesins in host tissue attachment, colonization and subsequent infection by both pathogenic and non-pathogenic Escherichia coli as well as other bacterial species. The structural properties of these Ig-domain containing adhesins, along with their interactions with specific Ig-like and non Ig-like binding partners present on the host cell surface have been discussed in detail.


Subject(s)
Adhesins, Bacterial/chemistry , Bacterial Infections/immunology , Bacterial Infections/microbiology , Immunoglobulins/chemistry , Adhesins, Bacterial/genetics , Animals , Host-Pathogen Interactions/immunology , Humans
13.
Biochem Biophys Res Commun ; 534: 504-510, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33220924

ABSTRACT

Nectins are a family of four cell surface glycoproteins belonging to the immunoglobulin superfamily that mediate cell-cell adhesion and associated signalling pathways, thereby regulating several physiological processes including morphogenesis, growth and development of multicellular organisms. Nectins interact among themselves through their extracellular domains from the adjacent cells in both homophilic and heterophilic fashions to support cell-cell adhesion. Although nectins form homodimers as demonstrated in experimental set-ups, only the specific heterophilic interactions among nectins are physiologically relevant as shown by in vivo studies. It has been hypothesised that a conserved charged residue present at the binding interface acts as the molecular switch for heterophilic nectin-nectin recognitions. In this work, we have analysed the energetics of homophilic and heterophilic interactions of nectins, followed by surface plasmon resonance-based binding studies and complementary in silico analyses. Our findings confirm that the conserved charged residues at the binding interfaces dictate the specificity of the nectin-nectin heterophilic interactions. Furthermore, these residues also play a role in conferring higher affinity to the heterophilic interactions, thereby making them physiologically more prevalent compared to homophilic interactions. Thus, this work reveals the molecular basis of heterophilic recognitions among nectins that contribute to their physiological functions.


Subject(s)
Nectins/metabolism , Amino Acid Sequence , Animals , Conserved Sequence , Humans , Models, Molecular , Nectins/chemistry , Protein Interaction Domains and Motifs , Protein Interaction Maps , Protein Multimerization , Static Electricity
14.
J Mol Biol ; 432(22): 5938-5950, 2020 11 06.
Article in English | MEDLINE | ID: mdl-32976909

ABSTRACT

T cell costimulation is mediated by the interaction of a number of receptors and ligands present on the surface of the T cell and antigen-presenting cell, respectively. Stimulatory or inhibitory signals from these receptor-ligand interactions work in tandem to preserve immune homeostasis. BTNL2 is a type-1 membrane protein that provides inhibitory signal to T cells and plays an important role in several inflammatory and autoimmune diseases. Therefore, manipulation of the molecular interaction of BTNL2 with its putative receptor could provide strategies to restore immune homeostasis in these diseases. Hence, it is imperative to study the structural characteristics of this molecule, which will provide important insights into its function as well. In this study, the membrane-distal ectodomain of murine BTNL2 was expressed in bacteria as inclusion bodies, refolded in vitro and purified for functional and structural characterization. The domain is monomeric in solution as demonstrated by size-exclusion chromatography and analytical ultracentrifugation, and also binds to its putative receptor on naïve B cells and activated T cell subsets. Importantly, for the first time, we report the structure of BTNL2 as determined by solution NMR spectroscopy and also the picosecond-nanosecond timescale backbone dynamics of this domain. The N-terminal ectodomain of BTNL2, which was able to inhibit T cell function as well, exhibits distinctive structural features. The N-terminal ectodomain of BTNL2 has a significantly reduced surface area in the front sheet due to the non-canonical conformation of the CC' loop, which provides important insights into the recognition of its presently unknown binding partner.


Subject(s)
Butyrophilins/chemistry , Immunoglobulin Domains , T-Lymphocytes/immunology , Animals , Butyrophilins/genetics , Homeostasis , Ligands , Lymphocyte Activation , Membrane Proteins/chemistry , Mice , Models, Molecular , Protein Conformation
15.
Sci Rep ; 10(1): 9434, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32523039

ABSTRACT

Nectin and nectin-like cell adhesion molecules (collectively referred as nectin family henceforth) are known to mediate cell-cell adhesion and related functions. While current literature suggests that nectins are prevalent in vertebrates, there are no in-depth analyses regarding the evolution of nectin family as a whole. In this work, we examine the evolutionary origin of the nectin family, using selected multicellular metazoans representing diverse clades whose whole genome sequencing data is available. Our results show that this family may have appeared earlier during metazoan evolution than previously believed. Systematic analyses indicate the order in which various members of nectin family seem to have evolved, with some nectin-like molecules appearing first, followed by the evolution of other members. Furthermore, we also found a few possible ancient homologues of nectins. While our study confirms the previous grouping of the nectin family into nectins and nectin-like molecules, it also shows poliovirus receptor (PVR/nectin-like-5) to possess characteristics that are intermediate between these two groups. Interestingly, except for PVR, the other nectins show surprising sequence conservations across species, suggesting evolutionary constraints due to critical roles played by these proteins.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion/genetics , Nectins/genetics , Animals , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line , Computational Biology/methods , Evolution, Molecular , Humans , Nectins/metabolism
16.
Proc Natl Acad Sci U S A ; 116(7): 2634-2639, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30683721

ABSTRACT

Random amino acid copolymers used in the treatment of multiple sclerosis in man or experimental autoimmune encephalomyelitis (EAE) in mice [poly(Y,E,A,K)n, known as Copaxone, and poly(Y,F,A,K)n] function at least in part by generation of IL-10-secreting regulatory T cells that mediate bystander immunosuppression. The mechanism through which these copolymers induce Tregs is unknown. To investigate this question, four previously described Vα3.2 Vß14 T cell receptor (TCR) cDNAs, the dominant clonotype generated in splenocytes after immunization of SJL mice, that differed only in their CDR3 sequences were utilized to generate retrogenic mice. The high-level production of IL-10 as well as IL-5 and small amounts of the related cytokines IL-4 and IL-13 by CD4+ T cells isolated from the splenocytes of these mice strongly suggests that the TCR itself encodes information for specific cytokine secretion. The proliferation and production of IL-10 by these Tregs was costimulated by activation of glucocorticoid-induced TNF receptor (GITR) (expressed at high levels by these cells) through its ligand GITRL. A mechanism for generation of cells with this specificity is proposed. Moreover, retrogenic mice expressing these Tregs were protected from induction of EAE by the appropriate autoantigen.


Subject(s)
Hematopoietic Stem Cells/cytology , Interleukin-10/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Animals , DNA, Complementary , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Genetic Vectors , Immune Tolerance , Interleukins/metabolism , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Tumor Necrosis Factors/metabolism
17.
Proteins ; 86(11): 1157-1164, 2018 11.
Article in English | MEDLINE | ID: mdl-30183103

ABSTRACT

Cell adhesion molecules such as nectins and cadherins play important role in the formation of adherens junction. While nectins interact through their extracellular domains in both homophilic and heterophilic manner among themselves, extracellular domains of cadherins participate only in homophilic fashion to mediate cell-cell adhesion. It is well established that nectins recruit cadherins in the adhesion sites through an interplay of adaptor molecules in the cytoplasmic side thereby increasing the effective concentration of both the adhesion molecules on the cell surface. This study provides molecular and structural bases of the novel interaction between extracellular domains of nectin-2 and N-cadherin, by which nectins can also recruit cadherins at the site of adherens junction through an adaptor-independent mechanism. Surface plasmon resonance study demonstrates that nectin-2 can directly recognize N-cadherin with a KD of 3.5 ± 0.6 µM which is physiologically relevant considering the affinities between other cell adhesion molecules including cadherin dimerization. Furthermore, structural analysis of currently available homodimeric structures of both nectin-2 and N-cadherin followed by molecular docking as well as complementary mutagenesis studies revealed the binding interface of this novel interaction.


Subject(s)
Cadherins/metabolism , Nectins/metabolism , Cadherins/chemistry , Cadherins/genetics , Humans , Molecular Docking Simulation , Nectins/chemistry , Nectins/genetics , Point Mutation , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Maps , Protein Multimerization , Protein Refolding
18.
PLoS One ; 12(1): e0170333, 2017.
Article in English | MEDLINE | ID: mdl-28099529

ABSTRACT

Each cycle of translation initiation in bacterial cell requires free 50S and 30S ribosomal subunits originating from the post-translational dissociation of 70S ribosome from the previous cycle. Literature shows stable dissociation of 70S from model post-termination complexes by the concerted action of Ribosome Recycling Factor (RRF) and Elongation Factor G (EF-G) that interact with the rRNA bridge B2a/B2b joining 50S to 30S. In such experimental models, the role of full-length nascent protein was never considered seriously. We observed relatively slow release of full-length nascent protein from 50Sof post translation ribosome, and in that process, its toe prints on the rRNA in vivo and in in vitro translation with E.coli S30 extract. We reported earlier that a number of chemically unfolded proteins like bovine carbonic anhydrase (BCA), lactate dehydrogenase (LDH), malate dehydrogenase (MDH), lysozyme, ovalbumin etc., when added to free 70Sin lieu of the full length nascent proteins, also interact with identical RNA regions of the 23S rRNA. Interestingly the rRNA nucleotides that slow down release of the C-terminus of full-length unfolded protein were found in close proximity to the B2a/B2b bridge. It indicated a potentially important chemical reaction conserved throughout the evolution. Here we set out to probe that conserved role of unfolded protein conformation in splitting the free or post-termination 70S. How both the RRF-EFG dependent and the plausible nascent protein-EFG dependent ribosome recycling pathways might be relevant in bacteria is discussed here.


Subject(s)
Escherichia coli/metabolism , Peptide Chain Initiation, Translational/physiology , Peptide Chain Termination, Translational/physiology , Protein Biosynthesis/physiology , Ribosome Subunits, Large, Bacterial/metabolism , Ribosome Subunits, Small, Bacterial/metabolism , Animals , Carbonic Anhydrases/metabolism , Cattle , Chick Embryo , Escherichia coli/genetics , L-Lactate Dehydrogenase/metabolism , Malate Dehydrogenase/metabolism , Muramidase/metabolism , Ovalbumin/metabolism , Peptide Elongation Factor G/metabolism , Protein Folding , Ribosomal Proteins/metabolism , Swine
19.
Mol Immunol ; 81: 151-159, 2017 01.
Article in English | MEDLINE | ID: mdl-27978489

ABSTRACT

In addition to antigen-specific stimulation of T cell receptor (TCR) by a peptide-MHC complex, the functional outcome of TCR engagement is regulated by antigen-independent costimulatory signals. Costimulatory signals are provided by an array of interactions involving activating and inhibitory receptors expressed on T cells and their cognate ligands on antigen presenting cells. T cell immunoglobulin and ITIM domain (TIGIT), a recently identified immune receptor expressed on T and NK cells, upon interaction with either of its two ligands, nectin-2 or poliovirus receptor (PVR), inhibits activation of T and NK cells. Here we report the crystal structure of the human TIGIT ectodomain, which exhibits the classic two-layer ß-sandwich topology observed in other immunoglobulin super family (IgSF) members. Biophysical studies indicate that TIGIT is monomeric in solution but can form a dimer at high concentrations, consistent with the observation of a canonical immunoglobulin-like dimer interface in the crystalline state. Based on existing structural data, we present a model of the TIGIT:nectin-2 complex and utilized complementary biochemical studies to map the nectin-binding interface on TIGIT. Our data provide important structural and biochemical determinants responsible for the recognition of nectin-2 by TIGIT. Defining the TIGIT:nectin-2 binding interface provides the basis for rational manipulation of this molecular interaction for the development of immunotherapeutic reagents in autoimmunity and cancer.


Subject(s)
Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/immunology , Lymphocyte Activation/immunology , Models, Molecular , Receptors, Immunologic/chemistry , Receptors, Immunologic/immunology , Cell Adhesion Molecules/metabolism , Crystallography, X-Ray , Humans , Mutagenesis, Site-Directed , Nectins , Protein Binding , Protein Conformation , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/metabolism , Surface Plasmon Resonance
20.
Cell Mol Life Sci ; 72(4): 645-58, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25326769

ABSTRACT

Cell-cell adhesive processes are central to the physiology of multicellular organisms. A number of cell surface molecules contribute to cell-cell adhesion, and the dysfunction of adhesive processes underlies numerous developmental defects and inherited diseases. The nectins, a family of four immunoglobulin superfamily members (nectin-1 to -4), interact through their extracellular domains to support cell-cell adhesion. While both homophilic and heterophilic interactions among the nectins are implicated in cell-cell adhesion, cell-based and biochemical studies suggest heterophilic interactions are stronger than homophilic interactions and control a range of physiological processes. In addition to interactions within the nectin family, heterophilic associations with nectin-like molecules, immune receptors, and viral glycoproteins support a wide range of biological functions, including immune modulation, cancer progression, host-pathogen interactions and immune evasion. We review current structural and molecular knowledge of nectin recognition processes, with a focus on the biochemical and biophysical determinants of affinity and selectivity that drive distinct nectin associations. These proteins and interactions are discussed as potential targets for immunotherapy.


Subject(s)
Cell Adhesion Molecules/metabolism , Cadherins/chemistry , Cadherins/metabolism , Cell Adhesion/physiology , Cell Adhesion Molecules/chemistry , Herpesvirus 1, Human/metabolism , Host-Pathogen Interactions/immunology , Humans , Immunoglobulins/chemistry , Nectins , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL
...