Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 18(4): 412-421, 2017 04.
Article in English | MEDLINE | ID: mdl-28166218

ABSTRACT

Type 1 regulatory T cells (Tr1 cells) are induced by interleukin-27 (IL-27) and have critical roles in the control of autoimmunity and resolution of inflammation. We found that the transcription factors IRF1 and BATF were induced early on after treatment with IL-27 and were required for the differentiation and function of Tr1 cells in vitro and in vivo. Epigenetic and transcriptional analyses revealed that both transcription factors influenced chromatin accessibility and expression of the genes required for Tr1 cell function. IRF1 and BATF deficiencies uniquely altered the chromatin landscape, suggesting that these factors serve a pioneering function during Tr1 cell differentiation.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/immunology , Chromatin/metabolism , Interferon Regulatory Factor-1/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmunity , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Chromatin/genetics , Cluster Analysis , Cytokines/metabolism , Cytokines/pharmacology , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Interferon Regulatory Factor-1/genetics , Mice , Mice, Knockout , Promoter Regions, Genetic , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , Transcription Factors/metabolism , Transcriptome
2.
Eur J Immunol ; 54(7): e2350847, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38643381

ABSTRACT

Cenicriviroc, a dual CCR2/CCR5 antagonist, initially developed as an anti-HIV drug, has shown promising results in nonalcoholic steatohepatitis phase 2 clinical trials. It inhibits the infiltration and activation of CCR2+/CCR5+ monocytes and macrophages to the site of liver injury, preventing liver fibrosis. However, the role of Cenicriviroc in the modulation of helper T cell differentiation and functions remains to be explored. In inflamed colons of Crohn's disease patients, CCR2+ and CCR5+ CD4+ T cells are enriched. Considering the role of CCR2+ and CCR5+ T cells in IBD pathogenesis, we investigated the potential role of Cenicriviroc in colitis. Our in vitro studies revealed that Cenicriviroc inhibits Th1-, Th2-, and Th17-cell differentiation while promoting the generation of type 1 regulatory T cells (Tr1), known for preventing inflammation through induction of IL-10. This study is the first to report that Cenicriviroc promotes Tr1 cell generation by up-regulating the signature of Tr1 cell transcription factors such as c-Maf, Prdm1, Irf-1, Batf, and EGR-2. Cenicriviroc displayed a protective effect in experimental colitis models by preventing body weight loss and intestinal inflammation and preserving epithelial barrier integrity. We show that Cenicriviroc induced IL-10 and inhibited the generation of pro-inflammatory cytokines IFN-γ, IL-17, IL-6, and IL-1ß during colitis. Based on our data, we propose Cenicriviroc as a potential therapeutic in controlling tissue inflammation by inhibiting the generation and functions of effector T cells and promoting the induction of anti-inflammatory Tr1 cells.


Subject(s)
CCR5 Receptor Antagonists , Cell Differentiation , Colitis , Receptors, CCR2 , Receptors, CCR5 , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Animals , Mice , Receptors, CCR2/metabolism , Receptors, CCR2/antagonists & inhibitors , Colitis/immunology , Colitis/drug therapy , Colitis/chemically induced , Cell Differentiation/drug effects , Cell Differentiation/immunology , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use , Receptors, CCR5/metabolism , Humans , Th17 Cells/immunology , Th17 Cells/drug effects , Sulfoxides/pharmacology , Mice, Inbred C57BL , Th1 Cells/immunology , Th1 Cells/drug effects , Interleukin-10/metabolism , Th2 Cells/immunology , Imidazoles
3.
Eur J Immunol ; 54(7): e2350624, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38655818

ABSTRACT

Pathogenic infections cause thymic atrophy, perturb thymic T-cell development, and alter immunological response. Previous studies reported dysregulated T-cell function and lymphopenia in coronavirus disease-19 (COVID-19). However, immunopathological changes in the thymus associated with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection have not been elucidated. Here, we report that SARS-CoV-2 infects thymocytes, and induces CD4+CD8+ (double positive; DP) T-cell apoptosis leading to thymic atrophy and loss of peripheral TCR repertoire in K18-hACE2 transgenic mice. Infected thymus led to increased CD44+CD25- T-cells, indicating an early arrest in the T-cell maturation pathway. Thymic atrophy was notably higher in male hACE2-Tg mice than in females and involved an upregulated de-novo synthesis pathway of thymic glucocorticoid. Further, IFN-γ was crucial for thymic atrophy, as anti-IFN-γ -antibody neutralization blunted thymic involution. Therapeutic use of Remdesivir also rescued thymic atrophy. While the Omicron variant and its sub-lineage BA.5 variant caused marginal thymic atrophy, the delta variant of SARS-CoV-2 exhibited severe thymic atrophy characterized by severely depleted DP T-cells. Recently characterized broadly SARS-CoV-2 neutralizing monoclonal antibody P4A2 was able to rescue thymic atrophy and restore the thymic maturation pathway of T-cells. Together, we report SARS-CoV-2-associated thymic atrophy resulting from impaired T-cell maturation pathway which may contribute to dyregulated T cell response during COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2 , Atrophy , COVID-19 , Interferon-gamma , Mice, Transgenic , SARS-CoV-2 , Thymus Gland , Animals , COVID-19/immunology , COVID-19/pathology , SARS-CoV-2/immunology , Thymus Gland/pathology , Thymus Gland/immunology , Mice , Interferon-gamma/metabolism , Interferon-gamma/immunology , Atrophy/immunology , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Female , Humans , Male , Thymocytes/immunology , Apoptosis , CD8-Positive T-Lymphocytes/immunology
4.
Clin Gastroenterol Hepatol ; 22(6): 1295-1306.e7, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38278200

ABSTRACT

BACKGROUND & AIMS: Coconut water (CW) is anti-inflammatory, can manipulate the gut microbiome, and is a rich source of potassium. Gut microbiome modulation improves outcomes in ulcerative colitis (UC), and potassium possesses in vitro anti-inflammatory property. We evaluated the effect of CW as an adjunct therapy for patients with mild-moderate UC. METHODS: This single-center, double-blind, placebo-controlled trial randomized patients with mild to moderate (Simple Clinical Colitis Activity Index [SCCAI]: 3-9) endoscopically active UC (Ulcerative Colitis Endoscopic Index of Severity [UCEIS] >1) in 1:1 ratio to CW + standard medical therapy (SMT) vs placebo + SMT. Four hundred mL of CW was administered for 8 weeks. Primary outcome measure was clinical remission (SCCAI ≤2), and secondary outcome measures were clinical response (SCCAI decline ≥3) and adverse events at 8 weeks. Microbiome was analyzed at baseline and 8 weeks. RESULTS: Of 121 patients screened, 95 were included for modified intention to treat analysis (CW, n = 49; placebo, n = 46) (mean age, 37.2 ± 11.2 years; males, 54.1%; disease duration, 48 months [interquartile range (IQR), 24-90 months]; pancolitis, 26.1%; SCCAI, 5 [IQR, 4-6]; UCEIS, 4 [IQR, 3-5]). Clinical response (57.1% vs 28.3%; odds ratio [OR], 3.4; 95% confidence interval [CI], 1.4-7.9; P = .01), remission (53.1% vs 28.3%; OR, 2.9; 95% CI, 1.2-6.7; P = .02), and proportion of patients with fecal calprotectin (FCP) <150 µg/g (30.6% vs 6.5%; OR, 6.3; 95% CI, 1.7-23.6; P = .003) were significantly higher in CW. The relative abundance of bacterial taxa that had a significant or trend towards negative correlation with SCCAI, UCEIS, or FCP increased at 8 weeks in CW, and this effect was independent of disease activity and dietary fiber. Adverse events were comparable, and no patient developed hyperkalemia. CONCLUSIONS: CW was more effective than placebo for induction of clinical remission in patients with mild to moderate UC. The trial was prospectively registered on Clinical Trials Registry of India (ctri.nic.in, Number: CTRI/2019/03/01827).


Subject(s)
Cocos , Colitis, Ulcerative , Humans , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/therapy , Male , Female , Double-Blind Method , Adult , Middle Aged , Treatment Outcome , Placebos/administration & dosage , Young Adult , Gastrointestinal Microbiome , Aged , Remission Induction , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/administration & dosage , Severity of Illness Index
5.
PLoS Pathog ; 18(4): e1010465, 2022 04.
Article in English | MEDLINE | ID: mdl-35482816

ABSTRACT

Although efficacious vaccines have significantly reduced the morbidity and mortality of COVID-19, there remains an unmet medical need for treatment options, which monoclonal antibodies (mAbs) can potentially fill. This unmet need is exacerbated by the emergence and spread of SARS-CoV-2 variants of concern (VOCs) that have shown some resistance to vaccine responses. Here we report the isolation of five neutralizing mAbs from an Indian convalescent donor, out of which two (THSC20.HVTR04 and THSC20.HVTR26) showed potent neutralization of SARS-CoV-2 VOCs at picomolar concentrations, including the Delta variant (B.1.617.2). One of these (THSC20.HVTR26) also retained activity against the Omicron variant. These two mAbs target non-overlapping epitopes on the receptor-binding domain (RBD) of the spike protein and prevent virus attachment to its host receptor, human angiotensin converting enzyme-2 (hACE2). Furthermore, the mAb cocktail demonstrated protection against the Delta variant at low antibody doses when passively administered in the K18 hACE2 transgenic mice model, highlighting their potential as a cocktail for prophylactic and therapeutic applications. Developing the capacity to rapidly discover and develop mAbs effective against highly transmissible pathogens like coronaviruses at a local level, especially in a low- and middle-income country (LMIC) such as India, will enable prompt responses to future pandemics as an important component of global pandemic preparedness.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , Mice , Spike Glycoprotein, Coronavirus
6.
PLoS Pathog ; 18(12): e1010994, 2022 12.
Article in English | MEDLINE | ID: mdl-36508467

ABSTRACT

The emergence of new variants of SARS-CoV-2 necessitates unremitting efforts to discover novel therapeutic monoclonal antibodies (mAbs). Here, we report an extremely potent mAb named P4A2 that can neutralize all the circulating variants of concern (VOCs) with high efficiency, including the highly transmissible Omicron. The crystal structure of the P4A2 Fab:RBD complex revealed that the residues of the RBD that interact with P4A2 are a part of the ACE2-receptor-binding motif and are not mutated in any of the VOCs. The pan coronavirus pseudotyped neutralization assay confirmed that the P4A2 mAb is specific for SARS-CoV-2 and its VOCs. Passive administration of P4A2 to K18-hACE2 transgenic mice conferred protection, both prophylactically and therapeutically, against challenge with VOCs. Overall, our data shows that, the P4A2 mAb has immense therapeutic potential to neutralize the current circulating VOCs. Due to the overlap between the P4A2 epitope and ACE2 binding site on spike-RBD, P4A2 may also be highly effective against a number of future variants.


Subject(s)
Angiotensin-Converting Enzyme 2 , Antibodies, Neutralizing , COVID-19 , SARS-CoV-2 , Animals , Humans , Mice , Angiotensin-Converting Enzyme 2/chemistry , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , COVID-19/immunology , COVID-19/therapy , Mice, Transgenic , Neutralization Tests , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/genetics
7.
Nat Immunol ; 13(10): 991-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22961052

ABSTRACT

Interleukin 17 (IL-17)-producing helper T cells (T(H)17 cells) are often present at the sites of tissue inflammation in autoimmune diseases, which has led to the conclusion that T(H)17 cells are main drivers of autoimmune tissue injury. However, not all T(H)17 cells are pathogenic; in fact, T(H)17 cells generated with transforming growth factor-ß1 (TGF-ß1) and IL-6 produce IL-17 but do not readily induce autoimmune disease without further exposure to IL-23. Here we found that the production of TGF-ß3 by developing T(H)17 cells was dependent on IL-23, which together with IL-6 induced very pathogenic T(H)17 cells. Moreover, TGF-ß3-induced T(H)17 cells were functionally and molecularly distinct from TGF-ß1-induced T(H)17 cells and had a molecular signature that defined pathogenic effector T(H)17 cells in autoimmune disease.


Subject(s)
Autoimmune Diseases/immunology , Interleukin-17/biosynthesis , Th17 Cells/immunology , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta3/immunology , Animals , Cell Differentiation/immunology , Humans , Inflammation/immunology , Interleukin-23/immunology , Interleukin-6/immunology , Mice , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta3/metabolism
8.
Scand J Immunol ; 99(2): e13345, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38441373

ABSTRACT

COVID-19 disease has plagued the world economy and affected the overall well-being and life of most of the people. Natural infection as well as vaccination leads to the development of an immune response against the pathogen. This involves the production of antibodies, which can neutralize the virus during future challenges. In addition, the development of cellular immune memory with memory B and T cells provides long-lasting protection. The longevity of the immune response has been a subject of intensive research in this field. The extent of immunity conferred by different forms of vaccination or natural infections remained debatable for long. Hence, understanding the effectiveness of these responses among different groups of people can assist government organizations in making informed policy decisions. In this article, based on the publicly available data, we have reviewed the memory response generated by some of the vaccines against SARS-CoV-2 and its variants, particularly B cell memory in different groups of individuals.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , COVID-19/prevention & control , COVID-19 Vaccines , Antibodies , Immunologic Memory
9.
Semin Cancer Biol ; 80: 87-106, 2022 05.
Article in English | MEDLINE | ID: mdl-32068087

ABSTRACT

Plant lectins, a natural source of glycans with a therapeutic potential may lead to the discovery of new targeted therapies. Glycans extracted from plant lectins are known to act as ligands for C-type lectin receptors (CLRs) that are primarily present on immune cells. Plant-derived glycosylated lectins offer diversity in their N-linked oligosaccharide structures that can serve as a unique source of homogenous and heterogenous glycans. Among the plant lectins-derived glycan motifs, Man9GlcNAc2Asn exhibits high-affinity interactions with CLRs that may resemble glycan motifs of pathogens. Thus, such glycan domains when presented along with antigens complexed with a nanocarrier of choice may bewilder the immune cells and direct antigen cross-presentation - a cytotoxic T lymphocyte immune response mediated by CD8+ T cells. Glycan structure analysis has attracted considerable interest as glycans are looked upon as better therapeutic alternatives than monoclonal antibodies due to their cost-effectiveness, reduced toxicity and side effects, and high specificity. Furthermore, this approach will be useful to understand whether the multivalent glycan presentation on the surface of nanocarriers can overcome the low-affinity lectin-ligand interaction and thereby modulation of CLR-dependent immune response. Besides this, understanding how the heterogeneity of glycan structure impacts the antigen cross-presentation is pivotal to develop alternative targeted therapies. In the present review, we discuss the findings on structural analysis of glycans from natural lectins performed using GlycanBuilder2 - a software tool based on a thorough literature review of natural lectins. Additionally, we discuss how multiple parameters like the orientation of glycan ligands, ligand density, simultaneous targeting of multiple CLRs and design of antigen delivery nanocarriers may influence the CLR targeting efficacy. Integrating this information will eventually set the ground for new generation immunotherapeutic vaccine design for the treatment of various human malignancies.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Antigen Presentation , Dendritic Cells , Humans , Immunotherapy , Lectins, C-Type/chemistry , Ligands , Neoplasms/therapy , Plant Lectins , Polysaccharides/chemistry
10.
Med Microbiol Immunol ; 212(1): 103-122, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36583790

ABSTRACT

The SARS-CoV-2 virus has been rapidly evolving over the time and the genetic variation has led to the generation of Variants of Concerns (VoC), which have shown increased fitness. These VoC viruses contain the key mutations in the spike protein which have allowed better survival and evasion of host defense mechanisms. The D614G mutation in the spike domain is found in the majority of VoC; additionally, the P681R/H mutation at the S1/S2 furin cleavage site junction is also found to be highly conserved in major VoCs; Alpha, Delta, Omicron, and its' current variants. The impact of these genetic alterations of the SARS-CoV-2 VoCs on the host cell entry, transmissibility, and infectivity has not been clearly identified. In our study, Delta and D614G + P681R synthetic double mutant pseudoviruses showed a significant increase in the cell entry, cell-to-cell fusion and infectivity. In contrast, the Omicron and P681H synthetic single mutant pseudoviruses showed TMPRSS2 independent cell entry, less fusion and infectivity as compared to Delta and D614G + P681R double mutants. Addition of exogenous trypsin further enhanced fusion in Delta viruses as compared to Omicron. Furthermore, Delta viruses showed susceptibility to both E64d and Camostat mesylate inhibitors suggesting, that the Delta virus could exploit both endosomal and TMPRSS2 dependent entry pathways as compared to the Omicron virus. Taken together, these results indicate that the D614G and P681R/H mutations in the spike protein are pivotal which might be favoring the VoC replication in different host compartments, and thus allowing a balance of mutation vs selection for better long-term adaptation.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Mutation
11.
J Immunol ; 206(7): 1540-1548, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33648937

ABSTRACT

IL-17A and IL-22 derived from Th17 cells play a significant role in mucosal immunity and inflammation. TGF-ß and IL-6 promote Th17 differentiation; however, these cytokines have multiple targets. The identification and screening of additional molecules that regulate IL-17A and IL-22 responses in certain inflammatory conditions is of great clinical significance. In this study, we show that CDDO-Im, a specific Nrf2 activator, promotes IL-17A and IL-22 responses in murine Th17 cells. In contrast, CDDO-Im inhibits IL-17A response in multiple sclerosis patient-derived PBMCs. However, Nrf2 specifically regulates IL-22 response in vivo. Nrf2 acts through the regulation of antioxidant response element (ARE) binding motifs in target genes to induce or repress transcription. Promoter analysis revealed that Il17a, Rorc, and Ahr genes have several ARE motifs. We showed that Nrf2 bound to ARE repressor (ARE-R2) of Rorc and inhibited Rorc-dependent IL-17A transactivation. The luciferase reporter assay data showed that CDDO-Im regulated Ahr promoter activity. Chromatin immunoprecipitation quantitative PCR data showed that Nrf2 bound to ARE of AhR. Finally, we confirmed that the CDDO-Im-mediated induction of IL-22 production in CD4+ T cells was abrogated in CD4-specific Ahr knockout mice (AhrCD4 ). CH-223191, a specific AhR antagonist, inhibits CDDO-Im-induced IL-22 production in CD4+ T cells, which further confirmed the AhR-dependent regulation. Collectively, our data showed that Nrf2 via AhR pathways regulated IL-22 response in CD4+ T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interleukins/metabolism , Multiple Sclerosis/immunology , NF-E2-Related Factor 2/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Th17 Cells/immunology , Animals , Azo Compounds/metabolism , Gene Expression Regulation , Humans , Imidazoles/metabolism , Interleukin-17/genetics , Interleukin-17/metabolism , Lymphocyte Activation , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Oleanolic Acid/analogs & derivatives , Oleanolic Acid/metabolism , Promoter Regions, Genetic/genetics , Pyrazoles/metabolism , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Interleukin-22
12.
J Biol Chem ; 295(42): 14352-14366, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32817314

ABSTRACT

One strategy for the development of a next generation influenza vaccine centers upon using conserved domains of the virus to induce broader and long-lasting immune responses. The production of artificial proteins by mimicking native-like structures has shown to be a promising approach for vaccine design against diverse enveloped viruses. The amino terminus of influenza A virus matrix 2 ectodomain (M2e) is highly conserved among influenza subtypes, and previous studies have shown M2e-based vaccines are strongly immunogenic, making it an attractive target for further exploration. We hypothesized that stabilizing M2e protein in the mammalian system might influence the immunogenicity of M2e with the added advantage to robustly produce the large scale of proteins with native-like fold and hence can act as an efficient vaccine candidate. In this study, we created an engineered construct in which the amino terminus of M2e is linked to the tetramerizing domain tGCN4, expressed the construct in a mammalian system, and tested for immunogenicity in BALB/c mice. We have also constructed a stand-alone M2e construct (without tGCN4) and compared the protein expressed in mammalian cells and in Escherichia coli using in vitro and in vivo methods. The mammalian-expressed protein was found to be more stable, more antigenic than the E. coli protein, and form higher-order oligomers. In an intramuscular protein priming and boosting regimen in mice, these proteins induced high titers of antibodies and elicited a mixed Th1/Th2 response. These results highlight the mammalian-expressed M2e soluble proteins as a promising vaccine development platform.


Subject(s)
Influenza A Virus, H1N1 Subtype/metabolism , Viral Matrix Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Escherichia coli/metabolism , HEK293 Cells , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Influenza Vaccines/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Protein Domains , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
13.
Arch Biochem Biophys ; 713: 109059, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34673001

ABSTRACT

Approved mAbs that block the protein-protein interaction (PPI) interface of the PD-1/PD-L1 immune checkpoint axis have led to significant improvements in cancer treatment. Despite having drawbacks of mAbs only few a compounds are reported till date against this axis. Inhibiting PPIs using small molecules has emerged as a significant therapeutic opportunity, demanding for the identification of drug-like molecules at an accelerated pace under the hit-to-lead campaigns. Due to the PD-L1's cross-talk with PD-1/CD80 and its overexpression on cancer cells, as well as the availability of its crystal structures with small molecules, it is an enticing therapeutic target for structure-assisted small molecule design. Furthermore, the selection of chemical databases enriched with focused designing for PPI interfaces is crucial. Therefore, in this study we have utilized the Asinex signature library for structure-assisted virtual screening to find the potential PD-L1 inhibitors by targeting the cryptic PD-L1 interface, followed by induced fit docking for pose refinements in the pocket. The obtained hits were then subjected to interaction fingerprinting and ligand-based drug-likeness investigations in order to evaluate and analyze their drug-like qualities (ADME). Twelve compounds qualified for molecular dynamics simulations, followed by thermodynamic calculations for evaluation of their stability and energetics inside the pocket. Two novel compounds with different chemical moieties have been identified that are consistent throughout the simulation, mimicking the interactions and binding energies with BMS-1166. These compounds appear as potential therapeutic candidates to be explored experimentally, thereby paving the way for the development of novel leads as immunomodulators.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Immune Checkpoint Inhibitors/metabolism , Small Molecule Libraries/metabolism , Amino Acid Sequence , B7-H1 Antigen/chemistry , Binding Sites , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Thermodynamics
14.
J Chem Inf Model ; 61(1): 358-384, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33433201

ABSTRACT

The dynamics and plasticity of the PD-1/PD-L1 axis are the bottlenecks for the discovery of small-molecule antagonists to perturb this interaction interface significantly. Understanding the process of this protein-protein interaction (PPI) is of fundamental biological interest in structure-based drug designing. Food and Drug Administration (FDA)-approved anti-PD-1 monoclonal antibodies (mAbs) are the first-in-class with distinct binding modes to access this axis clinically; however, their mechanistic aspects remain elusive. Here, we have unveiled the interactive interfaces with PD-L1 and mAbs to investigate the native plasticity of PD-1 at global (structural and dynamical) and local (residue side-chain orientations) levels. We found that the structural stability and coordinated Cα movements are increased in the presence of PD-1's binding partners. The rigorous analysis of these PPIs using computational biophysical approaches revealed PD-1's intrinsic plasticity, its concerted loops' movement (BC, FG, and CC'), distal side-chain motions, and the thermodynamic landscape, which are perturbed remarkably from its unbound to bound states. Based on intra-/inter-residues' contact networks and energetics, the hot-spots have been identified that were found to be essential to arrest the dynamical motions of PD-1 significantly for the rational design of therapeutic agents by mimicking the mAbs mechanism.


Subject(s)
Programmed Cell Death 1 Receptor , Models, Molecular , Protein Binding , Protein Conformation
15.
World J Microbiol Biotechnol ; 37(4): 67, 2021 Mar 22.
Article in English | MEDLINE | ID: mdl-33748926

ABSTRACT

Infectious diseases are one of the main grounds of death and disabilities in human beings globally. Lack of effective treatment and immunization for many deadly infectious diseases and emerging drug resistance in pathogens underlines the need to either develop new vaccines or sufficiently improve the effectiveness of currently available drugs and vaccines. In this review, we discuss the application of advanced tools like bioinformatics, genomics, proteomics and associated techniques for a rational vaccine design.


Subject(s)
Bacterial Vaccines , Drug Development , Bacteria , Computational Biology , Genomics , Humans , Immunization , Proteomics
16.
Environ Dev Sustain ; 23(6): 8147-8160, 2021.
Article in English | MEDLINE | ID: mdl-32994752

ABSTRACT

The novel coronavirus disease is known as COVID-19, which is declared as a pandemic by the World Health Organization during March 2020. In this study, the COVID-19 connection with various weather parameters like temperature, wind speed, and relative humidity is investigated and the future scenario of COVID-19 is predicted based on the Gaussian model (GM). This study is conducted in Delhi, the capital city of India, during the lowest mobility rate due to strict lockdown nationwide for about two months from March 15 to May 17, 2020. Spearman correlation is applied to obtain the interconnection of COVID-19 cases with weather parameters. Based on statistical analysis, this has been observed that the temperature parameter shows a significant positive trend during the period of study. The number of confirmed cases of COVID-19 is fitted with respect to the number of days by using the Gaussian curve and it is estimated on the basis of the model that maximum cases will go up to 123,886 in number. The maximum number of cases will be observed during the range of 166 ± 36 days. It is also estimated by using the width of the fitted GM that it will take minimum of 10 months for the complete recovery from COVID-19. Additionally, the linear regression technique is used to find the trend of COVID-19 cases with temperature and it is estimated that with an increase in temperature by 1 °C, 30 new COVID-19 cases on daily basis will be expected to observe. This study is believed to be a preliminary study and to better understand the concrete relationship of coronavirus, at least one complete cycle is essential to investigate. The laboratory-based study is essential to be done to support the present field-based study. Henceforth, based on preliminary studies, significant inputs are put forth to the research community and government to formulate thoughtful strategies like medical facilities such as ventilators, beds, testing centers, quarantine centers, etc., to curb the effects of COVID-19.

17.
Nat Immunol ; 9(12): 1347-55, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18997793

ABSTRACT

Transcription factor Foxp3 is critical for generating regulatory T cells (T(reg) cells). Transforming growth factor-beta (TGF-beta) induces Foxp3 and suppressive T(reg) cells from naive T cells, whereas interleukin 6 (IL-6) inhibits the generation of inducible T(reg) cells. Here we show that IL-4 blocked the generation of TGF-beta-induced Foxp3(+) T(reg) cells and instead induced a population of T helper cells that produced IL-9 and IL-10. The IL-9(+)IL-10(+) T cells demonstrated no regulatory properties despite producing abundant IL-10. Adoptive transfer of IL-9(+)IL-10(+) T cells into recombination-activating gene 1-deficient mice induced colitis and peripheral neuritis, the severity of which was aggravated if the IL-9(+)IL-10(+) T cells were transferred with CD45RB(hi) CD4(+) effector T cells. Thus IL-9(+)IL-10(+) T cells lack suppressive function and constitute a distinct population of helper-effector T cells that promote tissue inflammation.


Subject(s)
Forkhead Transcription Factors/immunology , Interleukin-4/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Forkhead Transcription Factors/metabolism , GATA3 Transcription Factor/immunology , GATA3 Transcription Factor/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-4/metabolism , Interleukin-9/immunology , Interleukin-9/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Regulatory/cytology , Transforming Growth Factor beta/metabolism
18.
Immunity ; 35(2): 156-7, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21867924

ABSTRACT

Neural progenitor cells (NPCs) can repair damaged myelin in neurodegenerative diseases. In this issue of Immunity, Cao et al. (2011) report that NPCs also produce the cytokine LIF, which suppresses Th17 cell-driven inflammation and autoimmunity by upregulating the protein Socs3.

19.
Nat Immunol ; 8(12): 1380-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17994022

ABSTRACT

Regulatory T cells (T(reg) cells) expressing the transcription factor Foxp3 are key in maintaining the balance of immune homeostasis. However, distinct induced T regulatory type 1 (Tr1) cells that lack Foxp3 expression also regulate T cell function, mainly by producing the immunosuppressive cytokine interleukin 10 (IL-10). However, the factors required for the induction of IL-10-producing suppressive T cells are not fully understood. Here we demonstrate that dendritic cells modified by T(reg) cells induced the generation of IL-10-producing Tr1 cells. The differentiation of naive CD4+ T cells into IL-10-producing cells was mediated by IL-27 produced by the T(reg) cell-modified dendritic cells, and transforming growth factor-beta amplified the generation of induced IL-10+ Tr1 cells by IL-27. Thus, IL-27 and transforming growth factor-beta promote the generation of IL-10-producing Tr1 cells.


Subject(s)
Interleukin-10/physiology , Interleukin-17/physiology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/biosynthesis , Animals , Humans , Interleukin-10/immunology , Interleukin-17/metabolism , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology
20.
Nature ; 496(7446): 461-8, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23467089

ABSTRACT

Despite their importance, the molecular circuits that control the differentiation of naive T cells remain largely unknown. Recent studies that reconstructed regulatory networks in mammalian cells have focused on short-term responses and relied on perturbation-based approaches that cannot be readily applied to primary T cells. Here we combine transcriptional profiling at high temporal resolution, novel computational algorithms, and innovative nanowire-based perturbation tools to systematically derive and experimentally validate a model of the dynamic regulatory network that controls the differentiation of mouse TH17 cells, a proinflammatory T-cell subset that has been implicated in the pathogenesis of multiple autoimmune diseases. The TH17 transcriptional network consists of two self-reinforcing, but mutually antagonistic, modules, with 12 novel regulators, the coupled action of which may be essential for maintaining the balance between TH17 and other CD4(+) T-cell subsets. Our study identifies and validates 39 regulatory factors, embeds them within a comprehensive temporal network and reveals its organizational principles; it also highlights novel drug targets for controlling TH17 cell differentiation.


Subject(s)
Cell Differentiation/genetics , Gene Regulatory Networks/genetics , Th17 Cells/cytology , Th17 Cells/metabolism , Animals , Cells, Cultured , DNA/genetics , DNA/metabolism , Forkhead Transcription Factors/metabolism , Gene Knockdown Techniques , Genome/genetics , Interferon-gamma/biosynthesis , Interleukin-2/genetics , Mice , Mice, Inbred C57BL , Nanowires , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Silicon , Th17 Cells/immunology , Time Factors , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL