1
|
Nanda S, Zafar MA, Lamba T, Malik JA, Khan MA, Bhardwaj P, Bisht B, Ghadi R, Kaur G, Bhalla V, Owais M, Jain S, Sehrawat S, Agrewala JN. A novel strategy to elicit enduring anti-morphine immunity and relief from addiction by targeting Acr1 protein nano vaccine through TLR-2 to dendritic cells. Int J Biol Macromol 2024; 274:133188. [PMID: 38880456 DOI: 10.1016/j.ijbiomac.2024.133188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Morphine addiction poses a significant challenge to global healthcare. Current opioid substitution therapies, such as buprenorphine, naloxone and methadone are effective but often lead to dependence. Thus, exploring alternative treatments for opioid addiction is crucial. We have developed a novel vaccine that presents morphine and Pam3Cys (a TLR-2 agonist) on the surface of Acr1 nanoparticles. This vaccine has self-adjuvant properties and targets TLR-2 receptors on antigen-presenting cells, particularly dendritic cells. Our vaccination strategy promotes the proliferation and differentiation of morphine-specific B-cells and Acr1-reactive CD4 T-cells. Additionally, the vaccine elicited the production of high-affinity anti-morphine antibodies, effectively eliminating morphine from the bloodstream and brain in mice. It also reduced the expression of addiction-associated μ-opioid receptor and dopamine genes. The significant increase in memory CD4 T-cells and B-cells indicates the vaccine's ability to induce long-lasting immunity against morphine. This vaccine holds promise as a prophylactic measure against morphine addiction.
Collapse
Affiliation(s)
- Sidhanta Nanda
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Mohammad Adeel Zafar
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Taruna Lamba
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Jonaid Ahmad Malik
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Mohammad Affan Khan
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Priya Bhardwaj
- CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Bhawana Bisht
- CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Rohan Ghadi
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Mohali, India
| | - Gurpreet Kaur
- Department of Biotechnology, Chandigarh Group of Colleges, Mohali, India
| | | | - Mohammad Owais
- Department of Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Mohali, India
| | - Sharvan Sehrawat
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Javed N Agrewala
- Immunology Laboratory, Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India.
| |
Collapse
|
2
|
Ayodele S, Kumar P, van Eyk A, Choonara YE. Advances in immunomodulatory strategies for host-directed therapies in combating tuberculosis. Biomed Pharmacother 2023; 162:114588. [PMID: 36989709 DOI: 10.1016/j.biopha.2023.114588] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/14/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Tuberculosis (TB) maintains its infamous status regarding its detrimental effect on global health, causing the highest mortality by a single infectious agent. The presence of resistance and immune compromising disease favours the disease in maintaining its footing in the health care burden despite various anti-TB drugs used to fight it. Main factors contributing to resistance and difficulty in treating disease include prolonged treatment duration (at least 6 months) and severe toxicity, which further leads to patient non-compliance, and thus a ripple effect leading to therapeutic non-efficacy. The efficacy of new regimens demonstrates that targeting host factors concomitantly with the Mycobacterium tuberculosis (M.tb) strain is urgently required. Due to the huge expenses and time required of up to 20 years for new drug research and development, drug repurposing may be the most economical, circumspective, and conveniently faster journey to embark on. Host-directed therapy (HDT) will dampen the burden of the disease by acting as an immunomodulator, allowing it to defend the body against antibiotic-resistant pathogens whilst minimizing the possibility of developing new resistance to susceptible drugs. Repurposed drugs in TB act as host-directed therapies, acclimatizing the host immune cell to the presence of TB, improving its antimicrobial activity and time taken to get rid of the disease, whilst minimizing inflammation and tissue damage. In this review, we, therefore, explore possible immunomodulatory targets, HDT immunomodulatory agents, and their ability to improve clinical outcomes whilst minimizing the risk of drug resistance, through various pathway targeting and treatment duration reduction.
Collapse
|
3
|
Hasankhani A, Bahrami A, Mackie S, Maghsoodi S, Alawamleh HSK, Sheybani N, Safarpoor Dehkordi F, Rajabi F, Javanmard G, Khadem H, Barkema HW, De Donato M. In-depth systems biological evaluation of bovine alveolar macrophages suggests novel insights into molecular mechanisms underlying Mycobacterium bovis infection. Front Microbiol 2022; 13:1041314. [PMID: 36532492 PMCID: PMC9748370 DOI: 10.3389/fmicb.2022.1041314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/04/2022] [Indexed: 08/26/2023] Open
Abstract
Objective Bovine tuberculosis (bTB) is a chronic respiratory infectious disease of domestic livestock caused by intracellular Mycobacterium bovis infection, which causes ~$3 billion in annual losses to global agriculture. Providing novel tools for bTB managements requires a comprehensive understanding of the molecular regulatory mechanisms underlying the M. bovis infection. Nevertheless, a combination of different bioinformatics and systems biology methods was used in this study in order to clearly understand the molecular regulatory mechanisms of bTB, especially the immunomodulatory mechanisms of M. bovis infection. Methods RNA-seq data were retrieved and processed from 78 (39 non-infected control vs. 39 M. bovis-infected samples) bovine alveolar macrophages (bAMs). Next, weighted gene co-expression network analysis (WGCNA) was performed to identify the co-expression modules in non-infected control bAMs as reference set. The WGCNA module preservation approach was then used to identify non-preserved modules between non-infected controls and M. bovis-infected samples (test set). Additionally, functional enrichment analysis was used to investigate the biological behavior of the non-preserved modules and to identify bTB-specific non-preserved modules. Co-expressed hub genes were identified based on module membership (MM) criteria of WGCNA in the non-preserved modules and then integrated with protein-protein interaction (PPI) networks to identify co-expressed hub genes/transcription factors (TFs) with the highest maximal clique centrality (MCC) score (hub-central genes). Results As result, WGCNA analysis led to the identification of 21 modules in the non-infected control bAMs (reference set), among which the topological properties of 14 modules were altered in the M. bovis-infected bAMs (test set). Interestingly, 7 of the 14 non-preserved modules were directly related to the molecular mechanisms underlying the host immune response, immunosuppressive mechanisms of M. bovis, and bTB development. Moreover, among the co-expressed hub genes and TFs of the bTB-specific non-preserved modules, 260 genes/TFs had double centrality in both co-expression and PPI networks and played a crucial role in bAMs-M. bovis interactions. Some of these hub-central genes/TFs, including PSMC4, SRC, BCL2L1, VPS11, MDM2, IRF1, CDKN1A, NLRP3, TLR2, MMP9, ZAP70, LCK, TNF, CCL4, MMP1, CTLA4, ITK, IL6, IL1A, IL1B, CCL20, CD3E, NFKB1, EDN1, STAT1, TIMP1, PTGS2, TNFAIP3, BIRC3, MAPK8, VEGFA, VPS18, ICAM1, TBK1, CTSS, IL10, ACAA1, VPS33B, and HIF1A, had potential targets for inducing immunomodulatory mechanisms by M. bovis to evade the host defense response. Conclusion The present study provides an in-depth insight into the molecular regulatory mechanisms behind M. bovis infection through biological investigation of the candidate non-preserved modules directly related to bTB development. Furthermore, several hub-central genes/TFs were identified that were significant in determining the fate of M. bovis infection and could be promising targets for developing novel anti-bTB therapies and diagnosis strategies.
Collapse
Affiliation(s)
- Aliakbar Hasankhani
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
- Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Shayan Mackie
- Faculty of Science, Earth Sciences Building, University of British Columbia, Vancouver, BC, Canada
| | - Sairan Maghsoodi
- Faculty of Paramedical Sciences, Kurdistan University of Medical Sciences, Kurdistan, Iran
| | - Heba Saed Kariem Alawamleh
- Department of Basic Scientific Sciences, AL-Balqa Applied University, AL-Huson University College, AL-Huson, Jordan
| | - Negin Sheybani
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | - Farhad Safarpoor Dehkordi
- Halal Research Center of IRI, FDA, Tehran, Iran
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Fatemeh Rajabi
- Department of Agronomy and Plant Breeding, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Ghazaleh Javanmard
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Hosein Khadem
- Department of Agronomy and Plant Breeding, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Herman W. Barkema
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Marcos De Donato
- Regional Department of Bioengineering, Tecnológico de Monterrey, Monterrey, Mexico
| |
Collapse
|
4
|
Bartens MC, Gibson AJ, Etherington GJ, Di Palma F, Holder A, Werling D, Willcocks S. Single Nucleotide Polymorphisms in the Bovine TLR2 Extracellular Domain Contribute to Breed and Species-Specific Innate Immune Functionality. Front Immunol 2022; 12:764390. [PMID: 35003078 PMCID: PMC8732954 DOI: 10.3389/fimmu.2021.764390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/26/2021] [Indexed: 11/13/2022] Open
Abstract
Recent evidence suggests that several cattle breeds may be more resistant to infection with the zoonotic pathogen Mycobacterium bovis. Our data presented here suggests that the response to mycobacterial antigens varies in macrophages generated from Brown Swiss (BS) and Holstein Friesian (HF) cattle, two breeds belonging to the Bos taurus family. Whole genome sequencing of the Brown Swiss genome identified several potential candidate genes, in particular Toll-like Receptor-2 (TLR2), a pattern recognition receptor (PRR) that has previously been described to be involved in mycobacterial recognition. Further investigation revealed single nucleotide polymorphisms (SNP) in TLR2 that were identified between DNA isolated from cells of BS and HF cows. Interestingly, one specific SNP, H326Q, showed a different genotype frequency in two cattle subspecies, Bos (B.) taurus and Bos indicus. Cloning of the TLR2 gene and subsequent gene-reporter and chemokine assays revealed that this SNP, present in BS and Bos indicus breeds, resulted in a significantly higher response to mycobacterial antigens as well as tri-acylated lipopeptide ligands in general. Comparing wild-type and H326Q containing TLR2 responses, wild-type bovine TLR2 response showed clear, diminished mycobacterial antigen responses compared to human TLR2, however bovine TLR2 responses containing H326Q were found to be partially recovered compared to human TLR2. The creation of human:bovine TLR2 chimeras increased the response to mycobacterial antigens compared to the full-length bovine TLR2, but significantly reduced the response compared to the full-length human TLR2. Thus, our data, not only present evidence that TLR2 is a major PRR in the mammalian species-specific response to mycobacterial antigens, but furthermore, that there are clear differences between the response seen in different cattle breeds, which may contribute to their enhanced or reduced susceptibility to mycobacterial infection.
Collapse
Affiliation(s)
- Marie-Christine Bartens
- Molecular Immunology Group, Centre for Vaccinology and Regenerative Medicine, Department of Pathobiology and Population Science, Royal Veterinary College, Hatfield, United Kingdom.,Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Amanda J Gibson
- Molecular Immunology Group, Centre for Vaccinology and Regenerative Medicine, Department of Pathobiology and Population Science, Royal Veterinary College, Hatfield, United Kingdom
| | | | - Federica Di Palma
- Genome British Columbia, Vancouver, BC, Canada.,Department of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| | - Angela Holder
- Molecular Immunology Group, Centre for Vaccinology and Regenerative Medicine, Department of Pathobiology and Population Science, Royal Veterinary College, Hatfield, United Kingdom
| | - Dirk Werling
- Molecular Immunology Group, Centre for Vaccinology and Regenerative Medicine, Department of Pathobiology and Population Science, Royal Veterinary College, Hatfield, United Kingdom
| | - Sam Willcocks
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
5
|
Kaur H, Ali SA. Probiotics and gut microbiota: mechanistic insights into gut immune homeostasis through TLR pathway regulation. Food Funct 2022; 13:7423-7447. [DOI: 10.1039/d2fo00911k] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Consumption of probiotics as a useful functional food improves the host's wellbeing, and, when paired with prebiotics (indigestible dietary fibre/carbohydrate), often benefits the host through anaerobic fermentation.
Collapse
Affiliation(s)
- Harpreet Kaur
- Animal Biochemistry Division, ICAR-NDRI, 132001, India
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
6
|
Blanco FC, Gravisaco MJ, Bigi MM, García EA, Marquez C, McNeil M, Jackson M, Bigi F. Identifying Bacterial and Host Factors Involved in the Interaction of Mycobacterium bovis with the Bovine Innate Immune Cells. Front Immunol 2021; 12:674643. [PMID: 34335572 PMCID: PMC8319915 DOI: 10.3389/fimmu.2021.674643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
Bovine tuberculosis is an important animal and zoonotic disease caused by Mycobacterium bovis. The innate immune response is the first line of defense against pathogens and is also crucial for the development of an efficient adaptive immune response. In this study we used an in vitro co-culture model of antigen presenting cells (APC) and autologous lymphocytes derived from peripheral blood mononuclear cells to identify the cell populations and immune mediators that participate in the development of an efficient innate response capable of controlling the intracellular replication of M. bovis. After M. bovis infection, bovine immune cell cultures displayed upregulated levels of iNOS, IL-22 and IFN-γ and the induction of the innate immune response was dependent on the presence of differentiated APC. Among the analyzed M. bovis isolates, only a live virulent M. bovis isolate induced an efficient innate immune response, which was increased upon stimulation of cell co-cultures with the M. bovis culture supernatant. Moreover, we demonstrated that an allelic variation of the early secreted protein ESAT-6 (ESAT6 T63A) expressed in the virulent strain is involved in this increased innate immune response. These results highlight the relevance of the compounds secreted by live M. bovis as well as the variability among the assessed M. bovis strains to induce an efficient innate immune response.
Collapse
Affiliation(s)
- Federico Carlos Blanco
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
- (Consejo Nacional de Investigaciones Científicas y Tecnológicas) National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - María José Gravisaco
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
| | - María Mercedes Bigi
- (Facultad de Agronomía, Universidad de Buenos Aires) School of Agronomy, University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Elizabeth Andrea García
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
| | - Cecilia Marquez
- High Technology Analytical Centre, Laboratory, Buenos Aires, Argentina
| | - Mike McNeil
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Mary Jackson
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Fabiana Bigi
- (Instituto de Biotecnología, Instituto Nacional de Tecnología Agropecuaria) Institute of Biotechnology, National Institute of Agricultural Technology (INTA), Buenos Aires, Argentina
- (Consejo Nacional de Investigaciones Científicas y Tecnológicas) National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
7
|
Sharma A, De Rosa M, Singla N, Singh G, Barnwal RP, Pandey A. Tuberculosis: An Overview of the Immunogenic Response, Disease Progression, and Medicinal Chemistry Efforts in the Last Decade toward the Development of Potential Drugs for Extensively Drug-Resistant Tuberculosis Strains. J Med Chem 2021; 64:4359-4395. [PMID: 33826327 DOI: 10.1021/acs.jmedchem.0c01833] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tuberculosis (TB) is a slow growing, potentially debilitating disease that has plagued humanity for centuries and has claimed numerous lives across the globe. Concerted efforts by researchers have culminated in the development of various strategies to combat this malady. This review aims to raise awareness of the rapidly increasing incidences of multidrug-resistant (MDR) and extensively drug-resistant (XDR) tuberculosis, highlighting the significant modifications that were introduced in the TB treatment regimen over the past decade. A description of the role of pathogen-host immune mechanisms together with strategies for prevention of the disease is discussed. The struggle to develop novel drug therapies has continued in an effort to reduce the treatment duration, improve patient compliance and outcomes, and circumvent TB resistance mechanisms. Herein, we give an overview of the extensive medicinal chemistry efforts made during the past decade toward the discovery of new chemotypes, which are potentially active against TB-resistant strains.
Collapse
Affiliation(s)
- Akanksha Sharma
- Department of Biophysics, Panjab University, Chandigarh 160014, India.,UIPS, Panjab University, Chandigarh 160014, India
| | - Maria De Rosa
- Drug Discovery Unit, Ri.MED Foundation, Palermo 90133, Italy
| | - Neha Singla
- Department of Biophysics, Panjab University, Chandigarh 160014, India
| | - Gurpal Singh
- UIPS, Panjab University, Chandigarh 160014, India
| | - Ravi P Barnwal
- Department of Biophysics, Panjab University, Chandigarh 160014, India
| | - Ankur Pandey
- Department of Chemistry, Panjab University, Chandigarh 160014, India
| |
Collapse
|
8
|
Innate Immune Pattern Recognition Receptors of Mycobacterium tuberculosis: Nature and Consequences for Pathogenesis of Tuberculosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:179-215. [PMID: 34661896 DOI: 10.1007/978-3-030-67452-6_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Innate immunity against Mycobacterium tuberculosis is a critical early response to prevent the establishment of the infection. Despite recent advances in understanding the host-pathogen dialogue in the early stages of tuberculosis (TB), much has yet to be learnt. The nature and consequences of this dialogue ultimately determine the path of infection: namely, either early clearance of M. tuberculosis, or establishment of M. tuberculosis infection leading to active TB disease and/or latent TB infection. On the frontline in innate immunity are pattern recognition receptors (PRRs), with soluble factors (e.g. collectins and complement) and cell surface factors (e.g. Toll-like receptors and other C-type lectin receptors (Dectin 1/2, Nod-like receptors, DC-SIGN, Mincle, mannose receptor, and MCL) that play a central role in recognising M. tuberculosis and facilitating its clearance. However, in a 'double-edged sword' scenario, these factors can also be involved in enhancement of pathogenesis as well. Furthermore, innate immunity is also a critical bridge in establishing the subsequent adaptive immune response, which is also responsible for granuloma formation that cordons off M. tuberculosis infection, establishing latency and acting as a reservoir for bacterial persistence and dissemination of future disease. This chapter discusses the current understanding of pattern recognition of M. tuberculosis by innate immunity and the role this plays in the pathogenesis and protection against TB.
Collapse
|
9
|
Ryu C, Walia A, Ortiz V, Perry C, Woo S, Reeves BC, Sun H, Winkler J, Kanyo JE, Wang W, Vukmirovic M, Ristic N, Stratton EA, Meena SR, Minasyan M, Kurbanov D, Liu X, Lam TT, Farina G, Gomez JL, Gulati M, Herzog EL. Bioactive Plasma Mitochondrial DNA Is Associated With Disease Progression in Scleroderma-Associated Interstitial Lung Disease. Arthritis Rheumatol 2020; 72:1905-1915. [PMID: 32602227 PMCID: PMC8081728 DOI: 10.1002/art.41418] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 06/23/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Systemic sclerosis-associated interstitial lung disease (SSc-ILD) is characterized by variable clinical outcomes, activation of innate immune pattern-recognition receptors (PRRs), and accumulation of α-smooth muscle actin (α-SMA)-expressing myofibroblasts. The aim of this study was to identify an association between these entities and mitochondrial DNA (mtDNA), an endogenous ligand for the intracellular DNA-sensing PRRs Toll-like receptor 9 (TLR-9) and cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS/STING), which has yet to be determined. METHODS Human lung fibroblasts (HLFs) from normal donors and SSc-ILD explants were treated with synthetic CpG DNA and assayed for α-SMA expression and extracellular mtDNA using quantitative polymerase chain reaction for the human MT-ATP6 gene. Plasma MT-ATP6 concentrations were evaluated in 2 independent SSc-ILD cohorts and demographically matched controls. The ability of SSc-ILD and control plasma to induce TLR-9 and cGAS/STING activation was evaluated with commercially available HEK 293 reporter cells. Plasma concentrations of type I interferons (IFNs), interleukin-6 (IL-6), and oxidized DNA were measured using electrochemiluminescence and enzyme-linked immunosorbent assay-based methods. Extracellular vesicles (EVs) precipitated from plasma were evaluated for MT-ATP6 concentrations and proteomics via liquid chromatography mass spectrometry. RESULTS Normal HLFs and SSc-ILD fibroblasts developed increased α-SMA expression and MT-ATP6 release following CpG stimulation. Plasma mtDNA concentrations were increased in the 2 SSc-ILD cohorts, reflective of ventilatory decline, and were positively associated with both TLR-9 and cGAS/STING activation as well as type I IFN and IL-6 expression. Plasma mtDNA was not oxidized and was conveyed by EVs displaying a proteomics profile consistent with a multicellular origin. CONCLUSION These findings demonstrate a previously unrecognized connection between EV-encapsulated mtDNA, clinical outcomes, and intracellular DNA-sensing PRR activation in SSc-ILD. Further study of these interactions could catalyze novel mechanistic and therapeutic insights into SSc-ILD and related disorders.
Collapse
Affiliation(s)
- Changwan Ryu
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Anjali Walia
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Vivian Ortiz
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Carrighan Perry
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Sam Woo
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Benjamin C. Reeves
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Huanxing Sun
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Julia Winkler
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Jean E. Kanyo
- Yale MS & Proteomics Resource, WM Keck Foundation Biotechnology Resource Laboratory, New Haven, CT
| | - Weiwei Wang
- Yale MS & Proteomics Resource, WM Keck Foundation Biotechnology Resource Laboratory, New Haven, CT
| | - Milica Vukmirovic
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Nicholas Ristic
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Eric A. Stratton
- Boston University School of Medicine, Department of Rheumatology
| | - Sita Ram Meena
- Yale University School of Medicine, Department of Cellular and Molecular Physiology
| | - Maksym Minasyan
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Daniel Kurbanov
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Xinran Liu
- Yale University School of Medicine, Department of Cell Biology, Center for Cellular and Molecular Imaging
| | - TuKiet T. Lam
- Yale MS & Proteomics Resource, WM Keck Foundation Biotechnology Resource Laboratory, New Haven, CT
- Yale University School of Medicine, Department of Molecular Biophysics and Biochemistry
| | | | - Jose L. Gomez
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Mridu Gulati
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
| | - Erica L. Herzog
- Yale University School of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine
- Yale University School of Medicine, Department of Pathology
| |
Collapse
|
10
|
Negi S, Pahari S, Bashir H, Agrewala JN. Intestinal microbiota disruption limits the isoniazid mediated clearance of Mycobacterium tuberculosis in mice. Eur J Immunol 2020; 50:1976-1987. [PMID: 32673409 DOI: 10.1002/eji.202048556] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 07/05/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022]
Abstract
Tuberculosis (TB) continues to remain a global threat due to the emergence of drug-resistant Mycobacterium tuberculosis (Mtb) strains and toxicity associated with TB drugs. Intestinal microbiota has been reported to affect the host response to immunotherapy and drugs. However, how it affects the potency of first-line TB drug isoniazid (INH) is largely unknown. Here, we examined the impact of gut microbial dysbiosis on INH efficiency to kill Mtb. In this study, we employed in vivo mouse model, pretreated with broad-spectrum antibiotics (Abx) cocktail to disrupt their intestinal microbial population prior to Mtb infection and subsequent INH therapy. We demonstrated that microbiota disruption results in the impairment of INH-mediated Mtb clearance, and aggravated TB-associated tissue pathology. Further, it suppressed the innate immunity and reduced CD4 T-cell response against Mtb. Interestingly, a distinct shift of gut microbial profile was noted with abundance of Enterococcus and reduction of Lactobacillus and Bifidobacterium population. Our results show that the intestinal microbiota is crucial determinant in efficacy of INH to kill Mtb and impacts the host immune response against infection. This work provides an intriguing insight into the potential links between host gut microbiota and potency of INH.
Collapse
Affiliation(s)
- Shikha Negi
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India.,Present address: Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Susanta Pahari
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India.,Immunology Division, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Hilal Bashir
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed N Agrewala
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India.,Centre for Biomedical Engineering, Indian Institute of Technology-Ropar, Rupnagar, Punjab, India
| |
Collapse
|
11
|
Abreu R, Giri P, Quinn F. Host-Pathogen Interaction as a Novel Target for Host-Directed Therapies in Tuberculosis. Front Immunol 2020; 11:1553. [PMID: 32849525 PMCID: PMC7396704 DOI: 10.3389/fimmu.2020.01553] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 06/12/2020] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB) has been a transmittable human disease for many thousands of years, and M. tuberculosis is again the number one cause of death worldwide due to a single infectious agent. The intense 6- to 10-month process of multi-drug treatment, combined with the adverse side effects that can run the spectrum from gastrointestinal disturbances to liver toxicity or peripheral neuropathy are major obstacles to patient compliance and therapy completion. The consequent increase in multidrug resistant TB (MDR-TB) and extensively drug resistant TB (XDR-TB) cases requires that we increase our arsenal of effective drugs, particularly novel therapeutic approaches. Over the millennia, host and pathogen have evolved mechanisms and relationships that greatly influence the outcome of infection. Understanding these evolutionary interactions and their impact on bacterial clearance or host pathology will lead the way toward rational development of new therapeutics that favor enhancing a host protective response. These host-directed therapies have recently demonstrated promising results against M. tuberculosis, adding to the effectiveness of currently available anti-mycobacterial drugs that directly kill the organism or slow mycobacterial replication. Here we review the host-pathogen interactions during M. tuberculosis infection, describe how M. tuberculosis bacilli modulate and evade the host immune system, and discuss the currently available host-directed therapies that target these bacterial factors. Rather than provide an exhaustive description of M. tuberculosis virulence factors, which falls outside the scope of this review, we will instead focus on the host-pathogen interactions that lead to increased bacterial growth or host immune evasion, and that can be modulated by existing host-directed therapies.
Collapse
Affiliation(s)
| | | | - Fred Quinn
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
| |
Collapse
|
12
|
Kaur D, Arora C, Raghava GPS. A Hybrid Model for Predicting Pattern Recognition Receptors Using Evolutionary Information. Front Immunol 2020; 11:71. [PMID: 32082326 PMCID: PMC7002473 DOI: 10.3389/fimmu.2020.00071] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/13/2020] [Indexed: 12/17/2022] Open
Abstract
This study describes a method developed for predicting pattern recognition receptors (PRRs), which are an integral part of the immune system. The models developed here were trained and evaluated on the largest possible non-redundant PRRs, obtained from PRRDB 2.0, and non-pattern recognition receptors (Non-PRRs), obtained from Swiss-Prot. Firstly, a similarity-based approach using BLAST was used to predict PRRs and got limited success due to a large number of no-hits. Secondly, machine learning-based models were developed using sequence composition and achieved a maximum MCC of 0.63. In addition to this, models were developed using evolutionary information in the form of PSSM composition and achieved maximum MCC value of 0.66. Finally, we developed hybrid models that combined a similarity-based approach using BLAST and machine learning-based models. Our best model, which combined BLAST and PSSM based model, achieved a maximum MCC value of 0.82 with an AUROC value of 0.95, utilizing the potential of both similarity-based search and machine learning techniques. In order to facilitate the scientific community, we also developed a web server "PRRpred" based on the best model developed in this study (http://webs.iiitd.edu.in/raghava/prrpred/).
Collapse
Affiliation(s)
- Dilraj Kaur
- Department of Computational Biology, Indraprastha Institute of Information Technology, New Delhi, India
| | - Chakit Arora
- Department of Computational Biology, Indraprastha Institute of Information Technology, New Delhi, India
| | - Gajendra P S Raghava
- Department of Computational Biology, Indraprastha Institute of Information Technology, New Delhi, India
| |
Collapse
|
13
|
Negi S, Das DK, Pahari S, Nadeem S, Agrewala JN. Potential Role of Gut Microbiota in Induction and Regulation of Innate Immune Memory. Front Immunol 2019; 10:2441. [PMID: 31749793 PMCID: PMC6842962 DOI: 10.3389/fimmu.2019.02441] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/01/2019] [Indexed: 12/14/2022] Open
Abstract
The gut microbiota significantly regulates the development and function of the innate and adaptive immune system. The attribute of immunological memory has long been linked only with adaptive immunity. Recent evidence indicates that memory is also present in the innate immune cells such as monocytes/macrophages and natural killer cells. These cells exhibit pattern recognition receptors (PRRs) that recognize microbe- or pathogen-associated molecular patterns (MAMPs or PAMPs) expressed by the microbes. Interaction between PRRs and MAMPs is quite crucial since it triggers the sequence of signaling events and epigenetic rewiring that not only play a cardinal role in modulating the activation and function of the innate cells but also impart a sense of memory response. We discuss here how gut microbiota can influence the generation of innate memory and functional reprogramming of bone marrow progenitors that helps in protection against infections. This article will broaden our current perspective of association between the gut microbiome and innate memory. In the future, this knowledge may pave avenues for development and designing of novel immunotherapies and vaccination strategies.
Collapse
Affiliation(s)
- Shikha Negi
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India.,Gastroenterology Division, Washington University in St. Louis, St. Louis, MO, United States
| | - Deepjyoti Kumar Das
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Susanta Pahari
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India.,Immunology Division, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Sajid Nadeem
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India.,Department of Microbiology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Javed N Agrewala
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India.,Center for Biomedical Engineering, Indian Institute of Technology-Ropar, Rupnagar, India
| |
Collapse
|
14
|
Pahari S, Negi S, Aqdas M, Arnett E, Schlesinger LS, Agrewala JN. Induction of autophagy through CLEC4E in combination with TLR4: an innovative strategy to restrict the survival of Mycobacterium tuberculosis. Autophagy 2019; 16:1021-1043. [PMID: 31462144 DOI: 10.1080/15548627.2019.1658436] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Host-directed therapies are gaining considerable impetus because of the emergence of drug-resistant strains of pathogens due to antibiotic therapy. Therefore, there is an urgent need to exploit alternative and novel strategies directed at host molecules to successfully restrict infections. The C-type lectin receptor CLEC4E and Toll-like receptor TLR4 expressed by host cells are among the first line of defense in encountering pathogens. Therefore, we exploited signaling of macrophages through CLEC4E in association with TLR4 agonists (C4.T4) to control the growth of Mycobacterium tuberculosis (Mtb). We observed significant improvement in host immunity and reduced bacterial load in the lungs of Mtb-infected mice and guinea pigs treated with C4.T4 agonists. Further, intracellular killing of Mtb was achieved with a 10-fold lower dose of isoniazid or rifampicin in conjunction with C4.T4 than the drugs alone. C4.T4 activated MYD88, PtdIns3K, STAT1 and RELA/NFKB, increased lysosome biogenesis, decreased Il10 and Il4 gene expression and enhanced macroautophagy/autophagy. Macrophages from autophagy-deficient (atg5 knockout or Becn1 knockdown) mice showed elevated survival of Mtb. The present findings also unveiled the novel role of CLEC4E in inducing autophagy through MYD88, which is required for control of Mtb growth. This study suggests a unique immunotherapeutic approach involving CLEC4E in conjunction with TLR4 to restrict the survival of Mtb through autophagy. ABBREVIATIONS 3MA: 3 methyladenine; AO: acridine orange; Atg5: autophagy related 5; AVOs: acidic vesicular organelles; BECN1: beclin 1, autophagy related; BMDMs: bone marrow derived macrophages; bw: body weight; C4.T4: agonists of CLEC4E (C4/TDB) and TLR4 (T4/ultra-pure-LPS); CFU: colony forming unit; CLEC4E/Mincle: C-type lectin domain family 4, member e; CLR: c-type lectin receptor; INH: isoniazid; LAMP1: lysosomal-associated membrane protein 1; MφC4.T4: Mtb-infected C4.T4 stimulated macrophages; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MDC: monodansylcadaverine; MTOR: mechanistic target of rapamycin kinase; MYD88: myeloid differentiation primary response 88; NFKB: nuclear factor of kappa light polypeptide gene enhance in B cells; NLR: NOD (nucleotide-binding oligomerization domain)-like receptors; PFA: paraformaldehyde; PPD: purified protein derivative; PtdIns3K: class III phosphatidylinositol 3-kinase; RELA: v-rel reticuloendotheliosis viral oncogene homolog A (avian); RIF: rifampicin; RLR: retinoic acid-inducible gene-I-like receptors; TDB: trehalose-6,6´-dibehenate; TLR4: toll-like receptor 4; Ultra-pure-LPS: ultra-pure lipopolysaccharide-EK; V-ATPase: vacuolar-type H+ ATPase.
Collapse
Affiliation(s)
- Susanta Pahari
- Immunology Division, CSIR-Institute of Microbial Technology , Chandigarh, India.,Host-Pathogen Interactions Program, Texas Biomedical Research Institute , San Antonio, TX, USA
| | - Shikha Negi
- Immunology Division, CSIR-Institute of Microbial Technology , Chandigarh, India
| | - Mohammad Aqdas
- Immunology Division, CSIR-Institute of Microbial Technology , Chandigarh, India
| | - Eusondia Arnett
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute , San Antonio, TX, USA
| | - Larry S Schlesinger
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute , San Antonio, TX, USA
| | - Javed N Agrewala
- Immunology Division, CSIR-Institute of Microbial Technology , Chandigarh, India.,Biomedical Engineering Department, Indian Institute of Technology Ropar , Rupnagar, India
| |
Collapse
|
15
|
Negi S, Pahari S, Bashir H, Agrewala JN. Gut Microbiota Regulates Mincle Mediated Activation of Lung Dendritic Cells to Protect Against Mycobacterium tuberculosis. Front Immunol 2019; 10:1142. [PMID: 31231363 PMCID: PMC6558411 DOI: 10.3389/fimmu.2019.01142] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 05/07/2019] [Indexed: 12/11/2022] Open
Abstract
Gut microbial components serve as ligand for various pattern recognition receptors (PRRs) present on immune cells and thereby regulates host immunity. Dendritic cells (DCs) are highly specialized innate cells involved in immune response to Mycobacterium tuberculosis (Mtb) infection. The gut-lung axis is a potential therapeutic target in tuberculosis; however, understanding of the innate immune mechanism underlying the interaction of gut microbiota and lung still remains obscure. We investigated if antibiotics (Abx) induced gut dysbiosis is able to affect the activation of innate receptor, macrophage inducible C-type lectin (mincle) in lungs during Mtb infection. We found that dysbiosis reduced the lung mincle expression with a concomitant increase in Mtb survival. Further, Abx diminished the effector and memory T cell population, while elevating frequency of regulatory T cells (Tregs) in the lungs. Here, we show that dysbiotic mice exhibited low mincle expression on lung DCs. These DCs with impaired phenotype and functions had reduced ability to activate naïve CD4 T cells, and thus unable to restrict Mtb survival. In vivo administration of trehalose-6,6-dibehenate (TDB: mincle ligand) efficiently rescued this immune defect by enhancing lung DCs function and subsequent T cell response. Further, gut microbial profiling revealed augmentation of Lactobacillus upon mincle stimulation in microbiota depleted animals. Accordingly, supplementation with Lactobacillus restored mincle expression on lung DCs along with anti-Mtb response. Our data demonstrate that gut microbiota is crucial to maintain DC-dependent lung immune response against Mtb, mediated by mincle. Abx interrupt this process to induce impaired T cell-response and increased susceptibility to Mtb.
Collapse
Affiliation(s)
- Shikha Negi
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Susanta Pahari
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India.,Immunology Division, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Hilal Bashir
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed N Agrewala
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India.,Center for Biomedical Engineering, Indian Institute of Technology, Rupnagar, India
| |
Collapse
|
16
|
Negi S, Pahari S, Das DK, Khan N, Agrewala JN. Curdlan Limits Mycobacterium tuberculosis Survival Through STAT-1 Regulated Nitric Oxide Production. Front Microbiol 2019; 10:1173. [PMID: 31191491 PMCID: PMC6547911 DOI: 10.3389/fmicb.2019.01173] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 05/08/2019] [Indexed: 12/16/2022] Open
Abstract
Host-directed therapies have emerged as an innovative and promising approach in tuberculosis (TB) treatment due to the observed limitations of current TB regimen such as lengthy duration and emergence of drug resistance. Thus, we explored the role of curdlan (beta glucan polysaccharide) as a novel strategy to activate macrophages against Mycobacterium tuberculosis (Mtb). The aim of the study was to investigate the role of curdlan in restricting the Mtb growth both in vitro and in vivo. Further, the immunomodulatory potential of curdlan against Mtb and the underlying mechanism is largely unknown. We found that curdlan treatment enhanced the antigen presentation, pro-inflammatory cytokines, Mtb uptake and killing activity of macrophages. In vivo studies showed that curdlan therapy significantly reduced the Mtb burden in lung and spleen of mice. Administration of curdlan triggered the protective Th1 and Th17 immunity while boosting the central and effector memory response in Mtb infected mice. Curdlan mediated anti-Mtb activity is through signal transducer and activator of transcription-1 (STAT-1), which regulates nitric oxide (NO) production through inducible NO synthase (iNOS) induction; along with this activation of nuclear factor kappa B (NF-κB) was also evident in Mtb infected macrophages. Thus, we demonstrate that curdlan exerts effective anti-tuberculous activity anti-tuberculous activity. It can be used as a potential host-directed therapy against Mtb.
Collapse
Affiliation(s)
- Shikha Negi
- Immunology Division, CSIR - Institute of Microbial Technology, Chandigarh, India
| | - Susanta Pahari
- Immunology Division, CSIR - Institute of Microbial Technology, Chandigarh, India.,Immunology Division, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Deepjyoti Kumar Das
- Immunology Division, CSIR - Institute of Microbial Technology, Chandigarh, India
| | - Nargis Khan
- Immunology Division, CSIR - Institute of Microbial Technology, Chandigarh, India.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Javed N Agrewala
- Immunology Division, CSIR - Institute of Microbial Technology, Chandigarh, India.,Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| |
Collapse
|
17
|
Chatterjee D, Kaur G, Muradia S, Singh B, Agrewala JN. ImmtorLig_DB: repertoire of virtually screened small molecules against immune receptors to bolster host immunity. Sci Rep 2019; 9:3092. [PMID: 30816123 PMCID: PMC6395627 DOI: 10.1038/s41598-018-36179-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 11/15/2018] [Indexed: 10/31/2022] Open
Abstract
Host directed therapies to boost immunity against infection are gaining considerable impetus following the observation that use of antibiotics has become a continuous source for the emergence of drug resistant strains of pathogens. Receptors expressed by the cells of immune system play a cardinal role in initiating sequence of events necessary to ameliorate many morbid conditions. Although, ligands for the immune receptors are available; but their use is limited due to complex structure, synthesis and cost-effectiveness. Virtual screening (VS) is an integral part of chemoinformatics and computer-aided drug design (CADD) and aims to streamline the process of drug discovery. ImmtorLig_DB is a repertoire of 5000 novel small molecules, screened from ZINC database and ranked using structure based virtual screening (SBVS) against 25 immune receptors which play a pivotal role in defending and initiating the activation of immune system. Consequently, in the current study, small molecules were screened by docking on the essential domains present on the receptors expressed by cells of immune system. The screened molecules exhibited efficacious binding to immune receptors, and indicated a possibility of discovering novel small molecules. Other features of ImmtorLig_DB include information about availability, clustering analysis, and estimation of absorption, distribution, metabolism, and excretion (ADME) properties of the screened small molecules. Structural comparisons indicate that predicted small molecules may be considered novel. Further, this repertoire is available via a searchable graphical user interface (GUI) through http://bioinfo.imtech.res.in/bvs/immtor/ .
Collapse
Affiliation(s)
| | - Gurkirat Kaur
- CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Shilpa Muradia
- CSIR-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Balvinder Singh
- CSIR-Institute of Microbial Technology, Chandigarh, 160036, India.
| | | |
Collapse
|
18
|
Gupta CL, Babu Khan M, Ampasala DR, Akhtar S, Dwivedi UN, Bajpai P. Pharmacophore-based virtual screening approach for identification of potent natural modulatory compounds of human Toll-like receptor 7. J Biomol Struct Dyn 2019; 37:4721-4736. [PMID: 30661449 DOI: 10.1080/07391102.2018.1559098] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Toll-like receptor 7 (TLR7) is a transmembrane glycoprotein playing very crucial role in the signaling pathways involved in innate immunity and has been demonstrated to be useful in fighting against infectious disease by recognizing viral ssRNA & specific small molecule agonists. In order to find novel human TLR7 (hTLR7) modulators, computational ligand-based pharmacophore modeling approach was used to identify the molecular chemical features required for the modulation of hTLR7 protein. A training set of 20 TLR7 agonists with their known experimental activity was used to create pharmacophore model using 3D-QSAR pharmacophore generation (HypoGen algorithm) module in Discovery Studio. The best developed hypothesis consists of four pharmacophoric features namely, one hydrogen bond donor (HBD), one ring aromatic (RA), and two hydrophobic (HY) character. The developed hypothesis was then validated by different methods such as cost analysis, test set method, and Fischer's test method for consistency. Hence, this validated model was further employed for screening of natural hit compounds from InterBioScreen Natural product database, consisting of more than 60,000 natural compounds and derivatives. The screened hit compounds were subsequently filtered by Lipinski's rule of 5, ADME and toxicity parameters and molecular docking studies to remove the false positive rates. Finally, molecular docking analysis led to identification of the (3a'S,6a'R)-3'-(3,4-dihydroxybenzyl)-5'-(3,4-dimethoxyphenethyl)-5-ethyl-3',3a'-dihydro-2'H-spiro[indoline-3,1'-pyrrolo[3,4-c]pyrrole]-2,4',6'(5'H,6a'H)-trione (Compound ID: STOCK1N-65837) as potent hTLR7 modulator due to its better docking score and molecular interactions compared to other compounds. The result of virtual screening was further validated using molecular dynamics (MD) simulation analysis. Thus, a 30 ns MD simulation analysis revealed high stability and effective binding of STOCK1N-65837 within the binding site of hTLR7. Therefore, the present study provides confidence for the utility of the selected chemical feature based pharmacophore model to design novel TLR7 modulators with desired biological activity.
Collapse
Affiliation(s)
- Chhedi Lal Gupta
- Institute for Development of Advanced computing, ONGC Centre for Advanced studies, University of Lucknow , Lucknow , UP , India.,Molecular Immunology Laboratory, Department of Biosciences, Integral University , Lucknow , UP , India
| | - Mohd Babu Khan
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University , Puducherry , India
| | - Dinakara Rao Ampasala
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University , Puducherry , India
| | - Salman Akhtar
- Department of Bioengineering, Integral University , Lucknow , UP , India
| | - Upendra Nath Dwivedi
- Institute for Development of Advanced computing, ONGC Centre for Advanced studies, University of Lucknow , Lucknow , UP , India.,Department of Biochemistry, Centre of Excellence in Bioinformatics, Bioinformatics Infrastructure Facility, University of Lucknow , Lucknow , UP , India
| | - Preeti Bajpai
- Molecular Immunology Laboratory, Department of Biosciences, Integral University , Lucknow , UP , India
| |
Collapse
|
19
|
Mubin N, Pahari S, Owais M, Zubair S. Mycobacterium tuberculosis host cell interaction: Role of latency associated protein Acr-1 in differential modulation of macrophages. PLoS One 2018; 13:e0206459. [PMID: 30395609 PMCID: PMC6218195 DOI: 10.1371/journal.pone.0206459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 10/13/2018] [Indexed: 11/19/2022] Open
Abstract
Mycobacterium tuberculosis (M.tb) contrives intracellular abode as a strategy to combat antibody onslaught. Additionally, to thrive against hostile ambiance inside host macrophages, the pathogen inhibits phago-lysosomal fusion. Finally, to further defy host cell offensives, M.tb opts for dormant phase, where it turns off or slows down most of its metabolic process as an added stratagem. While M.tb restrains most of its metabolic activities during dormancy, surprisingly latency-associated alpha-crystallin protein (Acr-1) is expressed most prominently during this phase. Interestingly, several previous studies described the potential of Acr-1 to induce the robust immuno-prophylactic response in the immunized host. It is intriguing to comprehend the apparent discrepancy that the microbe M.tb overexpresses a protein that has the potential to prime host immune system against the pathogen itself. Keeping this apparent ambiguity into consideration, it is imperative to unravel intricacies involved in the exploitation of Acr-1 by M.tb during its interaction with host immune cells. The present study suggests that Acr-1 exhibits diverse role in the maturation of macrophages (MΦs) and related immunological responses. The early encounter of bone marrow derived immune cells (pre-exposure during differentiation to MΦs) with Acr-1 (AcrMΦpre), results in hampering of their function. The pre-exposure of naïve MΦs with Acr-1 induces the expression of TIM-3 and IL-10. In contrast, exposure of fully differentiated MΦs to Acr-1 results in their down-modulation and induces the phosphorylation of STAT-1 and STAT-4 in host MΦs. Furthermore, Acr-1 mediated activation of MΦs results in the induction of Th1 and Th17 phenotype by activated T lymphocyte.
Collapse
Affiliation(s)
- Nida Mubin
- Molecular Immunology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Susanta Pahari
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Mohammad Owais
- Molecular Immunology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
- * E-mail: (SZ); (MO)
| | - Swaleha Zubair
- Department of Computer Science, Aligarh Muslim University, Aligarh, India
- * E-mail: (SZ); (MO)
| |
Collapse
|
20
|
Hertz D, Schneider B. Sex differences in tuberculosis. Semin Immunopathol 2018; 41:225-237. [PMID: 30361803 DOI: 10.1007/s00281-018-0725-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022]
Abstract
Tuberculosis is the most prevalent bacterial infectious disease in humans and the leading cause of death from a single infectious agent, ranking above HIV/AIDS. The causative agent, Mycobacterium tuberculosis, is carried by an estimated two billion people globally and claims more than 1.5 million lives each year. Tuberculosis rates are significantly higher in men than in women, reflected by a male-to-female ratio for worldwide case notifications of 1.7. This phenomenon is not new and has been reported in various countries and settings over the last century. However, the reasons for the observed gender bias are not clear, potentially highly complex and discussed controversially in the literature. Both gender- (referring to sociocultural roles and behavior) and sex-related factors (referring to biological aspects) likely contribute to higher tuberculosis rates in men and will be discussed.
Collapse
Affiliation(s)
- David Hertz
- Coinfection Unit, Priority Research Area Infections, Research Center Borstel, Parkallee 1-40, 23847, Borstel, Germany
| | - Bianca Schneider
- Coinfection Unit, Priority Research Area Infections, Research Center Borstel, Parkallee 1-40, 23847, Borstel, Germany.
| |
Collapse
|
21
|
Hu S, Du X, Huang Y, Fu Y, Yang Y, Zhan X, He W, Wen Q, Zhou X, Zhou C, Zhong XP, Yang J, Xiong W, Wang R, Gao Y, Ma L. NLRC3 negatively regulates CD4+ T cells and impacts protective immunity during Mycobacterium tuberculosis infection. PLoS Pathog 2018; 14:e1007266. [PMID: 30133544 PMCID: PMC6122840 DOI: 10.1371/journal.ppat.1007266] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/04/2018] [Accepted: 08/08/2018] [Indexed: 02/06/2023] Open
Abstract
NLRC3, a member of the NLR family, has been reported as a negative regulator of inflammatory signaling pathways in innate immune cells. However, the direct role of NLRC3 in modulation of CD4+ T-cell responses in infectious diseases has not been studied. In the present study, we showed that NLRC3 plays an intrinsic role by suppressing the CD4+ T cell phenotype in lung and spleen, including differentiation, activation, and proliferation. NLRC3 deficiency in CD4+ T cells enhanced the protective immune response against Mycobacterium tuberculosis infection. Finally, we demonstrated that NLRC3 deficiency promoted the activation, proliferation, and cytokine production of CD4+ T cells via negatively regulating the NF-κB and MEK-ERK signaling pathways. This study reveals a critical role of NLRC3 as a direct regulator of the adaptive immune response and its protective effects on immunity during M. tuberculosis infection. Our findings also suggested that NLRC3 serves as a potential target for therapeutic intervention against tuberculosis.
Collapse
Affiliation(s)
- Shengfeng Hu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xialin Du
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yulan Huang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yuling Fu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yalong Yang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiaoxia Zhan
- Department of laboratory medicine, The first Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenting He
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Qian Wen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xinying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Chaoying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiao-Ping Zhong
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Department of Pediatrics, Division of Allergy and Immunology, Duke University Medical Center, Durham, NC, United States of America
| | - Jiahui Yang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Wenjing Xiong
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Ruining Wang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yuchi Gao
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- * E-mail:
| |
Collapse
|
22
|
Sharma R, Dubey S, Mody N, Sharma G, Kushwah V, Jain S, Katare OP, Vyas SP. Release promoter-based systematically designed nanocomposite(s): a novel approach for site-specific delivery of tumor-associated antigen(s) (TAAs). ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:776-789. [DOI: 10.1080/21691401.2018.1469137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Rajeev Sharma
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Central University, Sagar, India
| | - Surabhi Dubey
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Central University, Sagar, India
| | - Nishi Mody
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Central University, Sagar, India
| | - Gajanand Sharma
- University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Studies, Panjab University, Chandigarh, India
| | - Varun Kushwah
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Mohali, Punjab, India
| | - Om Prakash Katare
- University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Studies, Panjab University, Chandigarh, India
| | - Suresh P. Vyas
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour Central University, Sagar, India
| |
Collapse
|
23
|
López-Hernández Y, Rivas-Santiago CE, López JA, Mendoza-Almanza G, Hernandez-Pando R. Tuberculosis and cigarette smoke exposure: An update of in vitro and in vivo studies. Exp Lung Res 2018; 44:113-126. [PMID: 29565741 DOI: 10.1080/01902148.2018.1444824] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tuberculosis (TB) has been declared the first cause of death by an infectious agent. Annually, 10.4 million people suffer active TB. Most infected individuals live in low-income countries, where social and economic conditions enhance the dissemination and progression of the disease. These countries have a high percentage of smokers. Thousands of studies have linked cigarette smoke (CS) with increased risk of many diseases, such as cancer and lung diseases. Numerous in vitro studies have been conducted to evaluate the general and specific toxic effects of CS in lung immune function. Smoke exposure increases the risk of TB development three-fold. However, until now, only few animal studies have been performed to analyze the association between smoke and TB. In the present work, we review in vitro and in vivo studies whose aim was to analyze the molecular basis of TB susceptibility caused by exposure to CS.
Collapse
Affiliation(s)
- Y López-Hernández
- a CONACyT, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - C E Rivas-Santiago
- a CONACyT, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - J A López
- b Laboratorio de MicroRNAs, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - G Mendoza-Almanza
- a CONACyT, Unidad Academica de Ciencias Biologicas , Universidad Autónoma de Zacatecas , Zacatecas , Mexico
| | - R Hernandez-Pando
- c Departamento de Patologia, Unidad de Patologia Experimental , Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran , Mexico
| |
Collapse
|
24
|
Pahari S, Kaur G, Negi S, Aqdas M, Das DK, Bashir H, Singh S, Nagare M, Khan J, Agrewala JN. Reinforcing the Functionality of Mononuclear Phagocyte System to Control Tuberculosis. Front Immunol 2018; 9:193. [PMID: 29479353 PMCID: PMC5811511 DOI: 10.3389/fimmu.2018.00193] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022] Open
Abstract
The mononuclear phagocyte system (MPS) constitutes dendritic cells, monocytes, and macrophages. This system contributes to various functions that are essential for maintaining homeostasis, activation of innate immunity, and bridging it with the adaptive immunity. Consequently, MPS is highly important in bolstering immunity against the pathogens. However, MPS is the frontline cells in destroying Mycobacterium tuberculosis (Mtb), yet the bacterium prefers to reside in the hostile environment of macrophages. Therefore, it may be very interesting to study the struggle between Mtb and MPS to understand the outcome of the disease. In an event when MPS predominates Mtb, the host remains protected. By contrast, the situation becomes devastating when the pathogen tames and tunes the host MPS, which ultimately culminates into tuberculosis (TB). Hence, it becomes extremely crucial to reinvigorate MPS functionality to overwhelm Mtb and eliminate it. In this article, we discuss the strategies to bolster the function of MPS by exploiting the molecules associated with the innate immunity and highlight the mechanisms involved to overcome the Mtb-induced suppression of host immunity. In future, such approaches may provide an insight to develop immunotherapeutics to treat TB.
Collapse
Affiliation(s)
- Susanta Pahari
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Gurpreet Kaur
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Shikha Negi
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Mohammad Aqdas
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Deepjyoti K Das
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Hilal Bashir
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Sanpreet Singh
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Mukta Nagare
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Junaid Khan
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed N Agrewala
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|