Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Microsyst Technol ; 27(9): 3279-3284, 2021.
Article in English | MEDLINE | ID: mdl-33191983

ABSTRACT

The present research proposes the novel corona virus kit using silicon based two (2D) photonic structure. The basic principle of the estimation of different corona viruses relies on the computation of reflectance, absorbance, and transmittance at the signal of 412 nm. Here reflectance is investigated through the analysis of photonic band gap where absorbance is calculated using analytical treatment. The present investigation is made for different corona virus such as N5H1, N5H2, H9N2, H4N6, FAdV, and IBV. The numerical analysis indicates that the sample could be affected by novel corona virus if the transmitted signal lies with red spectrum. Similarly, sample could be normal viruses if the transmitted signal would green spectrum.

2.
J Biol Chem ; 294(52): 19862-19876, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31653701

ABSTRACT

Paired two-component systems (TCSs), having a sensor kinase (SK) and a cognate response regulator (RR), enable the human pathogen Mycobacterium tuberculosis to respond to the external environment and to persist within its host. Here, we inactivated the SK gene of the TCS MtrAB, mtrB, generating the strain ΔmtrB We show that mtrB loss reduces the bacterium's ability to survive in macrophages and increases its association with autophagosomes and autolysosomes. Notably, the ΔmtrB strain was markedly defective in establishing lung infection in mice, with no detectable lung pathology following aerosol challenge. ΔmtrB was less able to withstand hypoxic and acid stresses and to form biofilms and had decreased viability under hypoxia. Transcriptional profiling of ΔmtrB by gene microarray analysis, validated by quantitative RT-PCR, indicated down-regulation of the hypoxia-associated dosR regulon, as well as genes associated with other pathways linked to adaptation of M. tuberculosis to the host environment. Using in vitro biochemical assays, we demonstrate that MtrB interacts with DosR (a noncognate RR) in a phosphorylation-independent manner. Electrophoretic mobility shift assays revealed that MtrB enhances the binding of DosR to the hspX promoter, suggesting an unexpected role of MtrB in DosR-regulated gene expression in M. tuberculosis Taken together, these findings indicate that MtrB functions as a regulator of DosR-dependent gene expression and in the adaptation of M. tuberculosis to hypoxia and the host environment. We propose that MtrB may be exploited as a chemotherapeutic target against tuberculosis.


Subject(s)
Bacterial Proteins/metabolism , Mycobacterium tuberculosis/physiology , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Autophagosomes/metabolism , Bacterial Proteins/genetics , Biofilms/growth & development , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Gene Regulatory Networks , Host-Pathogen Interactions , Humans , Lung Diseases/microbiology , Lung Diseases/pathology , Lung Diseases/veterinary , Lysosomes/metabolism , Macrophages/cytology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/growth & development , Phosphorylation , Promoter Regions, Genetic , Protein Binding , RNA-Binding Proteins/genetics , Transcription Factors/genetics
3.
PLoS Pathog ; 13(5): e1006410, 2017 May.
Article in English | MEDLINE | ID: mdl-28558034

ABSTRACT

For efficient clearance of Mycobacterium tuberculosis (Mtb), macrophages tilt towards M1 polarization leading to the activation of transcription factors associated with the production of antibacterial effector molecules such as nitric oxide (NO) and proinflammatory cytokines such as interleukin 1 ß (IL-1ß) and tumor necrosis factor α (TNF-α). At the same time, resolution of inflammation is associated with M2 polarization with increased production of arginase and cytokines such as IL-10. The transcriptional and post-transcriptional mechanisms that govern the balance between M1 and M2 polarization, and bacteria-containing processes such as autophagy and trafficking of Mtb to lysosomes, are incompletely understood. Here we report for the first time, that the transcription factor KLF4 is targeted by microRNA-26a (miR-26a). During Mtb infection, downregulation of miR-26a (observed both ex vivo and in vivo) facilitates upregulation of KLF4 which in turn favors increased arginase and decreased iNOS activity. We further demonstrate that KLF4 prevents trafficking of Mtb to lysosomes. The CREB-C/EBPß signaling axis also favors M2 polarization. Downregulation of miR-26a and upregulation of C/ebpbeta were observed both in infected macrophages as well as in infected mice. Knockdown of C/ebpbeta repressed the expression of selected M2 markers such as Il10 and Irf4 in infected macrophages. The importance of these pathways is substantiated by observations that expression of miR-26a mimic or knockdown of Klf4 or Creb or C/ebpbeta, attenuated the survival of Mtb in macrophages. Taken together, our results attribute crucial roles for the miR-26a/KLF4 and CREB-C/EBPßsignaling pathways in regulating the survival of Mtb in macrophages. These studies expand our understanding of how Mtb hijacks host signaling pathways to survive in macrophages, and open up new exploratory avenues for host-targeted interventions.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/immunology , CREB-Binding Protein/immunology , Kruppel-Like Transcription Factors/immunology , Lysosomes/microbiology , Macrophages/immunology , MicroRNAs/immunology , Mycobacterium tuberculosis/physiology , Tuberculosis/immunology , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CREB-Binding Protein/genetics , Cell Polarity , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Lysosomes/genetics , Lysosomes/immunology , Macrophages/cytology , Macrophages/microbiology , Mice , MicroRNAs/genetics , Mycobacterium tuberculosis/immunology , RAW 264.7 Cells , Signal Transduction , Tuberculosis/genetics , Tuberculosis/microbiology , Tuberculosis/physiopathology
4.
Sci Rep ; 6: 25851, 2016 05 16.
Article in English | MEDLINE | ID: mdl-27181265

ABSTRACT

The genome of M. tuberculosis (Mtb) encodes eleven paired two component systems (TCSs) consisting of a sensor kinase (SK) and a response regulator (RR). The SKs sense environmental signals triggering RR-dependent gene expression pathways that enable the bacterium to adapt in the host milieu. We demonstrate that a conserved motif present in the C-terminal domain regulates the DNA binding functions of the OmpR family of Mtb RRs. Molecular docking studies against this motif helped to identify two molecules with a thiazolidine scaffold capable of targeting multiple RRs, and modulating their regulons to attenuate bacterial replication in macrophages. The changes in the bacterial transcriptome extended to an altered immune response with increased autophagy and NO production, leading to compromised survival of Mtb in macrophages. Our findings underscore the promise of targeting multiple RRs as a novel yet unexplored approach for development of new anti-mycobacterial agents particularly against drug-resistant Mtb.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Bacterial Proteins/chemistry , Mycobacterium tuberculosis/genetics , Tuberculosis/immunology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Autophagy , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cells, Cultured , DNA/metabolism , Gene Expression Profiling/methods , Humans , Macrophages/cytology , Macrophages/immunology , Macrophages/microbiology , Mice , Models, Molecular , Molecular Docking Simulation , Mutation , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/metabolism , Nitric Oxide/metabolism , Protein Binding , RAW 264.7 Cells , Tuberculosis/microbiology
5.
Cell Microbiol ; 18(5): 679-91, 2016 May.
Article in English | MEDLINE | ID: mdl-26513648

ABSTRACT

Autophagy plays a crucial role in the control of bacterial burden during Mycobacterium tuberculosis infection. MicroRNAs (miRNAs) are small non-coding RNAs that regulate immune signalling and inflammation in response to challenge by pathogens. Appreciating the potential of host-directed therapies designed to control autophagy during mycobacterial infection, we focused on the role of miRNAs in regulating M. tuberculosis-induced autophagy in macrophages. Here, we demonstrate that M. tuberculosis infection leads to downregulation of miR-17 and concomitant upregulation of its targets Mcl-1 and STAT3, a transcriptional activator of Mcl-1. Forced expression of miR-17 reduces expression of Mcl-1 and STAT3 and also the interaction between Mcl-1 and Beclin-1. This is directly linked to enhanced autophagy, because Mcl-1 overexpression attenuates the effects of miR-17. At the same time, transfection with a kinase-inactive mutant of protein kinase C δ (PKCδ) (an activator of STAT3) augments M. tuberculosis-induced autophagy, and miR-17 overexpression diminishes phosphorylation of PKCδ, suggesting that an miR-17/PKC δ/STAT3 axis regulates autophagy during M. tuberculosis infection.


Subject(s)
MicroRNAs/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , STAT3 Transcription Factor/genetics , Tuberculosis/genetics , Animals , Autophagy/genetics , Beclin-1/genetics , HEK293 Cells , Humans , Macrophages/metabolism , Macrophages/microbiology , Mice , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Protein Kinase C-delta/genetics , RAW 264.7 Cells , Signal Transduction/genetics , Tuberculosis/microbiology
6.
Cell Host Microbe ; 17(3): 345-356, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25683052

ABSTRACT

The outcome of the interaction between Mycobacterium tuberculosis (Mtb) and a macrophage depends on the interplay between host defense and bacterial immune subversion mechanisms. MicroRNAs critically regulate several host defense mechanisms, but their role in the Mtb-macrophage interplay remains unclear. MicroRNA profiling of Mtb-infected macrophages revealed the downregulation of miR-let-7f in a manner dependent on the Mtb secreted effector ESAT-6. We establish that let-7f targets A20, a feedback inhibitor of the NF-κB pathway. Expression of let-7f decreases and A20 increases with progression of Mtb infection in mice. Mtb survival is attenuated in A20-deficient macrophages, and the production of TNF, IL-1ß, and nitrite, which are mediators of immunity to Mtb, is correspondingly increased. Further, let-7f overexpression diminishes Mtb survival and augments the production of cytokines including TNF and IL-1ß. These results uncover a role for let-7f and its target A20 in regulating immune responses to Mtb and controlling bacterial burden.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , DNA-Binding Proteins/biosynthesis , Host-Pathogen Interactions , Intracellular Signaling Peptides and Proteins/biosynthesis , Macrophages/immunology , MicroRNAs/antagonists & inhibitors , Mycobacterium tuberculosis/immunology , NF-kappa B/metabolism , Nuclear Proteins/biosynthesis , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Epithelial Cells/immunology , Epithelial Cells/microbiology , Gene Expression Profiling , Humans , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/microbiology , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Sequence Analysis, DNA , Tumor Necrosis Factor alpha-Induced Protein 3
SELECTION OF CITATIONS
SEARCH DETAIL
...