Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biomol Concepts ; 11(1): 76-85, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32271156

ABSTRACT

Interferon-gamma (IFN-γ) is a key cytokine that mediates immunity to tuberculosis (TB). Mycobacterium tuberculosis (M. tb) is known to downregulate the surface expression of IFN-γ receptor (IFN-γR) on macrophages and peripheral blood mononuclear cells (PBMCs) of patients with active TB disease. Many M. tb antigens also downmodulate IFN-γR levels in macrophages when compared with healthy controls. In the current study, we aimed at deciphering key factors involved in M. tb mediated downregulation of IFN-γR levels on macrophage surface. Our data showed that both M. tb H37Rv and M. bovis BCG infections mediate downmodulation of IFN-γR on human macrophages. This downmodulation is regulated at the level of TLR signaling pathway, second messengers such as calcium and cellular kinases i.e. PKC and ERK-MAPK, indicating that fine tuning of calcium response is critical to maintaining IFN-γR levels on macrophage surface. In addition, genes in the calcium and cysteine protease pathways which were previously identified by us to play a negative role during M. tb infection, also regulated IFN-γR expression. Thus, modulations in IFN-γR levels by utilizing host machinery may be a key immune suppressive strategy adopted by the TB pathogen to ensure its persistence and thwart host defense.


Subject(s)
Calcium/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/physiology , Receptors, Interferon/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Cytokines/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , Female , Homeostasis , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Leukocytes, Mononuclear/metabolism , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Inbred BALB C , Mycobacterium bovis/physiology , Protein Kinase C/metabolism , RNA, Small Interfering , Receptors, Interferon/genetics , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Interferon gamma Receptor
2.
Biomol Concepts ; 11(1): 230-239, 2020 Dec 31.
Article in English | MEDLINE | ID: mdl-33726488

ABSTRACT

Staphylococcus aureus (S. aureus) is a gram-positive bacteria, which causes various fatal respiratory infections including pneumonia. The emergence of Methicillin-Resistance Staphylococcus aureus (MRSA) demands a thorough understanding of host-pathogen interactions. Here we report the role of calcium in regulating defence responses of S. aureus in macrophages. Regulating calcium fluxes in cells by different routes differentially governs the expression of T cell costimulatory molecule CD80 and Th1 promoting IL-12 receptor. Inhibiting calcium influx from extracellular medium increased expression of IFN-γ and IL-10 while blocking calcium release from the intracellular stores inhibited TGF-ß levels. Blocking voltage-gated calcium channels (VGCC) inhibited the expression of multiple cytokines. While VGCC regulated the expression of apoptosis protein Bax, extracellular calcium-regulated the expression of Cytochrome-C. Similarly, VGCC regulated the expression of autophagy initiator Beclin-1. Blocking VGCC or calcium release from intracellular stores promoted phagosome-lysosome fusion, while activating VGCC inhibited phagosomelysosome fusion. Finally, calcium homeostasis regulated intracellular growth of Staphylococcus, although using different mechanisms. While blocking extracellular calcium influx seems to rely on IFN-γ and IL-12Rß receptor mediated reduction in bacterial survival, blocking either intracellular calcium release or via VGCC route seem to rely on enhanced autophagy mediated reduction of intracellular bacterial survival. These results point to fine-tuning of defence responses by routes of calcium homeostasis.


Subject(s)
Calcium/metabolism , Macrophages/metabolism , Protective Agents/metabolism , Staphylococcus aureus/growth & development , Animals , Cells, Cultured , Cytokines/analysis , Cytokines/biosynthesis , Homeostasis , Mice
3.
Biomol Concepts ; 9(1): 64-79, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29856726

ABSTRACT

A key player in driving cellular immunity, IFN-γ is capable of orchestrating numerous protective functions to heighten immune responses in infections and cancers. It can exhibit its immunomodulatory effects by enhancing antigen processing and presentation, increasing leukocyte trafficking, inducing an anti-viral state, boosting the anti-microbial functions and affecting cellular proliferation and apoptosis. A complex interplay between immune cell activity and IFN-γ through coordinated integration of signals from other pathways involving cytokines and Pattern Recognition Receptors (PRRs) such as Interleukin (IL)-4, TNF-α, Lipopolysaccharide (LPS), Type-I Interferons (IFNS) etc. leads to initiation of a cascade of pro-inflammatory responses. Microarray data has unraveled numerous genes whose transcriptional regulation is influenced by IFN-γ. Consequently, IFN-γ stimulated cells display altered expression of many such target genes which mediate its downstream effector functions. The importance of IFN-γ is further reinforced by the fact that mice possessing disruptions in the IFN-γ gene or its receptor develop extreme susceptibility to infectious diseases and rapidly succumb to them. In this review, we attempt to elucidate the biological functions and physiological importance of this versatile cytokine. The functional implications of its biological activity in several infectious diseases and autoimmune pathologies are also discussed. As a counter strategy, many virulent pathogenic species have devised ways to thwart IFN-γ endowed immune-protection. Thus, IFN-γ mediated host-pathogen interactions are critical for our understanding of disease mechanisms and these aspects also manifest enormous therapeutic importance for the annulment of various infections and autoimmune conditions.


Subject(s)
Communicable Diseases/immunology , Interferon-gamma/physiology , Animals , Communicable Diseases/drug therapy , Communicable Diseases/metabolism , Cytokines/physiology , Gene Expression Regulation , Humans , Interferon-gamma/metabolism , Interferon-gamma/therapeutic use , Listeriosis/immunology , Mice , Mycobacterium Infections/immunology , Parasitic Diseases/immunology , Rats , Salmonella Infections/immunology
4.
J Leukoc Biol ; 102(5): 1249-1259, 2017 11.
Article in English | MEDLINE | ID: mdl-28877954

ABSTRACT

Microorganisms are known to devise various strategies to thwart protective responses by the host. One such strategy is to incorporate sequences and domains in their genes/proteins that have similarity to various domains of the host proteins. In this study, we report that Mycobacterium tuberculosis protein Rv3529c exhibits significant similarity to the death domain of the TLR pathway adaptor protein MyD88. Incubation of macrophages with Rv3529c specifically inhibited TLR2-mediated proinflammatory responses. This included attenuated oxidative burst, reduced phosphorylation of MAPK-ERK, reduced activation of transcription factor NF-κB and reduced secretion of proinflammatory cytokines IFN-γ, IL-6, and IL-17A with a concomitant increased secretion of suppressor cytokines IL-10 and TGF-ß. Importantly, Rv3529c significantly inhibited TLR2-induced association of MyD88 with IRAK1 by competitively binding with IRAK1. Further, Rv3529c mediated inhibition of apoptosis and phagosome-lysosome fusion. Lastly, incubation of macrophages with Rv3529c increased bacterial burden inside macrophages. The data presented show another strategy evolved by M. tuberculosis toward immune evasion that centers on incorporating sequences in proteins that are similar to crucial proteins in the innate immune system of the host.


Subject(s)
Bacterial Proteins/pharmacology , Immune Evasion , Macrophages/microbiology , Mycobacterium tuberculosis/immunology , Toll-Like Receptor 2/immunology , Animals , Bacterial Load , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Gene Expression Regulation , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lysosomes/drug effects , Lysosomes/immunology , Macrophages/drug effects , Macrophages/immunology , Membrane Fusion/drug effects , Membrane Fusion/immunology , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/immunology , Molecular Mimicry , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/pathogenicity , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Phagosomes/drug effects , Phagosomes/immunology , Primary Cell Culture , Protein Domains , Respiratory Burst/immunology , Signal Transduction , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
5.
PLoS One ; 11(10): e0163845, 2016.
Article in English | MEDLINE | ID: mdl-27723836

ABSTRACT

The prevalence of Mycobacterium tuberculosis (M. tb) strains eliciting drug resistance has necessitated the need for understanding the complexities of host pathogen interactions. The regulation of calcium homeostasis by Voltage Gated Calcium Channel (VGCCs) upon M. tb infection has recently assumed importance in this area. We previously showed a suppressor role of VGCC during M. tb infections and recently reported the mechanisms of its regulation by M. tb. Here in this report, we further characterize the role of VGCC in mediating defence responses of macrophages during mycobacterial infection. We report that activation of VGCC during infection synergistically downmodulates the generation of oxidative burst (ROS) by macrophages. This attenuation of ROS is regulated in a manner which is dependent on Toll like Receptor (TLR) and also on the route of calcium influx, Protein Kinase C (PKC) and by Mitogen Activation Protein Kinase (MAPK) pathways. VGCC activation during infection increases cell survival and downmodulates autophagy. Concomitantly, pro-inflammatory responses such as IL-12 and IFN-γ secretion and the levels of their receptors on cell surface are inhibited. Finally, the ability of phagosomes to fuse with lysosomes in M. bovis BCG and M. tb H37Rv infected macrophages is also compromised when VGCC activation occurs during infection. The results point towards a well-orchestrated strategy adopted by mycobacteria to supress protective responses mounted by the host. This begins with the increase in the surface levels of VGCCs by mycobacteria and their antigens by well-controlled and regulated mechanisms. Subsequent activation of the upregulated VGCC following tweaking of calcium levels by molecular sensors in turn mediates suppressor responses and prepare the macrophages for long term persistent infection.


Subject(s)
Calcium Channels, L-Type/immunology , Calcium Signaling/immunology , Macrophages/immunology , Mycobacterium Infections/immunology , Mycobacterium bovis/immunology , Animals , Cell Line, Tumor , Female , Humans , Interferon-gamma/immunology , Interleukin-12/immunology , Male , Mice , Mice, Inbred BALB C , Reactive Oxygen Species/immunology
6.
PLoS One ; 10(7): e0131767, 2015.
Article in English | MEDLINE | ID: mdl-26132135

ABSTRACT

The emergence of drug resistant strains of Mycobacterium tuberculosis (M. tuberculosis) together with reports of co-infections with the human immunodeficiency virus (HIV) has renewed interest to better understand the intricate mechanisms prevalent during co-infections. In this study we report a synergistic effect of M. tuberculosis and HIV-1, and their antigens Rv3416 and Nef, respectively, in inhibiting apoptosis of macrophages. This inhibition involves the TLR2 pathway and second messengers that play complementing and contrasting roles in regulating apoptosis. Interestingly, the route of calcium influx into cells differentially regulates apoptosis during antigenic co-stimulation. While calcium released from intracellular stores was anti-apoptotic, calcium influx from the external milieu was pro-apoptotic. Further, molecular sensors of intracellular calcium release aid in antigen mediated inhibition of apoptosis. A cross-regulation between oxidative burst and differential routing of calcium influx governed apoptosis. Interestingly, the HIV-1 Nef supported anti-apoptotic responses in macrophages whereas Vpu had no significant effect. These results point to a synergistic liaison between M. tuberculosis and HIV-1 in regulating macrophage apoptosis.


Subject(s)
Apoptosis , Calcium/metabolism , HIV-1 , Macrophages/metabolism , Mycobacterium tuberculosis , Toll-Like Receptor 2/metabolism , Antigens/metabolism , Bacterial Proteins/metabolism , Cells, Cultured , Coinfection , Cytochromes c/metabolism , Gene Expression Regulation , HEK293 Cells , HIV Infections/complications , Homeostasis , Humans , Leukocytes, Mononuclear/cytology , Macrophages/microbiology , Macrophages/virology , Membrane Potential, Mitochondrial , RNA, Small Interfering/metabolism , Respiratory Burst , Signal Transduction , Tuberculosis/complications , nef Gene Products, Human Immunodeficiency Virus/metabolism
7.
PLoS One ; 10(4): e0124263, 2015.
Article in English | MEDLINE | ID: mdl-25915405

ABSTRACT

We demonstrated earlier the inhibitory role played by Voltage Gated Calcium Channels (VGCCs) in regulating Mycobacterium tuberculosis (M. tb) survival and pathogenesis. In this report, we investigated mechanisms and key players that regulate the surface expression of VGCC-CACNA1S by Rv2463 and M. tb infection in macrophages. Our earlier work identified Rv2463 to be expressed at early times post infection in macrophages that induced suppressor responses to dendritic cells and macrophages. Our results in this study demonstrate a role of MyD88 independent TLR pathway in mediating CACNA1S expression. Dissecting the role for second messengers, we show that calcium homeostasis plays a key role in CACNA1S expression during M. tb infection. Using siRNAs against molecular sensors of calcium regulation, we show an involvement of ER associated Stromal Interaction Molecules 1 and 2 (STIM1 and STIM2), and transcription factor pCREB, towards CACNA1S expression that also involved the MyD88 independent pathway. Interestingly, reactive oxygen species played a negative role in M. tb mediated CACNA1S expression. Further, a cross-regulation of ROS and pCREB was noted that governed CACNA1S expression. Characterizing the mechanisms governing CACNA1S expression would improve our understanding of the regulation of VGCC expression and its role in M. tb pathogenesis during M. tb infection.


Subject(s)
Calcium Channels/metabolism , Macrophages/immunology , Macrophages/metabolism , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tuberculosis/metabolism , Animals , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels, L-Type , Cell Adhesion Molecules/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Female , Flow Cytometry , Gene Expression , Membrane Proteins/metabolism , Mice , Models, Biological , Myeloid Differentiation Factor 88/metabolism , Neoplasm Proteins/metabolism , Reactive Oxygen Species/metabolism , Respiratory Burst/genetics , Signal Transduction , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2 , Transcription Factors/metabolism , Tuberculosis/genetics , Tuberculosis/microbiology
8.
PLoS One ; 9(5): e96427, 2014.
Article in English | MEDLINE | ID: mdl-24797940

ABSTRACT

Our previous work has demonstrated the roles played by L-type Voltage Gated Calcium Channels (VGCC) in regulating Mycobacterium tuberculosis (M. tb) survival and pathogenesis. Here we decipher mechanisms and pathways engaged by the pathogen to regulate VGCC expression in macrophages. We show that M. tb and its antigen Rv3416 use phospho-CREB (pCREB), Reactive Oxygen Species (ROS), Protein Kinase C (PKC) and Mitogen Activated Protein Kinase (MAPK) to modulate VGCC expression in macrophages. siRNA mediated knockdown of MyD88, IRAK1, IRAK2 or TRAF6 significantly inhibited antigen mediated VGCC expression. Inhibiting Protein Kinase C (PKC) or MEK-ERK1/2 further increased VGCC expression. Interestingly, inhibiting intracellular calcium release upregulated antigen mediated VGCC expression, while inhibiting extracellular calcium influx had no significant effect. siRNA mediated knockdown of transcription factors c-Jun, SOX5 and CREB significantly inhibited Rv3416 mediated VGCC expression. A dynamic reciprocal cross-regulation between ROS and pCREB was observed that in turn governed VGCC expression with ROS playing a limiting role in the process. Further dissection of the mechanisms such as the interplay between ROS and pCREB would improve our understanding of the regulation of VGCC expression during M. tb infection.


Subject(s)
Calcium Channels/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Mycobacterium tuberculosis/metabolism , Reactive Oxygen Species/metabolism , Antigens, Bacterial/physiology , Cell Line , Humans , Macrophages/metabolism , Metabolic Networks and Pathways/immunology , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , Mycobacterium tuberculosis/pathogenicity , Protein Kinase C/metabolism , Protein Kinase C/physiology
9.
EMBO J ; 33(9): 994-1010, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24431222

ABSTRACT

There is emerging evidence that stem cells can rejuvenate damaged cells by mitochondrial transfer. Earlier studies show that epithelial mitochondrial dysfunction is critical in asthma pathogenesis. Here we show for the first time that Miro1, a mitochondrial Rho-GTPase, regulates intercellular mitochondrial movement from mesenchymal stem cells (MSC) to epithelial cells (EC). We demonstrate that overexpression of Miro1 in MSC (MSCmiro(Hi)) leads to enhanced mitochondrial transfer and rescue of epithelial injury, while Miro1 knockdown (MSCmiro(Lo)) leads to loss of efficacy. Treatment with MSCmiro(Hi) was associated with greater therapeutic efficacy, when compared to control MSC, in mouse models of rotenone (Rot) induced airway injury and allergic airway inflammation (AAI). Notably, airway hyperresponsiveness and remodeling were reversed by MSCmiro(Hi) in three separate allergen-induced asthma models. In a human in vitro system, MSCmiro(Hi) reversed mitochondrial dysfunction in bronchial epithelial cells treated with pro-inflammatory supernatant of IL-13-induced macrophages. Anti-inflammatory MSC products like NO, TGF-ß, IL-10 and PGE2, were unchanged by Miro1 overexpression, excluding non-specific paracrine effects. In summary, Miro1 overexpression leads to increased stem cell repair.


Subject(s)
Lung Injury/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Mitochondria/metabolism , rho GTP-Binding Proteins/physiology , Animals , Biological Transport/genetics , Cells, Cultured , Genetic Therapy/methods , Humans , Lung/pathology , Lung Injury/pathology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mitochondria/transplantation , NIH 3T3 Cells , Nanotubes , Treatment Outcome , rho GTP-Binding Proteins/genetics
10.
J Biol Chem ; 287(14): 11108-21, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22337888

ABSTRACT

With rising incidence of acquired drug resistance among life-threatening pathogens, alternative approaches to improve therapy and vaccination have taken center stage. To this end, genome-wide and pathway-specific siRNA libraries are being employed increasingly to identify genes that regulate immune responses against a number of pathogens. In this study using calcium and cysteine protease pathway-specific siRNA libraries, we identified genes that play critical roles in modulating diverse functions of dendritic cells (DCs) during Mycobacterium tuberculosis infection. Knockdown of many of these genes in the two pathways resulted in reduced bacterial burden within DCs. These included genes that regulated activation of transcription factors, ubiquitin-specific peptidases, and genes that are involved in autophagy and neddylation. Knockdown of certain genes increased the expression of IL-12p40 and surface densities of costimulatory molecules in an antigen- and receptor-specific manner. Increased IL-12p40 and costimulatory molecules on DCs also promoted the development of Th1 responses from a Th2 inducing antigen. Furthermore, modulation of autophagy and oxidative burst appeared to be one of the mechanisms by which these genes regulated survival of M. tuberculosis within DCs. Although some genes regulated specific responses, others regulated multiple responses that included IL-12 production, T cell priming, as well as intracellular survival of M. tuberculosis. Further dissection of the mechanisms such as neddylation, by which these genes regulate immune responses, would improve our understanding of host parameters that are modulated during M. tuberculosis infection.


Subject(s)
Calcium/metabolism , Cysteine Proteases/metabolism , Dendritic Cells/immunology , Dendritic Cells/microbiology , Immunity/genetics , Mycobacterium tuberculosis/physiology , Animals , Autophagy/genetics , Autophagy/immunology , Calmodulin/metabolism , Cell Adhesion Molecules/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Gene Knockdown Techniques , Interleukin-12 Subunit p40/biosynthesis , Mice , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Respiratory Burst/genetics , Respiratory Burst/immunology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/microbiology , Toll-Like Receptor 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...