Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Biol Chem ; 290(44): 26576-86, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26391398

ABSTRACT

Specific and coordinated regulation of innate immune receptor-driven signaling networks often determines the net outcome of the immune responses. Here, we investigated the cross-regulation of toll-like receptor (TLR)2 and nucleotide-binding oligomerization domain (NOD)2 pathways mediated by Ac2PIM, a tetra-acylated form of mycobacterial cell wall component and muramyl dipeptide (MDP), a peptidoglycan derivative respectively. While Ac2PIM treatment of macrophages compromised their ability to induce NOD2-dependent immunomodulators like cyclooxygenase (COX)-2, suppressor of cytokine signaling (SOCS)-3, and matrix metalloproteinase (MMP)-9, no change in the NOD2-responsive NO, TNF-α, VEGF-A, and IL-12 levels was observed. Further, genome-wide microRNA expression profiling identified Ac2PIM-responsive miR-150 and miR-143 to target NOD2 signaling adaptors, RIP2 and TAK1, respectively. Interestingly, Ac2PIM was found to activate the SRC-FAK-PYK2-CREB cascade via TLR2 to recruit CBP/P300 at the promoters of miR-150 and miR-143 and epigenetically induce their expression. Loss-of-function studies utilizing specific miRNA inhibitors establish that Ac2PIM, via the miRNAs, abrogate NOD2-induced PI3K-PKCδ-MAPK pathway to suppress ß-catenin-mediated expression of COX-2, SOCS-3, and MMP-9. Our investigation has thus underscored the negative regulatory role of Ac2PIM-TLR2 signaling on NOD2 pathway which could broaden our understanding on vaccine potential or adjuvant utilities of Ac2PIM and/or MDP.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Animals , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/pharmacology , Epigenesis, Genetic , Immunity, Innate , Immunologic Factors/pharmacology , MAP Kinase Kinase Kinases/genetics , Macrophages/cytology , Macrophages/drug effects , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Mitogen-Activated Protein Kinases , Nitric Oxide/metabolism , Nod2 Signaling Adaptor Protein/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Polysaccharides, Bacterial/pharmacology , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Suppressor of Cytokine Signaling Proteins/pharmacology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism
4.
J Biol Chem ; 288(46): 33037-48, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24092752

ABSTRACT

Inflammatory bowel disease (IBD) is a debilitating chronic inflammatory disorder of the intestine. The interactions between enteric bacteria and genetic susceptibilities are major contributors of IBD etiology. Although genetic variants with loss or gain of NOD2 functions have been linked to IBD susceptibility, the mechanisms coordinating NOD2 downstream signaling, especially in macrophages, during IBD pathogenesis are not precisely identified. Here, studies utilizing the murine dextran sodium sulfate model of colitis revealed the crucial roles for inducible nitric-oxide synthase (iNOS) in regulating pathophysiology of IBDs. Importantly, stimulation of NOD2 failed to activate Sonic hedgehog (SHH) signaling in iNOS null macrophages, implicating NO mediated cross-talk between NOD2 and SHH signaling. NOD2 signaling up-regulated the expression of a NO-responsive microRNA, miR-146a, that targeted NUMB gene and alleviated the suppression of SHH signaling. In vivo and ex vivo studies confirmed the important roles for miR-146a in amplifying inflammatory responses. Collectively, we have identified new roles for miR-146a that established novel cross-talk between NOD2-SHH signaling during gut inflammation. Potential implications of these observations in therapeutics could increase the possibility of defining and developing better regimes to treat IBD pathophysiology.


Subject(s)
Hedgehog Proteins/metabolism , Inflammatory Bowel Diseases/metabolism , MicroRNAs/metabolism , Nitric Oxide/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Signal Transduction , Animals , Dextran Sulfate/toxicity , Disease Models, Animal , Hedgehog Proteins/genetics , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Mice , Mice, Knockout , MicroRNAs/genetics , Nitric Oxide/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nod2 Signaling Adaptor Protein/genetics
5.
J Biol Chem ; 288(28): 20592-606, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23733190

ABSTRACT

Pathogenic mycobacteria employ several immune evasion strategies such as inhibition of class II transactivator (CIITA) and MHC-II expression, to survive and persist in host macrophages. However, precise roles for specific signaling components executing down-regulation of CIITA/MHC-II have not been adequately addressed. Here, we demonstrate that Mycobacterium bovis bacillus Calmette-Guérin (BCG)-mediated TLR2 signaling-induced iNOS/NO expression is obligatory for the suppression of IFN-γ-induced CIITA/MHC-II functions. Significantly, NOTCH/PKC/MAPK-triggered signaling cross-talk was found critical for iNOS/NO production. NO responsive recruitment of a bifunctional transcription factor, KLF4, to the promoter of CIITA during M. bovis BCG infection of macrophages was essential to orchestrate the epigenetic modifications mediated by histone methyltransferase EZH2 or miR-150 and thus calibrate CIITA/MHC-II expression. NO-dependent KLF4 regulated the processing and presentation of ovalbumin by infected macrophages to reactive T cells. Altogether, our study delineates a novel role for iNOS/NO/KLF4 in dictating the mycobacterial capacity to inhibit CIITA/MHC-II-mediated antigen presentation by infected macrophages and thereby elude immune surveillance.


Subject(s)
Histocompatibility Antigens Class II/immunology , Kruppel-Like Transcription Factors/immunology , Nitric Oxide/immunology , Nuclear Proteins/immunology , Trans-Activators/immunology , Tuberculosis/immunology , Animals , Cell Line , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic/immunology , Gene Expression/drug effects , Gene Expression/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunoblotting , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/immunology , MicroRNAs/metabolism , Microscopy, Confocal , Mycobacterium bovis/immunology , Mycobacterium bovis/physiology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type II/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/immunology , Polycomb Repressive Complex 2/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Tuberculosis/microbiology , Tuberculosis/veterinary
6.
Mol Cancer ; 13: 210, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25208737

ABSTRACT

BACKGROUND: Increased incidence of lung cancer among pulmonary tuberculosis patients suggests mycobacteria-induced tumorigenic response in the host. The alveolar epithelial cells, candidate cells that form lung adenocarcinoma, constitute a niche for mycobacterial replication and infection. We thus explored the possible mechanism of M. bovis Bacillus Calmette-Guérin (BCG)-assisted tumorigenicity in type II epithelial cells, human lung adenocarcinoma A549 and other cancer cells. METHODS: Cancer cell lines originating from lung, colon, bladder, liver, breast, skin and cervix were treated with tumor necrosis factor (TNF)-α in presence or absence of BCG infection. p53, COP1 and sonic hedgehog (SHH) signaling markers were determined by immunoblotting and luciferase assays, and quantitative real time PCR was done for p53-responsive pro-apoptotic genes and SHH signaling markers. MTT assays and Annexin V staining were utilized to study apoptosis. Gain- and loss-of-function approaches were used to investigate the role for SHH and COP1 signaling during apoptosis. A549 xenografted mice were used to validate the contribution of BCG during TNF-α treatment. RESULTS: Here, we show that BCG inhibits TNF-α-mediated apoptosis in A549 cells via downregulation of p53 expression. Substantiating this observation, BCG rescued A549 xenografts from TNF-α-mediated tumor clearance in nude mice. Furthermore, activation of SHH signaling by BCG induced the expression of an E3 ubiquitin ligase, COP1. SHH-driven COP1 targeted p53, thereby facilitating downregulation of p53-responsive pro-apoptotic genes and inhibition of apoptosis. Similar effects of BCG could be shown for HCT116, T24, MNT-1, HepG2 and HELA cells but not for HCT116 p53(-/-) and MDA-MB-231 cells. CONCLUSION: Our results not only highlight possible explanations for the coexistence of pulmonary tuberculosis and lung cancer but also address probable reasons for failure of BCG immunotherapy of cancers.


Subject(s)
Apoptosis , Mycobacterium bovis/physiology , Neoplasms/pathology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HeLa Cells , Hep G2 Cells , Humans , Mice , Mice, Nude , Neoplasms/microbiology , Neoplasms, Experimental , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
7.
J Biol Chem ; 286(36): 31347-60, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21768114

ABSTRACT

Dendritic cells (DCs) as sentinels of the immune system are important for eliciting both primary and secondary immune responses to a plethora of microbial pathogens. Cooperative stimulation of a complex set of pattern-recognition receptors, including TLR2 and nucleotide-binding oligomerization domain (NOD)-like receptors on DCs, acts as a rate-limiting factor in determining the initiation and mounting of the robust immune response. It underscores the need for "decoding" these multiple receptor interactions. In this study, we demonstrate that TLR2 and NOD receptors cooperatively regulate functional maturation of human DCs. Intriguingly, synergistic stimulation of TLR2 and NOD receptors renders enhanced refractoriness to TGF-ß- or CTLA-4-mediated impairment of human DC maturation. Signaling perturbation data suggest that NOTCH1-PI3K signaling dynamics assume critical importance in TLR2- and NOD receptor-mediated surmounting of CTLA-4- and TGF-ß-suppressed maturation of human DCs. Interestingly, the NOTCH1-PI3K signaling axis holds the capacity to regulate DC functions by virtue of PKCδ-MAPK-dependent activation of NF-κB. This study provides mechanistic and functional insights into TLR2- and NOD receptor-mediated regulation of DC functions and unravels NOTCH1-PI3K as a signaling cohort for TLR2 and NOD receptors. These findings serve in building a conceptual foundation for the design of improved strategies for adjuvants and immunotherapies against infectious diseases.


Subject(s)
Dendritic Cells/cytology , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Receptor, Notch1/metabolism , Signal Transduction/immunology , Toll-Like Receptor 2/metabolism , CTLA-4 Antigen/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Humans , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Phosphatidylinositol 3-Kinase/immunology , Receptor, Notch1/immunology , Toll-Like Receptor 2/immunology , Transforming Growth Factor beta/immunology
8.
J Biol Chem ; 285(47): 36511-22, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20837474

ABSTRACT

Mycobacterium tuberculosis, an etiological agent of pulmonary tuberculosis, causes significant morbidity and mortality worldwide. Pathogenic mycobacteria survive in the host by subverting host innate immunity. Dendritic cells (DCs) are professional antigen-presenting cells that are vital for eliciting immune responses to infectious agents, including pathogenic mycobacteria. DCs orchestrate distinct Th responses based on the signals they receive. In this perspective, deciphering the interactions of the proline-glutamic acid/proline-proline-glutamic acid (PE/PPE) family of proteins of M. tuberculosis with DCs assumes significant pathophysiological attributes. In this study, we demonstrate that Rv1917c (PPE34), a representative member of the proline-proline-glutamic-major polymorphic tandem repeat family, interacts with TLR2 and triggers functional maturation of human DCs. Signaling perturbations implicated a critical role for integrated cross-talk among PI3K-MAPK and NF-κB signaling cascades in Rv1917c-induced maturation of DCs. However, this maturation of DCs was associated with a secretion of high amounts of anti-inflammatory cytokine IL-10, whereas Th1-polarizing cytokine IL-12 was not induced. Consistent with these results, Rv1917c-matured DCs favored secretion of IL-4, IL-5, and IL-10 from CD4(+) T cells and contributed to Th2-skewed cytokine balance ex vivo in healthy individuals and in patients with pulmonary tuberculosis. Interestingly, the Rv1917c-skewed Th2 immune response involved induced expression of cyclooxygenase-2 (COX-2) in DCs. Taken together, these results indicate that Rv1917c facilitates a shift in the ensuing immunity toward the Th2 phenotype and could aid in immune evasion by mycobacteria.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mycobacterium tuberculosis/metabolism , NF-kappa B/metabolism , Th2 Cells/immunology , Tuberculosis/metabolism , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Blotting, Western , Cells, Cultured , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation , Humans , Mycobacterium tuberculosis/immunology , NF-kappa B/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes , Th2 Cells/metabolism , Tuberculosis/immunology , Tuberculosis/microbiology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , src Homology Domains
9.
PLoS One ; 10(8): e0135842, 2015.
Article in English | MEDLINE | ID: mdl-26317499

ABSTRACT

Obesity-induced inflammation in visceral adipose tissue (VAT) is a major contributor to insulin resistance and type 2 diabetes. Whereas innate immune cells, notably macrophages, contribute to visceral adipose tissue (VAT) inflammation and insulin resistance, the role of adaptive immunity is less well defined. To address this critical gap, we used a model in which endogenous activation of T cells was suppressed in obese mice by blocking MyD88-mediated maturation of CD11c+ antigen-presenting cells. VAT CD11c+ cells from Cd11cCre+Myd88fl/fl vs. control Myd88fl/fl mice were defective in activating T cells in vitro, and VAT T and B cell activation was markedly reduced in Cd11cCre+Myd88fl/fl obese mice. However, neither macrophage-mediated VAT inflammation nor systemic inflammation were altered in Cd11cCre+Myd88fl/fl mice, thereby enabling a focused analysis on adaptive immunity. Unexpectedly, fasting blood glucose, plasma insulin, and the glucose response to glucose and insulin were completely unaltered in Cd11cCre+Myd88fl/fl vs. control obese mice. Thus, CD11c+ cells activate VAT T and B cells in obese mice, but suppression of this process does not have a discernible effect on macrophage-mediated VAT inflammation or systemic glucose homeostasis.


Subject(s)
Immunity, Innate , Insulin Resistance , Intra-Abdominal Fat/immunology , Lymphocyte Activation , Macrophages/immunology , Obesity/immunology , Animals , CD11 Antigens/genetics , CD11 Antigens/metabolism , Intra-Abdominal Fat/cytology , Lymphocyte Subsets/immunology , Male , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Obesity/metabolism
10.
Mol Cell Biol ; 33(3): 543-56, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23166298

ABSTRACT

Hedgehog (HH) signaling is a significant regulator of cell fate decisions during embryogenesis, development, and perpetuation of various disease conditions. Testing whether pathogen-specific HH signaling promotes unique innate recognition of intracellular bacteria, we demonstrate that among diverse Gram-positive or Gram-negative microbes, Mycobacterium bovis BCG, a vaccine strain, elicits a robust activation of Sonic HH (SHH) signaling in macrophages. Interestingly, sustained tumor necrosis factor alpha (TNF-α) secretion by macrophages was essential for robust SHH activation, as TNF-α(-/-) macrophages exhibited compromised ability to activate SHH signaling. Neutralization of TNF-α or blockade of TNF-α receptor signaling significantly reduced the infection-induced SHH signaling activation both in vitro and in vivo. Intriguingly, activated SHH signaling downregulated M. bovis BCG-mediated Toll-like receptor 2 (TLR2) signaling events to regulate a battery of genes associated with divergent functions of M1/M2 macrophages. Genome-wide expression profiling as well as conventional gain-of-function or loss-of-function analysis showed that SHH signaling-responsive microRNA 31 (miR-31) and miR-150 target MyD88, an adaptor protein of TLR2 signaling, thus leading to suppression of TLR2 responses. SHH signaling signatures could be detected in vivo in tuberculosis patients and M. bovis BCG-challenged mice. Collectively, these investigations identify SHH signaling to be what we believe is one of the significant regulators of host-pathogen interactions.


Subject(s)
Hedgehog Proteins/immunology , Host-Pathogen Interactions , MicroRNAs/immunology , Mycobacterium bovis/physiology , Toll-Like Receptor 2/immunology , Tuberculosis/immunology , Animals , Cell Line , Cells, Cultured , Gene Expression Regulation , Humans , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Myeloid Differentiation Factor 88/genetics , Signal Transduction , Transcriptome , Tuberculosis/genetics , Tuberculosis/veterinary , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
11.
Mol Cell Biol ; 32(12): 2239-53, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22473996

ABSTRACT

Pathogenic mycobacteria, including Mycobacterium tuberculosis and Mycobacterium bovis, cause significant morbidity and mortality worldwide. However, the vaccine strain Mycobacterium bovis BCG, unlike virulent strains, triggers extensive apoptosis of infected macrophages, a step necessary for the elicitation of robust protective immunity. We here demonstrate that M. bovis BCG triggers Toll-like receptor 2 (TLR2)-dependent microRNA-155 (miR-155) expression, which involves signaling cross talk among phosphatidylinositol 3-kinase (PI3K), protein kinase Cδ (PKCδ), and mitogen-activated protein kinases (MAPKs) and recruitment of NF-κB and c-ETS to miR-155 promoter. Genetic and signaling perturbations presented the evidence that miR-155 regulates PKA signaling by directly targeting a negative regulator of PKA, protein kinase inhibitor alpha (PKI-α). Enhanced activation of PKA signaling resulted in the generation of PKA C-α; phosphorylation of MSK1, cyclic AMP response element binding protein (CREB), and histone H3; and recruitment of phospho-CREB to the apoptotic gene promoters. The miR-155-triggered activation of caspase-3, BAK1, and cytochrome c translocation involved signaling integration of MAPKs and epigenetic or posttranslational modification of histones or CREB. Importantly, M. bovis BCG infection-induced apoptosis was severely compromised in macrophages derived from miR-155 knockout mice. Gain-of-function and loss-of-function studies validated the requirement of miR-155 for M. bovis BCG's ability to trigger apoptosis. Overall, M. bovis BCG-driven miR-155 dictates cell fate decisions of infected macrophages, strongly implicating a novel role for miR-155 in orchestrating cellular reprogramming during immune responses to mycobacterial infection.


Subject(s)
Macrophages/metabolism , Macrophages/microbiology , MicroRNAs/metabolism , Mycobacterium bovis , Animals , Apoptosis/immunology , Bacterial Vaccines , Cells, Cultured , Macrophages/immunology , Mice , MicroRNAs/immunology , Mycobacterium bovis/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Signal Transduction , Toll-Like Receptor 2/metabolism , Transcription Factor RelA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL