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










Publication year range
1.
Commun Biol ; 4(1): 1405, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34916605

ABSTRACT

Though various transgene expression switches have been adopted in a wide variety of organisms for basic and biomedical research, intrinsic obstacles of those existing systems, including toxicity and silencing, have been limiting their use in vertebrate transgenesis. Here we demonstrate a novel QF-based binary transgene switch (IQ-Switch) that is relatively free of driver toxicity and transgene silencing, and exhibits potent and highly tunable transgene activation by the chemical inducer tebufenozide, a non-toxic lipophilic molecule to developing zebrafish with negligible background. The interchangeable IQ-Switch makes it possible to elicit ubiquitous and tissue specific transgene expression in a spatiotemporal manner. We generated a RASopathy disease model using IQ-Switch and demonstrated that the RASopathy symptoms were ameliorated by the specific BRAF(V600E) inhibitor vemurafenib, validating the therapeutic use of the gene switch. The orthogonal IQ-Switch provides a state-of-the-art platform for flexible regulation of transgene expression in zebrafish, potentially applicable in cell-based systems and other model organisms.


Subject(s)
Animals, Genetically Modified/genetics , Gene Transfer Techniques , Genes, Switch , Transgenes , Zebrafish/genetics , Animals
2.
Clin Epigenetics ; 12(1): 66, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32398127

ABSTRACT

BACKGROUND: Atherosclerosis is the main cause of cardiovascular diseases such as ischemic stroke and coronary heart disease. Gene-specific promoter methylation changes have been suggested as one of the causes underlying the development of atherosclerosis. We aimed to identify and validate specific genes that are differentially expressed through promoter methylation in atherosclerotic plaques. We performed the present study in four steps: (1) profiling and identification of gene-specific promoter methylation changes in atherosclerotic tissues; (2) validation of the promoter methylation changes of genes in plaques by comparison with non-plaque intima; (3) evaluation of promoter methylation status of the genes in vascular cellular components composing atherosclerotic plaques; and (4) evaluation of promoter methylation differences in genes among monocytes, T cells, and B cells isolated from the blood of ischemic stroke patients. RESULTS: Upon profiling, AIRE1, ALOX12, FANK1, NETO1, and SERHL2 were found to have displayed changes in promoter methylation. Of these, AIRE1 and ALOX12 displayed higher methylation levels in plaques than in non-plaque intima, but lower than those in the buffy coat of blood. Between inflammatory cells, the three genes were significantly less methylated in monocytes than in T and B cells. In the vascular cells, AIRE1 methylation was lower in endothelial and smooth muscle cells. ALOX12 methylation was higher in endothelial, but lower in smooth muscle cells. Immunofluorescence staining showed that co-localization of ALOX12 and AIRE1 was more frequent in CD14(+)-monocytes than in CD4(+)-T cell in plaque than in non-plaque intima. CONCLUSIONS: Promoter methylation changes in AIRE1 and ALOX12 occur in atherosclerosis and can be considered as novel epigenetic markers.


Subject(s)
Arachidonate 12-Lipoxygenase/genetics , Atherosclerosis/genetics , Epigenesis, Genetic , Transcription Factors/genetics , Atherosclerosis/metabolism , Biomarkers/metabolism , DNA Methylation , Endothelium, Vascular/metabolism , Lymphocytes/metabolism , Monocytes/metabolism , Muscle, Smooth, Vascular/metabolism , Plaque, Atherosclerotic/genetics , Promoter Regions, Genetic , AIRE Protein
3.
Nat Commun ; 9(1): 4184, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30305619

ABSTRACT

Gamma-aminobutyric acid (GABA) is the principal inhibitory neurotransmitter in the brain; however, the roles of GABA in antimicrobial host defenses are largely unknown. Here we demonstrate that GABAergic activation enhances antimicrobial responses against intracellular bacterial infection. Intracellular bacterial infection decreases GABA levels in vitro in macrophages and in vivo in sera. Treatment of macrophages with GABA or GABAergic drugs promotes autophagy activation, enhances phagosomal maturation and antimicrobial responses against mycobacterial infection. In macrophages, the GABAergic defense is mediated via macrophage type A GABA receptor (GABAAR), intracellular calcium release, and the GABA type A receptor-associated protein-like 1 (GABARAPL1; an Atg8 homolog). Finally, GABAergic inhibition increases bacterial loads in mice and zebrafish in vivo, suggesting that the GABAergic defense plays an essential function in metazoan host defenses. Our study identified a previously unappreciated role for GABAergic signaling in linking antibacterial autophagy to enhance host innate defense against intracellular bacterial infection.


Subject(s)
Autophagy , Bacterial Infections/metabolism , Bacterial Infections/pathology , Host-Pathogen Interactions , Signal Transduction , gamma-Aminobutyric Acid/metabolism , Adenylate Kinase/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Autophagy/drug effects , Calcium/metabolism , Host-Pathogen Interactions/drug effects , Humans , Macrophages/metabolism , Macrophages/ultrastructure , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Mycobacterium tuberculosis/drug effects , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/ultrastructure , Receptors, GABA/metabolism , Signal Transduction/drug effects
4.
Sci Rep ; 7(1): 3431, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28611371

ABSTRACT

The induction of host cell autophagy by various autophagy inducers contributes to the antimicrobial host defense against Mycobacterium tuberculosis (Mtb), a major pathogenic strain that causes human tuberculosis. In this study, we present a role for the newly identified cyclic peptides ohmyungsamycins (OMS) A and B in the antimicrobial responses against Mtb infections by activating autophagy in murine bone marrow-derived macrophages (BMDMs). OMS robustly activated autophagy, which was essentially required for the colocalization of LC3 autophagosomes with bacterial phagosomes and antimicrobial responses against Mtb in BMDMs. Using a Drosophila melanogaster-Mycobacterium marinum infection model, we showed that OMS-A-induced autophagy contributed to the increased survival of infected flies and the limitation of bacterial load. We further showed that OMS triggered AMP-activated protein kinase (AMPK) activation, which was required for OMS-mediated phagosome maturation and antimicrobial responses against Mtb. Moreover, treating BMDMs with OMS led to dose-dependent inhibition of macrophage inflammatory responses, which was also dependent on AMPK activation. Collectively, these data show that OMS is a promising candidate for new anti-mycobacterial therapeutics by activating antibacterial autophagy via AMPK-dependent signaling and suppressing excessive inflammation during Mtb infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Autophagy , Mycobacterium Infections/drug therapy , Peptides, Cyclic/pharmacology , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Anti-Bacterial Agents/therapeutic use , Cells, Cultured , Humans , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/pathogenicity , Peptides, Cyclic/therapeutic use , Streptomyces/drug effects , Streptomyces/pathogenicity
5.
Immune Netw ; 17(2): 77-88, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28458619

ABSTRACT

Mitochondria are key organelles involved in energy production, functioning as the metabolic hubs of cells. Recent findings emphasize the emerging role of the mitochondrion as a key intracellular signaling platform regulating innate immune and inflammatory responses. Several mitochondrial proteins and mitochondrial reactive oxygen species have emerged as central players orchestrating the innate immune responses to pathogens and damaging ligands. This review explores our current understanding of the roles played by mitochondria in regulation of innate immunity and inflammatory responses. Recent advances in our understanding of the relationship between autophagy, mitochondria, and inflammasome activation are also briefly discussed. A comprehensive understanding of mitochondrial role in toll-like receptor-mediated innate immune responses and NLRP3 inflammasome complex activation, will facilitate development of novel therapeutics to treat various infectious, inflammatory, and autoimmune disorders.

6.
J Immunol ; 198(8): 3283-3295, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28275133

ABSTRACT

The role of peroxisome proliferator-activated receptor α (PPAR-α) in innate host defense is largely unknown. In this study, we show that PPAR-α is essential for antimycobacterial responses via activation of transcription factor EB (TFEB) transcription and inhibition of lipid body formation. PPAR-α deficiency resulted in an increased bacterial load and exaggerated inflammatory responses during mycobacterial infection. PPAR-α agonists promoted autophagy, lysosomal biogenesis, phagosomal maturation, and antimicrobial defense against Mycobacterium tuberculosis or M. bovis bacillus Calmette-Guérin. PPAR-α agonists regulated multiple genes involved in autophagy and lysosomal biogenesis, including Lamp2, Rab7, and Tfeb in bone marrow-derived macrophages. Silencing of TFEB reduced phagosomal maturation and antimicrobial responses, but increased macrophage inflammatory responses during mycobacterial infection. Moreover, PPAR-α activation promoted lipid catabolism and fatty acid ß-oxidation in macrophages during mycobacterial infection. Taken together, our data indicate that PPAR-α mediates antimicrobial responses to mycobacterial infection by inducing TFEB and lipid catabolism.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , Immunity, Innate/immunology , Lipid Metabolism/immunology , Mycobacterium Infections/immunology , PPAR alpha/immunology , Animals , Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Immunoblotting , Immunohistochemistry , Lipid Droplets/immunology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium , PPAR alpha/metabolism , Polymerase Chain Reaction
7.
Microbes Infect ; 19(6): 351-357, 2017 06.
Article in English | MEDLINE | ID: mdl-28245983

ABSTRACT

Mycobacterium marinum is a pathogenic mycobacterial species closely related to Mycobacterium tuberculosis. In this study, we established a mycobacterial infection model of Drosophila melanogaster to characterize the role played by cg6568, a homolog of the human cathelicidin gene, in the innate defense against infection. Drosophila cg6568 was expressed at various levels during all developmental stages, and the expression levels were modulated by M. marinum in a time-dependent manner. 20-hydroxyecdysone induced Drosophila cg6568 transcription both in vitro and in vivo. Using flies expressing cg6568 RNAi, we found that cg6568 was essential both for D. melanogaster survival and the exertion of antimicrobial effects during M. marinum infection. Thus, we named the gene product a cathelicidin-like antimicrobial protein of D. melanogaster (dCAMP). Our results indicate that dCAMP is crucial in terms of the innate D. melanogaster defense during M. marinum infection.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/microbiology , Host-Pathogen Interactions , Mycobacterium marinum , Animals , Antimicrobial Cationic Peptides/genetics , Disease Models, Animal , Drosophila Proteins/genetics , Ecdysterone/pharmacology , Gene Expression Regulation , Immunity, Innate , Male , Mycobacterium Infections, Nontuberculous/immunology , RNA Interference , Cathelicidins
8.
Immunol Cell Biol ; 95(7): 584-592, 2017 08.
Article in English | MEDLINE | ID: mdl-28356568

ABSTRACT

Inflammasomes are cytosolic multiprotein complexes that cause the release of biologically active interleukin-1ß. The best-characterized inflammasome is the NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 or Nod-like receptor protein 3) inflammasome. The NLRP3 inflammasome forms an assembly consisting of the ASC (apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain) adaptor protein and the effector, caspase-1 (cysteine-dependent aspartate-directed protease-1). Numerous agents and ligands derived from pathogens, modified self-cells and the environment induce NLRP3 inflammasome complex formation. NLRP3 inflammasome activation is tightly controlled at the transcriptional and post-translational levels to prevent unwanted excessive inflammation. Recent studies have highlighted the roles and mechanisms of several negative regulators that inhibit the assembly of NLRP3 inflammasome complexes and suppress inflammatory responses. The identification and characterization of new players in the regulation of NLRP3 inflammasome may lead to the development of inflammasome-targeting therapeutics against various inflammatory diseases related to NLRP3 inflammasome-associated pathogenesis.


Subject(s)
Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction , Animals , Humans , Models, Biological
9.
Radiat Res ; 187(1): 32-41, 2017 01.
Article in English | MEDLINE | ID: mdl-28001907

ABSTRACT

During radiotherapy for tumors, the innate immune system also responds to ionizing radiation and induces immune modulation. However, little is known about the molecular mechanisms by which radiation modulates innate immune responses. In this study, we observed that radiation triggered the generation of mitochondrial reactive oxygen species (mROS), leading to innate immune responses in murine bone marrow-derived macrophages (BMDM). Radiation-induced mROS was essential for robust induction of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and IL-12p40 mRNA and protein in BMDM. Exposure to radiation also led to rapid activation of the mitogen-activated protein kinase (MAPK) and nuclear factor (NF)-κB pathways in BMDM. Notably, radiation-induced MAPK activation and NF-κB signaling were regulated by mROS in macrophages. Additionally, radiation-induced expression of TNF-α, IL-6 and IL-12p40 was dependent on JNK, p38 and NF-κB activation in BMDM. These data suggest a key role for radiation-induced pro-inflammatory responses and activation of the MAPK and NF-κB pathways through a triggering mechanism involving mROS generation.


Subject(s)
Macrophages/immunology , Macrophages/radiation effects , Mitochondria/metabolism , Mitochondria/radiation effects , Animals , Bone Marrow Cells/cytology , Enzyme Activation/radiation effects , Female , Gene Expression Regulation/radiation effects , Interleukin-1beta/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/radiation effects , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Microbes Infect ; 19(1): 5-17, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27637463

ABSTRACT

Mycobacterial ESX systems are often related to pathogenesis during infection. However, little is known about the function of ESX systems of Mycobacterium abscessus (Mab). This study focuses on the Mab ESX-3 cluster, which contains major genes such as esxH (Rv0288, low molecular weight protein antigen 7; CFP-7) and esxG (Rv0287, ESAT-6 like protein). An esx-3 (MAB 2224c-2234c)-deletional mutant of Mab (Δesx) was constructed and used to infect murine and human macrophages. We then investigated whether Mab Δesx modulated innate host immune responses in macrophages. Mab Δesx infection resulted in less pathological and inflammatory responses. Additionally, Δesx resulted in significantly decreased activation of inflammatory signaling and cytokine production in macrophages compared to WT. Moreover, recombinant EsxG·EsxH (rEsxGH) proteins encoded by the ESX-3 region showed synergistic enhancement of inflammatory cytokine generation in macrophages infected with Δesx. Taken together, our data suggest that Mab ESX-3 plays an important role in inflammatory and pathological responses during Mab infection.


Subject(s)
Bacterial Proteins/metabolism , Multigene Family , Mycobacterium/pathogenicity , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Cytokines/metabolism , Female , Gene Deletion , Healthy Volunteers , Humans , Immunity, Innate , Macrophage Activation , Macrophages/immunology , Macrophages/microbiology , Mice, Inbred C57BL , Mycobacterium/genetics , Virulence Factors/genetics
11.
Arch Pharm Res ; 39(11): 1491-1502, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27699647

ABSTRACT

Innate immunity constitutes the first line of defense against pathogenic and dangerous insults. However, it is a double-edged sword, as it functions in both clearance of infection and inflammatory damage. It is therefore important that innate immune responses are tightly controlled to prevent harmful excessive inflammation. Nuclear receptors (NRs) are a family of transcription factors that play critical roles in various physiological responses. Orphan NRs are a subset of NRs for which the ligands and functions are unclear. Accumulating evidence has revealed that orphan NRs play essential roles in innate immune responses to prevent pathogenic inflammatory responses and to enhance antimicrobial host defenses. In this review, we describe current knowledge on the roles and mechanisms of orphan NRs in the regulation of innate immune responses. Discovery of new functions of orphan NRs would facilitate development of novel preventive and therapeutic strategies against human inflammatory diseases.


Subject(s)
Immunity, Innate/physiology , Orphan Nuclear Receptors/physiology , Animals , Bacterial Infections/immunology , Bacterial Infections/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Ligands , Orphan Nuclear Receptors/chemistry , Orphan Nuclear Receptors/genetics , Protein Processing, Post-Translational , Signal Transduction
12.
Endocrinol Metab (Seoul) ; 31(1): 17-24, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26754583

ABSTRACT

The nuclear receptor superfamily consists of the steroid and non-steroid hormone receptors and the orphan nuclear receptors. Small heterodimer partner (SHP) is an orphan family nuclear receptor that plays an essential role in the regulation of glucose and cholesterol metabolism. Recent studies reported a previously unidentified role for SHP in the regulation of innate immunity and inflammation. The innate immune system has a critical function in the initial response against a variety of microbial and danger signals. Activation of the innate immune response results in the induction of inflammatory cytokines and chemokines to promote anti-microbial effects. An excessive or uncontrolled inflammatory response is potentially harmful to the host, and can cause tissue damage or pathological threat. Therefore, the innate immune response should be tightly regulated to enhance host defense while preventing unwanted immune pathologic responses. In this review, we discuss recent studies showing that SHP is involved in the negative regulation of toll-like receptor-induced and NLRP3 (NACHT, LRR and PYD domains-containing protein 3)-mediated inflammatory responses in innate immune cells. Understanding the function of SHP in innate immune cells will allow us to prevent or modulate acute and chronic inflammation processes in cases where dysregulated innate immune activation results in damage to normal tissues.

13.
Immunity ; 43(1): 80-91, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26200012

ABSTRACT

The orphan nuclear receptor estrogen-related receptor α (ERRα; NR3B1) is a key metabolic regulator, but its function in regulating inflammation remains largely unknown. Here, we demonstrate that ERRα negatively regulates Toll-like receptor (TLR)-induced inflammation by promoting Tnfaip3 transcription and fine-tuning of metabolic reprogramming in macrophages. ERRα-deficient (Esrra(-/-)) mice showed increased susceptibility to endotoxin-induced septic shock, leading to more severe pro-inflammatory responses than control mice. ERRα regulated macrophage inflammatory responses by directly binding the promoter region of Tnfaip3, a deubiquitinating enzyme in TLR signaling. In addition, Esrra(-/-) macrophages showed an increased glycolysis, but impaired mitochondrial respiratory function and biogenesis. Further, ERRα was required for the regulation of NF-κB signaling by controlling p65 acetylation via maintenance of NAD(+) levels and sirtuin 1 activation. These findings unravel a previously unappreciated role for ERRα as a negative regulator of TLR-induced inflammatory responses through inducing Tnfaip3 transcription and controlling the metabolic reprogramming.


Subject(s)
Cysteine Endopeptidases/biosynthesis , Inflammation/immunology , Intracellular Signaling Peptides and Proteins/biosynthesis , Macrophages/metabolism , Receptors, Estrogen/genetics , Toll-Like Receptor 4/immunology , Acetylation , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cells, Cultured , Cysteine Endopeptidases/genetics , Enzyme Activation/genetics , Glycolysis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides , Macrophages/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , NAD/metabolism , Oxidative Phosphorylation , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Shock, Septic/immunology , Signal Transduction , Sirtuin 1/metabolism , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor RelA/metabolism , Transcription, Genetic/genetics , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitination , ERRalpha Estrogen-Related Receptor
14.
J Immunol ; 194(11): 5355-65, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25917095

ABSTRACT

MicroRNAs (miRNAs) are small noncoding nucleotides that play critical roles in the regulation of diverse biological functions, including the response of host immune cells. Autophagy plays a key role in activating the antimicrobial host defense against Mycobacterium tuberculosis. Although the pathways associated with autophagy must be tightly regulated at a posttranscriptional level, the contribution of miRNAs and whether they specifically influence the activation of macrophage autophagy during M. tuberculosis infection are largely unknown. In this study, we demonstrate that M. tuberculosis infection of macrophages leads to increased expression of miRNA-125a-3p (miR-125a), which targets UV radiation resistance-associated gene (UVRAG), to inhibit autophagy activation and antimicrobial responses to M. tuberculosis. Forced expression of miR-125a significantly blocked M. tuberculosis-induced activation of autophagy and phagosomal maturation in macrophages, and inhibitors of miR-125a counteracted these effects. Both TLR2 and MyD88 were required for biogenesis of miR-125a during M. tuberculosis infection. Notably, activation of the AMP-activated protein kinase significantly inhibited the expression of miR-125a in M. tuberculosis-infected macrophages. Moreover, either overexpression of miR-125a or silencing of UVRAG significantly attenuated the antimicrobial effects of macrophages against M. tuberculosis. Taken together, these data indicate that miR-125a regulates the innate host defense by inhibiting the activation of autophagy and antimicrobial effects against M. tuberculosis through targeting UVRAG.


Subject(s)
Autophagy/genetics , Macrophages/immunology , MicroRNAs/physiology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Tumor Suppressor Proteins/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/immunology , Animals , Cell Line , Gene Expression Regulation , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Myeloid Differentiation Factor 88 , RNA Interference , RNA, Small Interfering , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 2 , Tuberculosis, Pulmonary/genetics
15.
Nat Commun ; 6: 6115, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25655831

ABSTRACT

Excessive activation of the NLRP3 inflammasome results in damaging inflammation, yet the regulators of this process remain poorly defined. Herein, we show that the orphan nuclear receptor small heterodimer partner (SHP) is a negative regulator of NLRP3 inflammasome activation. NLRP3 inflammasome activation leads to an interaction between SHP and NLRP3, proteins that are both recruited to mitochondria. Overexpression of SHP competitively inhibits binding of NLRP3 to apoptosis-associated speck-like protein containing a CARD (ASC). SHP deficiency results in increased secretion of proinflammatory cytokines IL-1ß and IL-18, and excessive pathologic responses typically observed in mouse models of kidney tubular necrosis and peritoneal gout. Notably, the loss of SHP results in accumulation of damaged mitochondria and a sustained interaction between NLRP3 and ASC in the endoplasmic reticulum. These data are suggestive of a role for SHP in controlling NLRP3 inflammasome activation through a mechanism involving interaction with NLRP3 and maintenance of mitochondrial homeostasis.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , Endoplasmic Reticulum/metabolism , Enzyme Activation , HEK293 Cells , Homeostasis , Humans , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Binding , Protein Transport , Receptors, Cytoplasmic and Nuclear/deficiency
16.
Autophagy ; 10(5): 785-802, 2014 May.
Article in English | MEDLINE | ID: mdl-24598403

ABSTRACT

AMP-activated protein kinase (AMPK) is a crucial energy sensor and plays a key role in integration of cellular functions to maintain homeostasis. Despite this, it is largely unknown whether targeting the AMPK pathway can be used as a therapeutic strategy for infectious diseases. Herein, we show that AMPK activation robustly induces antibacterial autophagy, which contributes to antimicrobial defense against Mycobacterium tuberculosis (Mtb). AMPK activation led to inhibition of Mtb-induced phosphorylation of the mechanistic target of rapamycin (MTOR) in macrophages. In addition, AMPK activation increased the genes involved in oxidative phosphorylation, mitochondrial ATP production, and biogenesis in Mtb-infected macrophages. Notably, peroxisome proliferator-activated receptor-gamma, coactivator 1α (PPARGC1A) was required for AMPK-mediated antimicrobial activity, as well as enhancement of mitochondrial function and biogenesis, in macrophages. Further, the AMPK-PPARGC1A pathway was involved in the upregulation of multiple autophagy-related genes via CCAAT/enhancer binding protein (C/EBP), ß (CEBPB). PPARGC1A knockdown inhibited the AMPK-mediated induction of autophagy and impaired the fusion of phagosomes with MAP1LC3B (LC3B) autophagosomes in Mtb-infected macrophages. The link between autophagy, mitochondrial function, and antimicrobial activity was further demonstrated by studying LysMCre-mediated knockout of atg7, demonstrating mitochondrial ultrastructural defects and dysfunction, as well as blockade of antimicrobial activity against mycobacteria. Collectively, our results identify the AMPK-PPARGC1A axis as contributing to autophagy activation leading to an antimicrobial response, as a novel host defense mechanism.


Subject(s)
AMP-Activated Protein Kinases/physiology , Autophagy/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate/genetics , Transcription Factors/physiology , Animals , Cells, Cultured , Drosophila melanogaster , Host-Pathogen Interactions/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mycobacterium/immunology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction/genetics , Signal Transduction/immunology
17.
Cell Host Microbe ; 11(5): 457-68, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22607799

ABSTRACT

The current standard of treatment against tuberculosis consists of a cocktail of first-line drugs, including isoniazid and pyrazinamide. Although these drugs are known to be bactericidal, contribution of host cell responses in the context of antimycobacterial chemotherapy, if any, remains unknown. We demonstrate that isoniazid and pyrazinamide promote autophagy activation and phagosomal maturation in Mycobacterium tuberculosis (Mtb)-infected host cells. Treatment of Mtb-infected macrophages with isoniazid or pyrazinamide caused significant activation of cellular and mitochondrial reactive oxygen species and autophagy, which was triggered by bacterial hydroxyl radical generation. Mycobacterium marinum-infected autophagy-defective, atg7 mutant Drosophila exhibited decreased survival rates, which could not be rescued by antimycobacterial treatment, indicating that autophagy is required for effective antimycobacterial drug action in vivo. Moreover, activation of autophagy by antibiotic treatment dampened Mtb-induced proinflammatory responses in macrophages. Together, these findings underscore the importance of host autophagy in orchestrating successful antimicrobial responses to mycobacteria during chemotherapy.


Subject(s)
Antitubercular Agents/pharmacology , Autophagy/immunology , Mycobacterium marinum/immunology , Mycobacterium tuberculosis/immunology , Animals , Autophagy-Related Protein 7 , Cells, Cultured , Drosophila , Drosophila Proteins/deficiency , Drosophila Proteins/immunology , Humans , Isoniazid/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Mice , Mycobacterium marinum/drug effects , Mycobacterium marinum/pathogenicity , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/pathogenicity , Phagosomes/drug effects , Phagosomes/metabolism , Pyrazinamide/pharmacology , Reactive Oxygen Species/metabolism , Survival Analysis
18.
Nat Immunol ; 12(8): 742-51, 2011 Jul 03.
Article in English | MEDLINE | ID: mdl-21725320

ABSTRACT

The orphan nuclear receptor SHP (small heterodimer partner) is a transcriptional corepressor that regulates hepatic metabolic pathways. Here we identified a role for SHP as an intrinsic negative regulator of Toll-like receptor (TLR)-triggered inflammatory responses. SHP-deficient mice were more susceptible to endotoxin-induced sepsis. SHP had dual regulatory functions in a canonical transcription factor NF-κB signaling pathway, acting as both a repressor of transactivation of the NF-κB subunit p65 and an inhibitor of polyubiquitination of the adaptor TRAF6. SHP-mediated inhibition of signaling via the TLR was mimicked by macrophage-stimulating protein (MSP), a strong inducer of SHP expression, via an AMP-activated protein kinase-dependent signaling pathway. Our data identify a previously unrecognized role for SHP in the regulation of TLR signaling.


Subject(s)
NF-kappa B/immunology , Receptors, Cytoplasmic and Nuclear/immunology , Sepsis/immunology , Toll-Like Receptors/immunology , AMP-Activated Protein Kinases/immunology , Animals , Chromatin Immunoprecipitation , Female , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , TNF Receptor-Associated Factor 6/immunology , Ubiquitination/immunology
19.
PLoS Pathog ; 6(12): e1001230, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21187903

ABSTRACT

The "enhanced intracellular survival" (eis) gene of Mycobacterium tuberculosis (Mtb) is involved in the intracellular survival of M. smegmatis. However, its exact effects on host cell function remain elusive. We herein report that Mtb Eis plays essential roles in modulating macrophage autophagy, inflammatory responses, and cell death via a reactive oxygen species (ROS)-dependent pathway. Macrophages infected with an Mtb eis-deletion mutant H37Rv (Mtb-Δeis) displayed markedly increased accumulation of massive autophagic vacuoles and formation of autophagosomes in vitro and in vivo. Infection of macrophages with Mtb-Δeis increased the production of tumor necrosis factor-α and interleukin-6 over the levels produced by infection with wild-type or complemented strains. Elevated ROS generation in macrophages infected with Mtb-Δeis (for which NADPH oxidase and mitochondria were largely responsible) rendered the cells highly sensitive to autophagy activation and cytokine production. Despite considerable activation of autophagy and proinflammatory responses, macrophages infected with Mtb-Δeis underwent caspase-independent cell death. This cell death was significantly inhibited by blockade of autophagy and c-Jun N-terminal kinase-ROS signaling, suggesting that excessive autophagy and oxidative stress are detrimental to cell survival. Finally, artificial over-expression of Eis or pretreatment with recombinant Eis abrogated production of both ROS and proinflammatory cytokines, which depends on the N-acetyltransferase domain of the Eis protein. Collectively, these data indicate that Mtb Eis suppresses host innate immune defenses by modulating autophagy, inflammation, and cell death in a redox-dependent manner.


Subject(s)
Antigens, Bacterial/physiology , Autophagy , Bacterial Proteins/physiology , Host-Pathogen Interactions/immunology , Inflammation , Mycobacterium tuberculosis/physiology , Signal Transduction/physiology , Acetyltransferases , Animals , Cell Death , Immunity, Innate , Macrophages/metabolism , Macrophages/microbiology , Mice , Mycobacterium tuberculosis/chemistry , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism
20.
Cell Host Microbe ; 6(3): 231-43, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19748465

ABSTRACT

Autophagy and vitamin D3-mediated innate immunity have been shown to confer protection against infection with intracellular Mycobacterium tuberculosis. Here, we show that these two antimycobacterial defenses are physiologically linked via a regulatory function of human cathelicidin (hCAP-18/LL-37), a member of the cathelicidin family of antimicrobial proteins. We show that 1,25-dihydroxyvitamin D3 (1,25D3), the active form of vitamin D, induced autophagy in human monocytes via cathelicidin, which activated transcription of the autophagy-related genes Beclin-1 and Atg5. 1,25D3 also induced the colocalization of mycobacterial phagosomes with autophagosomes in human macrophages in a cathelicidin-dependent manner. Furthermore, the antimycobacterial activity in human macrophages mediated by physiological levels of 1,25D3 required autophagy and cathelicidin. These results indicate that human cathelicidin, a protein that has direct antimicrobial activity, also serves as a mediator of vitamin D3-induced autophagy.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Autophagy/drug effects , Cholecalciferol/pharmacology , Macrophages/cytology , Monocytes/cytology , Tuberculosis/physiopathology , Antimicrobial Cationic Peptides/genetics , Cells, Cultured , Host-Pathogen Interactions , Humans , Macrophages/drug effects , Macrophages/microbiology , Monocytes/drug effects , Monocytes/microbiology , Mycobacterium tuberculosis/physiology , Tuberculosis/genetics , Tuberculosis/immunology , Tuberculosis/microbiology , Cathelicidins
SELECTION OF CITATIONS
SEARCH DETAIL
...