Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
EMBO Rep ; 22(2): e50613, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33345425

ABSTRACT

One major factor that contributes to the virulence of Pseudomonas aeruginosa is its ability to reside and replicate unchallenged inside airway epithelial cells. The mechanism by which P. aeruginosa escapes destruction by intracellular host defense mechanisms, such as autophagy, is not known. Here, we show that the type III secretion system effector protein ExoS facilitates P. aeruginosa survival in airway epithelial cells by inhibiting autophagy in host cells. Autophagy inhibition is independent of mTOR activity, as the latter is also inhibited by ExoS, albeit by a different mechanism. Deficiency of the critical autophagy gene Atg7 in airway epithelial cells, both in vitro and in mouse models, greatly enhances the survival of ExoS-deficient P. aeruginosa but does not affect the survival of ExoS-containing bacteria. The inhibitory effect of ExoS on autophagy and mTOR depends on the activity of its ADP-ribosyltransferase domain. Inhibition of mTOR is caused by ExoS-mediated ADP ribosylation of RAS, whereas autophagy inhibition is due to the suppression of autophagic Vps34 kinase activity.


Subject(s)
ADP Ribose Transferases , Bacterial Toxins , Pseudomonas aeruginosa , ADP Ribose Transferases/genetics , Animals , Autophagy , Mice , TOR Serine-Threonine Kinases/genetics
2.
J Immunol ; 207(2): 661-670, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34193605

ABSTRACT

Inflammation contributes to the pathogenesis and morbidity of wide spectrum of human diseases. The inflammatory response must be actively controlled to prevent bystander damage to tissues. Yet, the mechanisms controlling excessive inflammatory responses are poorly understood. NLRP3 inflammasome plays an important role in innate immune response to cellular infection or stress. Its activation must be tightly regulated because uncontrolled inflammasome activation is associated with a number of human diseases. p38 MAPK signaling plays an essential role in the regulation of inflammation. The role of p38 MAPK in inflammatory response associated with the expression of proinflammatory molecules is known. However, the anti-inflammatory functions of p38 MAPK are largely unknown. In this study, we show that pharmacologic inhibition or genetic deficiency of p38 MAPK leads to hyperactivation of NLRP3 inflammasome, resulting in enhanced Caspase 1 activation and IL-1ß and IL-18 production. The deficiency of p38 MAPK activity induced an increase of cytosolic Ca2+ and excessive mitochondrial Ca2+ uptake, leading to exacerbation of mitochondrial damage, which was associated with hyperactivation of NLRP3 inflammasome. In addition, mice with deficiency of p38 MAPK in granulocytes had evidence of in vivo hyperactivation of NLRP3 inflammasome and were more susceptible to LPS-induced sepsis compared with wild-type mice. Our results suggest that p38 MAPK negatively regulates NLRP3 inflammasome through control of Ca2+ mobilization. Hyperactivity of inflammasome in p38-deficient mice causes lung inflammation and increased susceptibility to septic shock.


Subject(s)
Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , HEK293 Cells , Humans , Immunity, Innate/physiology , Inflammation/metabolism , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Sepsis/metabolism , Shock, Septic/metabolism , Signal Transduction/physiology
3.
Immunity ; 39(3): 537-47, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24035364

ABSTRACT

Autophagy and phagocytosis are conserved cellular functions involved in innate immunity. However, the nature of their interactions remains unclear. We evaluated the role of autophagy in regulating phagocytosis in macrophages from myeloid-specific autophagy-related gene 7-deficient (Atg7⁻/⁻) mice. Atg7⁻/⁻ macrophages exhibited higher bacterial uptake when infected with Mycobacterium tuberculosis (Mtb) or with M. tuberculosis var. bovis BCG (BCG). In addition, BCG-infected Atg7⁻/⁻ mice showed increased bacterial loads and exacerbated lung inflammatory responses. Atg7⁻/⁻ macrophages had increased expression of two class A scavenger receptors: macrophage receptor with collagenous structure (MARCO) and macrophage scavenger receptor 1 (MSR1). The increase in scavenger receptors was caused by increased activity of the nuclear factor (erythroid-derived 2)-like 2 (NFE2L2) transcription factor resulting from accumulated sequestosome 1 (SQSTM1 or p62) in Atg7⁻/⁻ macrophages. These insights increase our understanding of the host-pathogen relationship and suggest that therapeutic strategies should be designed to include modulation of both phagocytosis and autophagy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy/immunology , Heat-Shock Proteins/metabolism , Microtubule-Associated Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Phagocytosis/immunology , Receptors, Immunologic/biosynthesis , Scavenger Receptors, Class A/biosynthesis , Animals , Autophagy-Related Protein 7 , Bacterial Load/immunology , Cells, Cultured , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Mycobacterium Infections/immunology , Mycobacterium bovis/growth & development , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/immunology , Pneumonia/immunology , Pneumonia/microbiology , Receptors, Scavenger/biosynthesis , Receptors, Scavenger/immunology , Sequestosome-1 Protein , Up-Regulation
4.
EMBO J ; 36(17): 2544-2552, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28754656

ABSTRACT

TFEB is a master regulator for transcription of genes involved in autophagy and lysosome biogenesis. Activity of TFEB is inhibited upon its serine phosphorylation by mTOR The overall mechanisms by which TFEB activity in the cell is regulated are not well elucidated. Specifically, the mechanisms of TFEB turnover and how they might influence its activity remain unknown. Here, we show that STUB1, a chaperone-dependent E3 ubiquitin ligase, modulates TFEB activity by preferentially targeting inactive phosphorylated TFEB for degradation by the ubiquitin-proteasome pathway. Phosphorylated TFEB accumulated in STUB1-deficient cells and in tissues of STUB1-deficient mice resulting in reduced TFEB activity. Conversely, cellular overexpression of STUB1 resulted in reduced phosphorylated TFEB and increased TFEB activity. STUB1 preferentially interacted with and ubiqutinated phosphorylated TFEB, targeting it to proteasomal degradation. Consistent with reduced TFEB activity, accumulation of phosphorylated TFEB in STUB1-deficient cells resulted in reduced autophagy and reduced mitochondrial biogenesis. These studies reveal that the ubiquitin-proteasome pathway participates in regulating autophagy and lysosomal functions by regulating the activity of TFEB.


Subject(s)
Autophagy/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Lysosomes/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice, Knockout , Phosphorylation , Signal Transduction , Ubiquitin-Protein Ligases/genetics
5.
Int J Mol Sci ; 21(12)2020 Jun 17.
Article in English | MEDLINE | ID: mdl-32560429

ABSTRACT

Alpha one antitrypsin (α1AT), a serine proteinase inhibitor primarily produced by the liver, protects pulmonary tissue from neutrophil elastase digestion. Mutations of the SERPINA1 gene results in a misfolded α1AT protein which aggregates inside hepatocytes causing cellular damage. Therefore, inhibition of mutant α1AT production is one practical strategy to alleviate liver damage. Here we show that proteasome inhibitors can selectively downregulate α1AT expression in human hepatocytes by suppressing the translation of α1AT. Translational suppression of α1AT is mediated by phosphorylation of eukaryotic translation initiation factor 2α and increased association of RNA binding proteins, especially stress granule protein Ras GAP SH3 binding protein (G3BP1), with α1AT mRNA. Treatment of human-induced pluripotent stem cell-derived hepatocytes with a proteasome inhibitor also results in translational inhibition of mutant α1AT in a similar manner. Together we revealed a previously undocumented role of proteasome inhibitors in the regulation of α1AT translation.


Subject(s)
Gene Expression Regulation/drug effects , Proteasome Inhibitors/pharmacology , RNA Processing, Post-Transcriptional/drug effects , alpha 1-Antitrypsin/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Protein Biosynthesis/drug effects , Stress, Physiological , alpha 1-Antitrypsin/biosynthesis
6.
J Pharmacol Exp Ther ; 368(2): 169-178, 2019 02.
Article in English | MEDLINE | ID: mdl-30446578

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a fatal disease that destroys the structure and function of the lungs. Risk factors include advanced age and genetic predisposition. However, tobacco use is the chief modifiable risk factor. The prevalence of tobacco use in IPF reaches up to 80%. Although tobacco smoke contains over 5000 chemicals, nicotine is a major component. Nicotine is a bioactive molecule that acts upon nicotinic acetylcholine receptors expressed on neuronal and non-neuronal cells including endothelial cells. Accordingly, it has a pleiotropic effect on cell proliferation and angiogenesis. The angiogenic effect is partly mediated by stimulation of growth factors including fibroblast, platelet-derived, and vascular endothelial growth factors. Nintedanib, a Food and Drug Administration-approved drug for IPF, works by inhibiting receptors for these growth factors, suggesting a pathobiologic role of the growth factors in IPF and a potential mechanism by which tobacco use may exacerbate the disease process; additionally, nicotine downregulates anti-inflammatory microRNAs (miRs) in lung cells. Here, we profiled the expression of miRs in lung tissues explanted from a lung injury model and examined the effect of nicotine on one of the identified miRs (miR-24) and its downstream targets. Our data show that miR-24 is downregulated during lung injury and is suppressed by nicotine. We also found that nicotine upregulates the expression of inflammatory cytokines targeted by miR-24. Finally, nicotine stimulated growth factors, fibroblast proliferation, collagen release, and expression of myofibroblast markers. Taken together, nicotine, alone or as a component of tobacco smoke, may accelerate the disease process in IPF through stimulation of growth factors and downregulation of anti-inflammatory miRs.


Subject(s)
Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Nicotine/toxicity , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Intercellular Signaling Peptides and Proteins/agonists , Male , MicroRNAs/antagonists & inhibitors , Nicotinic Agonists/toxicity , Rats , Rats, Inbred F344 , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism
7.
Arterioscler Thromb Vasc Biol ; 37(8): 1524-1535, 2017 08.
Article in English | MEDLINE | ID: mdl-28596374

ABSTRACT

OBJECTIVE: Bacterial endotoxin (lipopolysaccharide)-mediated sepsis involves dysregulated systemic inflammation, which injures the lung and other organs, often fatally. Vascular endothelial cells act as both targets and mediators of lipopolysaccharide-induced inflammatory responses. Dysfunction of endothelium results in increases of proinflammatory cytokine production and permeability leakage. BMPER (bone morphogenetic protein-binding endothelial regulator), an extracellular modulator of bone morphogenetic protein signaling, has been identified as a vital component in chronic endothelial inflammatory responses and atherosclerosis. However, it is unclear whether BMPER also regulates inflammatory response in an acute setting such as sepsis. To address this question, we investigated the role of BMPER during lipopolysaccharide-induced acute lung injury. APPROACH AND RESULTS: Mice missing 1 allele of BMPER (BMPER+/- mice used in the place of BMPER-/- mice that die at birth) were used for lipopolysaccharide challenge. Lipopolysaccharide-induced pulmonary inflammation and injury was reduced in BMPER+/- mice as shown by several measures, including survival rate, infiltration of inflammatory cells, edema, and production of proinflammatory cytokines. Mechanistically, we have demonstrated that BMPER is required and sufficient for the activation of nuclear factor of activated T cells c1. This BMPER-induced nuclear factor of activated T cells activation is coordinated by multiple signaling pathways, including bone morphogenetic protein-independent low-density lipoprotein receptor-related protein 1-extracellular signal-regulated kinase activation, calcineurin signaling, and low-density lipoprotein receptor-related protein 1ß-mediated nuclear factor 45 nuclear export in response to BMPER treatment. CONCLUSIONS: We conclude that BMPER plays a pivotal role in pulmonary inflammatory response, which provides new therapeutic options against sepsis shock. The new signaling pathway initiated by BMPER/low-density lipoprotein receptor-related protein 1 axis broadens our understanding about BMPER's role in vascular homeostasis.


Subject(s)
Acute Lung Injury/metabolism , Carrier Proteins/metabolism , Endothelial Cells/metabolism , Endotoxins , Lung/blood supply , Pneumonia/metabolism , Receptors, LDL/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Apoptosis , Capillary Permeability , Carrier Proteins/genetics , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/pathology , Genetic Predisposition to Disease , Haploinsufficiency , Inflammation Mediators/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Lung/metabolism , Lung/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/metabolism , Nuclear Factor 45 Protein/metabolism , Phenotype , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/pathology , RNA Interference , Receptors, LDL/genetics , Time Factors , Transfection , Tumor Suppressor Proteins/genetics
8.
J Immunol ; 194(11): 5407-16, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25888640

ABSTRACT

Autophagy is an important component of the immune response. However, the functions of autophagy in human diseases are much less understood. We studied biological consequences of autophagy deficiency in mice lacking the essential autophagy gene Atg7 or Atg5 in myeloid cells. Surprisingly, these mice presented with spontaneous sterile lung inflammation, characterized by marked recruitment of inflammatory cells, submucosal thickening, goblet cell metaplasia, and increased collagen content. Lung inflammation was associated with increase in several proinflammatory cytokines in the bronchoalveolar lavage and in serum. This inflammation was largely driven by IL-18 as a result of constitutive inflammasome activation. Following i.p. LPS injection, autophagy-deficient mice had higher levels of proinflammatory cytokines in lungs and in serum, as well as increased mortality, than control mice. Intranasal bleomycin challenge exacerbated lung inflammation in autophagy-deficient mice and produced more severe fibrotic changes than in control mice. These results uncover a new and important role for autophagy as negative regulator of lung inflammation.


Subject(s)
Autophagy/immunology , Interleukin-18/immunology , Microtubule-Associated Proteins/genetics , Pneumonia/immunology , Animals , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Bleomycin/pharmacology , Bronchoalveolar Lavage Fluid/immunology , Collagen/metabolism , Cytokines/blood , Cytokines/metabolism , Fibrosis/genetics , Fibrosis/immunology , Goblet Cells/immunology , Inflammasomes/immunology , Interleukin-18/genetics , Lipopolysaccharides/administration & dosage , Lung/immunology , Lung/pathology , Metaplasia , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/mortality , Pneumonia/pathology
9.
J Biol Chem ; 289(38): 26525-26532, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25077962

ABSTRACT

Dendritic cells (DCs) are the most potent antigen-presenting cells (APCs) in the immune system. DCs present antigens to CD8 and CD4 T cells in the context of class I or II MHC. Recent evidence suggests that autophagy, a conserved intracellular degradation pathway, regulates class II antigen presentation. In vitro studies have shown that deletion of autophagy-related genes reduced antigen presentation by APCs to CD4 T cells. In vivo studies confirmed these findings in the context of infectious diseases. However, the relevance of autophagy-mediated antigen presentation in autoimmunity remains to be elucidated. Here, we report that loss of autophagy-related gene 7 (Atg7) in DCs ameliorated experimental autoimmune encephalomyelitis (EAE), a CD4 T cell-mediated mouse model of multiple sclerosis, by reducing in vivo priming of T cells. In contrast, severity of hapten-induced contact hypersensitivity, in which CD8 T cells and NK cells play major roles, was unaffected. Administration of the autophagy-lysosomal inhibitor chloroquine, before EAE onset, delayed disease progression and, when administered after the onset, reduced disease severity. Our data show that autophagy is required in DCs for induction of EAE and suggest that autophagy might be a potential target for treating CD4 T cell-mediated autoimmune conditions.


Subject(s)
Autophagy , Dendritic Cells/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Microtubule-Associated Proteins/genetics , Animals , Antigen Presentation , Autophagy/drug effects , Autophagy-Related Protein 7 , CD4-Positive T-Lymphocytes/immunology , Chloroquine/pharmacology , Chloroquine/therapeutic use , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Spleen/immunology , Spleen/pathology , Thymus Gland/immunology , Thymus Gland/pathology
10.
Cell Microbiol ; 16(11): 1637-45, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25237740

ABSTRACT

Eukaryotic cells utilize two main secretory pathways to transport proteins to the extracellular space. Proteins with a leader signal sequence often undergo co-translational transport into the endoplasmic reticulum (ER), and then to the Golgi apparatus before they reach their destination. This pathway is called the conventional secretory pathway. Proteins without signal peptides can bypass this ER-Golgi system and are secreted by a variety of mechanisms collectively called the unconventional secretory pathway. The molecular mechanisms of unconventional secretion are emerging. Autophagy is a conserved bulk degradation mechanism that regulates many intracellular functions. Recent evidence implicates autophagy in the secretory pathway. This review focuses on potential secretory roles of autophagy and how they could modulate the functions of innate immune cells that secrete a wide range of mediators in response to environmental and biological stimuli. We provide a brief overview of the secretory pathways, enumerate the potential mechanistic themes by which autophagy interacts with these pathways and describe their relevance in the context of innate immune cell function.


Subject(s)
Autophagy , Cells/immunology , Cells/metabolism , Proteins/metabolism , Animals , Humans , Mammals
11.
Am J Respir Crit Care Med ; 189(1): 16-29, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24251647

ABSTRACT

RATIONALE: IL-4Rα, the common receptor component for IL-4 and IL-13, plays a critical role in IL-4- and IL-13-mediated signaling pathways that regulate airway inflammation and remodeling. However, the regulatory mechanisms underlying IL-4Rα turnover and its signal termination remain elusive. OBJECTIVES: To evaluate the role of STUB1 (STIP1 homology and U-Box containing protein 1) in regulating IL-4R signaling in airway inflammation. METHODS: The roles of STUB1 in IL-4Rα degradation and its signaling were investigated by immunoblot, immunoprecipitation, and flow cytometry. The involvement of STUB1 in airway inflammation was determined in vivo by measuring lung inflammatory cells infiltration, mucus production, serum lgE levels, and alveolar macrophage M2 activation in STUB1(-/-) mice. STUB1 expression was evaluated in airway epithelium of patients with asthma and lung tissues of subjects with chronic obstructive pulmonary disease. MEASUREMENTS AND MAIN RESULTS: STUB1 interacted with IL-4Rα and targeted it for ubiquitination-mediated proteasomal degradation, terminating IL-4 or IL-13 signaling. STUB1 knockout cells showed increased levels of IL-4Rα and sustained STAT6 activation, whereas STUB1 overexpression reduced IL-4Rα levels. Mice deficient in STUB1 had spontaneous airway inflammation, alternative M2 activation of alveolar macrophage, and increased serum IgE. STUB1 levels were increased in airways of subjects with asthma or chronic obstructive pulmonary disease, suggesting that up-regulation of STUB1 might be an important feedback mechanism to dampen IL-4R signaling in airway inflammation. CONCLUSIONS: Our study identified a previously uncharacterized role for STUB1 in regulating IL-4R signaling, which might provide a new strategy for attenuating airway inflammation.


Subject(s)
Pneumonia/physiopathology , Receptors, Interleukin-4/physiology , Signal Transduction/physiology , Ubiquitin-Protein Ligases/physiology , Adult , Animals , Asthma/physiopathology , Child , Down-Regulation/physiology , Female , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , Macrophage Activation/physiology , Male , Mice , Mice, Knockout , Proteasome Endopeptidase Complex/physiology , Pulmonary Disease, Chronic Obstructive/physiopathology , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/physiology , STAT6 Transcription Factor/physiology , Ubiquitin/physiology
12.
J Biol Chem ; 288(50): 35886-95, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24178293

ABSTRACT

The association between altered proteostasis and inflammatory disorders has been increasingly recognized, but the underlying mechanisms are not well understood. In this study, we show that deficiency of either autophagy or sequestosome 1 (p62 or SQSTM) led to inflammasome hyperactivation in response to LPS and ATP in primary macrophages and in mice in vivo. Importantly, induction of protein misfolding by puromycin, thapsigargin, or geldanamycin resulted in inflammasome activation that was more pronounced in autophagy- or p62-deficient macrophages. Accumulation of misfolded proteins caused inflammasome activation by inducing generation of nonmitochondrial reactive oxygen species and lysosomal damage, leading to release of cathepsin B. Our results suggest that altered proteostasis results in inflammasome activation and thus provide mechanisms for the association of altered proteostasis with inflammatory disorders.


Subject(s)
Homeostasis , Inflammasomes/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Animals , Autophagy , Carrier Proteins/metabolism , Cathepsin B/metabolism , Cell Line , Heat-Shock Proteins/deficiency , Humans , Lysosomes/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Folding , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein
13.
J Biol Chem ; 287(23): 19687-98, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22518844

ABSTRACT

Failure to maintain protein homeostasis (proteostasis) leads to accumulation of unfolded proteins and contributes to the pathogenesis of many human diseases. Accumulation of unfolded proteins in the endoplasmic reticulum (ER) elicits unfolded protein response (UPR) that serves to attenuate protein translation, and increase protein refolding or degradation. In contrast to UPR in the ER, the regulatory molecules operative in cytosolic responses and their potential relation to ER stress are not well elucidated. Aggresome-like induced structures (ALIS) have been described as transient aggregation of ubiquitinated proteins in the cytosol. In this study, we show that cells respond to inflammation, infection or ER stress by cytosolic formation of ALIS, indicating that ALIS formation represents an early event in cellular adjustment to altered proteostasis that occurs under these conditions. This response was aided by rapid transcriptional up-regulation of polyubiqutin-binding protein p62. NF-κB and mTOR activation were also required for ALIS formation. Importantly, we show a cross talk between UPR in the ER and cytosolic ALIS. Down-regulation of ER UPR in XBP1 deficient cells increases cyotosolic ALIS formation. Furthermore, lysosomal activity but not macroautophagy is responsible for ALIS clearance. This study reveals the underlying regulatory mechanisms of ALIS formation and clearance, and provides a previously unrecognized common adaptive mechanism for cellular responses against inflammation and ER stress.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Ubiquitinated Proteins/metabolism , Unfolded Protein Response/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cytosol/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/physiology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Regulatory Factor X Transcription Factors , Sequestosome-1 Protein , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/physiology , Ubiquitinated Proteins/genetics , Up-Regulation/physiology , X-Box Binding Protein 1
14.
Blood ; 118(6): 1525-33, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21633090

ABSTRACT

The IFN-inducible immunity-related p47 GTPase Irgm1 has been linked to Crohn disease as well as susceptibility to tuberculosis. Previously we demonstrated that HSC quiescence and function are aberrant in mice lacking Irgm1. To investigate the molecular basis for these defects, we conducted microarray expression profiling of Irgm1-deficient HSCs. Cell-cycle and IFN-response genes are up-regulated in Irgm1(-/-) HSCs, consistent with dysregulated IFN signaling. To test the hypothesis that Irgm1 normally down-regulates IFN signaling in HSCs, we generated Irgm1(-/-)Ifngr1(-/-) and Irgm1(-/-)Stat1(-/-) double-knockout animals. Strikingly, hyperproliferation, self-renewal, and autophagy defects in Irgm1(-/-) HSCs were normalized in double-knockout animals. These defects were also abolished in Irgm1(-/-)Irgm3(-/-) double-knockout animals, indicating that Irgm1 may regulate Irgm3 activity. Furthermore, the number of HSCs was reduced in aged Irgm1(-/-) animals, suggesting that negative feedback inhibition of IFN signaling by Irgm1 is necessary to prevent hyperproliferation and depletion of the stem cell compartment. Collectively, our results indicate that Irgm1 is a powerful negative regulator of IFN-dependent stimulation in HSCs, with an essential role in preserving HSC number and function. The deleterious effects of excessive IFN signaling may explain how hematologic abnormalities arise in patients with inflammatory conditions.


Subject(s)
GTP-Binding Proteins/genetics , Hematopoietic Stem Cells/metabolism , Receptors, Interferon/genetics , Signal Transduction/genetics , Animals , Autophagy/drug effects , Autophagy/genetics , Bone Marrow Transplantation , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , Flow Cytometry , GTP-Binding Proteins/deficiency , Gene Expression/drug effects , Gene Expression Profiling , HEK293 Cells , Humans , Immunohistochemistry , Interferons/genetics , Interferons/metabolism , Interferons/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptors, Interferon/deficiency , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , Interferon gamma Receptor
15.
Curr Opin Pulm Med ; 19(1): 30-5, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23143196

ABSTRACT

PURPOSE OF REVIEW: Asthma is a common worldwide respiratory illness with significant morbidity and mortality. The disease is characterized by airway inflammation with involvement of multiple biological pathways. Genetic predisposition and increased susceptibility to severe respiratory viral infections are well known clinical features of asthma. Autophagy is an evolutionarily conserved cellular degradation process with significant impact on immunity and antiviral response. In this review we have described the role of autophagy in immune cell survival, proliferation and function. Autophagy has complex effects on immune response involved in inflammation, specifically Th2 immune response. Common respiratory viruses are associated with increased morbidity and mortality in asthmatic patients. RECENT FINDINGS: We describe recent studies showing the effect of autophagy on replication and immune response to common respiratory viruses. The role of autophagy in asthma has recently been investigated. Two studies have been published describing the association of autophagy with asthma. Genetic polymorphism in specific autophagy genes is associated with asthma and influences gene expression in an experimental in-vivo model. SUMMARY: These studies provide us with a window into the possible role of autophagy in asthma and offer new clues to pathogenesis. Modulation of autophagy has the potential to develop into a new therapeutic avenue to treat this common respiratory ailment.


Subject(s)
Asthma/physiopathology , Autophagy/physiology , Immunity/physiology , Asthma/genetics , Asthma/immunology , Autophagy/immunology , Autophagy-Related Protein 5 , Disease Susceptibility/immunology , Disease Susceptibility/physiopathology , Humans , Microtubule-Associated Proteins/genetics , Polymorphism, Genetic/genetics , Respiratory Tract Infections/immunology , Respiratory Tract Infections/physiopathology , Respiratory Tract Infections/virology
16.
Proc Natl Acad Sci U S A ; 107(31): 13854-9, 2010 Aug 03.
Article in English | MEDLINE | ID: mdl-20634424

ABSTRACT

Human Rpn13, also known as adhesion regulating molecule 1 (ADRM1), was recently identified as a novel 19S proteasome cap-associated protein, which recruits the deubiquitinating enzyme UCH37 to the 26S proteasome. Knockdown of Rpn13 by siRNA does not lead to global accumulation of ubiquitinated cellular proteins or changes in proteasome expression, suggesting that Rpn13 must have a specialized role in proteasome function. Thus, Rpn13 participation in protein degradation, by recruiting UCH37, is rather selective to specific proteins whose degradation critically depends on UCH37 deubiquitination activity. The specific substrates for the Rpn13/UCH37 complex have not been determined. Because of a previous discovery of an interaction between Rpn13 and inducible nitric oxide synthase (iNOS), we hypothesized that iNOS is one of the substrates for the Rpn13/UCH37 complex. In this study, we show that Rpn13 is involved in iNOS degradation and is required for iNOS interaction with the deubiquitination protein UCH37. Furthermore, we discovered that IkappaB-alpha, a protein whose proteasomal degradation activates the transcription factor NF-kappaB, is also a substrate for the Rpn13/UCH37 complex. Thus, this study defines two substrates, with important roles in inflammation and host defense for the Rpn13/UCH37 pathway.


Subject(s)
Cell Adhesion Molecules/metabolism , Membrane Glycoproteins/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Animals , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Cell Adhesion Molecules/genetics , Cell Line , Down-Regulation , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Mice , Protein Binding , RNA, Small Interfering/genetics , Ubiquitin Thiolesterase
17.
FASEB J ; 25(1): 99-110, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20826541

ABSTRACT

Cachectic muscle wasting is a frequent complication of many inflammatory conditions, due primarily to excessive muscle catabolism. However, the pathogenesis and intervention strategies against it remain to be established. Here, we tested the hypothesis that Toll-like receptor 4 (TLR4) is a master regulator of inflammatory muscle catabolism. We demonstrate that TLR4 activation by lipopolysaccharide (LPS) induces C2C12 myotube atrophy via up-regulating autophagosome formation and the expression of ubiquitin ligase atrogin-1/MAFbx and MuRF1. TLR4-mediated activation of p38 MAPK is necessary and sufficient for the up-regulation of atrogin1/MAFbx and autophagosomes, resulting in myotube atrophy. Similarly, LPS up-regulates muscle autophagosome formation and ubiquitin ligase expression in mice. Importantly, autophagy inhibitor 3-methyladenine completely abolishes LPS-induced muscle proteolysis, while proteasome inhibitor lactacystin partially blocks it. Furthermore, TLR4 knockout or p38 MAPK inhibition abolishes LPS-induced muscle proteolysis. Thus, TLR4 mediates LPS-induced muscle catabolism via coordinate activation of the ubiquitin-proteasome and the autophagy-lysosomal pathways.


Subject(s)
Lipopolysaccharides/pharmacology , Muscles/drug effects , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Autophagy/drug effects , Blotting, Western , Cell Line , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscles/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Phagosomes/drug effects , Phagosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Pyridines/pharmacology , RNA Interference , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Toll-Like Receptor 4/genetics , Ubiquitin/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Proc Natl Acad Sci U S A ; 106(4): 1211-5, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19139419

ABSTRACT

Nitric Oxide (NO), produced by inducible nitric oxide synthase (iNOS), has been implicated in the pathogenesis of various biological and inflammatory disorders. Recent evidence suggests that aggresome formation is a physiologic stress response not limited to misfolded proteins. That stress response, termed "physiologic aggresome," is exemplified by aggresome formation of iNOS, an important host defense protein. The functional significance of cellular formation of the iNOS aggresome is hitherto unknown. In this study, we used live cell imaging, fluorescence microscopy, and intracellular fluorescence NO probes to map the subcellular location of iNOS and NO under various conditions. We found that NO production colocalized with cytosolic iNOS but aggresomes containing iNOS were distinctly devoid of NO production. Further, cells expressing iNOS aggresomes produced significantly less NO as compared with cells not expressing aggresomes. Importantly, primary normal human bronchial epithelial cells, stimulated by cytokines to express iNOS, progressively sequestered iNOS to the aggresome, a process that correlated with marked reduction of NO production. These results suggest that bronchial epithelial cells used the physiologic aggresome mechanism for iNOS inactivation. Our studies reveal a novel cellular strategy to terminate NO production via formation of the iNOS aggresome.


Subject(s)
Inclusion Bodies/enzymology , Nitric Oxide Synthase Type II/metabolism , Stress, Physiological , Bronchi/cytology , Bronchi/enzymology , Cell Survival/drug effects , Cells, Cultured , Cytokines/pharmacology , Enzyme Activation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Humans , Inclusion Bodies/drug effects , Nitric Oxide/biosynthesis , Protein Transport/drug effects , Rhodamines/metabolism , Stress, Physiological/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology
19.
Sci Rep ; 12(1): 20668, 2022 11 30.
Article in English | MEDLINE | ID: mdl-36450789

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease of unknown etiology. Currently, pirfenidone and nintedanib are the only FDA-approved drugs for the treatment of IPF and are now the standard of care. This is a significant step in slowing down the progression of the disease. However, the drugs are unable to stop or reverse established fibrosis. Several retrospective clinical studies indicate that proton pump inhibitors (PPIs; FDA-approved to treat gastroesophageal reflux) are associated with favorable outcomes in patients with IPF, and emerging preclinical studies report that PPIs possess antifibrotic activity. In this study, we evaluated the antifibrotic efficacy of the PPI esomeprazole when combined with pirfenidone in vitro and in vivo. In cell culture studies of IPF lung fibroblasts, we assessed the effect of the combination on several fibrosis-related biological processes including TGFß-induced cell proliferation, cell migration, cell contraction, and collagen production. In an in vivo study, we used mouse model of TGFß-induced lung fibrosis to evaluate the antifibrotic efficacy of esomeprazole/pirfenidone combination. We also performed computational studies to understand the molecular mechanisms by which esomeprazole and/or pirfenidone regulate lung fibrosis. We found that esomeprazole significantly enhanced the anti-proliferative effect of pirfenidone and favorably modulated TGFß-induced cell migration and contraction of collagen gels. We also found that the combination significantly suppressed collagen production in response to TGFß in comparison to pirfenidone monotherapy. In addition, our animal study demonstrated that the combination therapy effectively inhibited the differentiation of lung fibroblasts into alpha smooth muscle actin (αSMA)-expressing myofibroblasts to attenuate the progression of lung fibrosis. Finally, our bioinformatics study of cells treated with esomeprazole or pirfenidone revealed that the drugs target several extracellular matrix (ECM) related pathways with esomeprazole preferentially targeting collagen family members while pirfenidone targets the keratins. In conclusion, our cell biological, computational, and in vivo studies show that the PPI esomeprazole enhances the antifibrotic efficacy of pirfenidone through complementary molecular mechanisms. This data supports the initiation of prospective clinical studies aimed at repurposing PPIs for the treatment of IPF and other fibrotic lung diseases where pirfenidone is prescribed.


Subject(s)
Esomeprazole , Idiopathic Pulmonary Fibrosis , Animals , Mice , Esomeprazole/pharmacology , Transforming Growth Factor beta , Prospective Studies , Retrospective Studies , Proton Pump Inhibitors/pharmacology , Idiopathic Pulmonary Fibrosis/drug therapy , Disease Models, Animal
20.
J Biol Chem ; 285(1): 784-92, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19875457

ABSTRACT

Src kinases are key regulators of cellular proliferation, survival, motility, and invasiveness. They play important roles in the regulation of inflammation and cancer. Overexpression or hyperactivity of c-Src has been implicated in the development of various types of cancer, including lung cancer. Src inhibition is currently being investigated as a potential therapy for non-small cell lung cancer in Phase I and II clinical trials. The mechanisms of Src implication in cancer and inflammation are linked to the ability of activated Src to phosphorylate multiple downstream targets that mediate its cellular effector functions. In this study, we reveal that inducible nitric-oxide synthase (iNOS), an enzyme also implicated in cancer and inflammation, is a downstream mediator of activated Src. We elucidate the molecular mechanisms of the association between Src and iNOS in models of inflammation induced by lipopolysaccharide and/or cytokines and in cancer cells and tissues. We identify human iNOS residue Tyr(1055) as a target for Src-mediated phosphorylation. These results are shown in normal cells and cancer cells as well as in vivo in mice. Importantly, such posttranslational modification serves to stabilize iNOS half-life. The data also demonstrate interactions and co-localization of iNOS and activated Src under inflammatory conditions and in cancer cells. This study demonstrates that phosphorylation of iNOS by Src plays an important role in the regulation of iNOS and nitric oxide production and hence could account for some Src-related roles in inflammation and cancer.


Subject(s)
Neoplasms/enzymology , Nitric Oxide Synthase Type II/metabolism , src-Family Kinases/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Stability/drug effects , Epidermal Growth Factor/pharmacology , Epithelium/drug effects , Epithelium/enzymology , Epithelium/pathology , Half-Life , Humans , Lung/drug effects , Lung/enzymology , Lung/pathology , Mice , Mice, Inbred C57BL , Models, Biological , Neoplasms/pathology , Phosphorylation/drug effects , Phosphoserine/metabolism , Pneumonia/enzymology , Pneumonia/pathology , Protein Transport/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL