Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1181876, 2023.
Article in English | MEDLINE | ID: mdl-37275891

ABSTRACT

Introduction: Resurgence of pertussis, caused by Bordetella pertussis, necessitates novel vaccines and vaccination strategies to combat this disease. Alum-adjuvanted acellular pertussis vaccines (aPV) delivered intramuscularly reduce bacterial numbers in the lungs of immunized animals and humans, but do not reduce nasal colonization. Thus, aPV-immunized individuals are sources of community transmission. We showed previously that modification of a commercial aPV (Boostrix) by addition of the Th1/17 polarizing adjuvant Bordetella Colonization Factor A (BcfA) attenuated Th2 responses elicited by alum and accelerated clearance of B. pertussis from mouse lungs. Here we tested whether a heterologous immunization strategy with systemic priming and mucosal booster (prime-pull) would reduce nasal colonization. Methods: Adult male and female mice were immunized intramuscularly (i.m.) with aPV or aPV/BcfA and boosted either i.m. or intranasally (i.n.) with the same formulation. Tissue-resident memory (TRM) responses in the respiratory tract were quantified by flow cytometry, and mucosal and systemic antibodies were quantified by ELISA. Immunized and naïve mice were challenged i.n. with Bordetella pertussis and bacterial load in the nose and lungs enumerated at days 1-14 post-challenge. Results: We show that prime-pull immunization with Boostrix plus BcfA (aPV/BcfA) generated IFNγ+ and IL-17+ CD4+ lung resident memory T cells (TRM), and CD4+IL-17+ TRM in the nose. In contrast, aPV alone delivered by the same route generated IL-5+ CD4+ resident memory T cells in the lungs and nose. Importantly, nasal colonization was only reduced in mice immunized with aPV/BcfA by the prime-pull regimen. Conclusions: These results suggest that TH17 polarized TRM generated by aPV/BcfA may reduce nasal colonization thereby preventing pertussis transmission and subsequent resurgence.


Subject(s)
Bordetella pertussis , Whooping Cough , Animals , Female , Male , Mice , Adjuvants, Immunologic , Adjuvants, Pharmaceutic , CD4-Positive T-Lymphocytes , Interleukin-17 , Pertussis Vaccine , Whooping Cough/prevention & control
3.
Sci Rep ; 11(1): 855, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441602

ABSTRACT

Burkholderia cenocepacia (B. cenocepacia) is an opportunistic bacterium; causing severe life threatening systemic infections in immunocompromised individuals including cystic fibrosis patients. The lack of gasdermin D (GSDMD) protects mice against endotoxin lipopolysaccharide (LPS) shock. On the other hand, GSDMD promotes mice survival in response to certain bacterial infections. However, the role of GSDMD during B. cenocepacia infection is not yet determined. Our in vitro study shows that GSDMD restricts B. cenocepacia replication within macrophages independent of its role in cell death through promoting mitochondrial reactive oxygen species (mROS) production. mROS is known to stimulate autophagy, hence, the inhibition of mROS or the absence of GSDMD during B. cenocepacia infections reduces autophagy which plays a critical role in the restriction of the pathogen. GSDMD promotes inflammation in response to B. cenocepacia through mediating the release of inflammasome dependent cytokine (IL-1ß) and an independent one (CXCL1) (KC). Additionally, different B. cenocepacia secretory systems (T3SS, T4SS, and T6SS) contribute to inflammasome activation together with bacterial survival within macrophages. In vivo study confirmed the in vitro findings and showed that GSDMD restricts B. cenocepacia infection and dissemination and stimulates autophagy in response to B. cenocepacia. Nevertheless, GSDMD promotes lung inflammation and necrosis in response to B. cenocepacia without altering mice survival. This study describes the double-edged functions of GSDMD in response to B. cenocepacia infection and shows the importance of GSDMD-mediated mROS in restriction of B. cenocepacia.


Subject(s)
Burkholderia Infections/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Animals , Autophagy/physiology , Burkholderia Infections/prevention & control , Burkholderia cenocepacia/pathogenicity , Caspases, Initiator/genetics , Caspases, Initiator/metabolism , Cell Death , Female , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Lipopolysaccharides/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/physiology , Reactive Oxygen Species/metabolism
4.
PLoS One ; 15(10): e0237520, 2020.
Article in English | MEDLINE | ID: mdl-33002030

ABSTRACT

OBJECTIVES: Gout is the most prevalent inflammatory arthritis. To study the effects of regular physical activity and exercise intensity on inflammation and clinical outcome, we examined inflammatory pathogenesis in an acute model of murine gout and analyzed human gout patient clinical data as a function of physical activity. METHODS: NF-κB-luciferase reporter mice were organized into four groups and exercised at 0 m/min (non-exercise), 8 m/min (low-intensity), 11 m/min (moderate-intensity), and 15 m/min (high-intensity) for two weeks. Mice subsequently received intra-articular monosodium urate (MSU) crystal injections (0.5mg) and the inflammatory response was analyzed 15 hours later. Ankle swelling, NF-κB activity, histopathology, and tissue infiltration by macrophages and neutrophils were measured. Toll-like receptor (TLR)2 was quantified on peripheral monocytes/neutrophils by flow cytometry and both cytokines and chemokines were measured in serum or synovial aspirates. Clinical data and questionnaires accessing overall physical activity levels were collected from gout patients. RESULTS: Injection of MSU crystals produced a robust inflammatory response with increased ankle swelling, NF-κB activity, and synovial infiltration by macrophages and neutrophils. These effects were partially mitigated by low and moderate-intensity exercise. Furthermore, IL-1ß was decreased at the site of MSU crystal injection, TLR2 expression on peripheral neutrophils was downregulated, and expression of CXCL1 in serum was suppressed with low and moderate-intensity exercise. Conversely, the high-intensity exercise group closely resembled the non-exercised control group by nearly all metrics of inflammation measured in this study. Physically active gout patients had significantly less flares/yr, decreased C-reactive protein (CRP) levels, and lower pain scores relative to physically inactive patients. CONCLUSIONS: Regular, moderate physical activity can produce a quantifiable anti-inflammatory effect capable of partially mitigating the pathologic response induced by intra-articular MSU crystals by downregulating TLR2 expression on circulating neutrophils and suppressing systemic CXCL1. Low and moderate-intensity exercise produces this anti-inflammatory effect to varying degrees, while high-intensity exercise provides no significant difference in inflammation compared to non-exercising controls. Consistent with the animal model, gout patients with higher levels of physical activity have more favorable prognostic data. Collectively, these data suggest the need for further research and may be the foundation to a future paradigm-shift in conventional exercise recommendations provided by Rheumatologists to gout patients.


Subject(s)
Chemokine CXCL1/blood , Gout/therapy , Inflammation/prevention & control , Physical Conditioning, Animal , Toll-Like Receptor 2/blood , Animals , Disease Models, Animal , Down-Regulation , Exercise/physiology , Female , Gout/blood , Gout/pathology , Humans , Inflammation/blood , Inflammation/pathology , Interleukin-1beta/blood , Interleukin-1beta/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neutrophils/metabolism , Neutrophils/pathology , Pain/prevention & control , Prognosis , Synovial Membrane/metabolism , Synovial Membrane/pathology
5.
Angew Chem Int Ed Engl ; 59(16): 6451-6458, 2020 04 16.
Article in English | MEDLINE | ID: mdl-31953912

ABSTRACT

With the infection rate of Bordetella pertussis at a 60-year high, there is an urgent need for new anti-pertussis vaccines. The lipopolysaccharide (LPS) of B. pertussis is an attractive antigen for vaccine development. With the presence of multiple rare sugars and unusual glycosyl linkages, the B. pertussis LPS is a highly challenging synthetic target. In this work, aided by molecular dynamics simulation and modeling, a pertussis-LPS-like pentasaccharide was chemically synthesized for the first time. The pentasaccharide was conjugated with a powerful carrier, bacteriophage Qß, as a vaccine candidate. Immunization of mice with the conjugate induced robust anti-glycan IgG responses with IgG titers reaching several million enzyme-linked immunosorbent assay (ELISA) units. The antibodies generated were long lasting and boostable and could recognize multiple clinical strains of B. pertussis, highlighting the potential of Qß-glycan as a new anti-pertussis vaccine.


Subject(s)
Oligosaccharides/immunology , Pertussis Vaccine/chemical synthesis , Animals , Cattle , Enzyme-Linked Immunosorbent Assay , Fucose/chemistry , Hemocyanins/chemistry , Immunoglobulin G/blood , Lipopolysaccharides/chemical synthesis , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Mice , Oligosaccharides/chemical synthesis , Oligosaccharides/chemistry , Pertussis Vaccine/chemistry , Pertussis Vaccine/immunology , Serum Albumin, Bovine/chemistry
6.
Front Immunol ; 10: 2519, 2019.
Article in English | MEDLINE | ID: mdl-31803174

ABSTRACT

Gout is characterized by attacks of arthritis with hyperuricemia and monosodium urate (MSU) crystal-induced inflammation within joints. Innate immune responses are the primary drivers for tissue destruction and inflammation in gout. MSU crystals engage the Nlrp3 inflammasome, leading to the activation of caspase-1 and production of IL-1ß and IL-18 within gout-affected joints, promoting the influx of neutrophils and monocytes. Here, we show that caspase-11-/- mice and their derived macrophages produce significantly reduced levels of gout-specific cytokines including IL-1ß, TNFα, IL-6, and KC, while others like IFNγ and IL-12p70 are not altered. IL-1ß induces the expression of caspase-11 in an IL-1 receptor-dependent manner in macrophages contributing to the priming of macrophages during sterile inflammation. The absence of caspase-11 reduced the ability of macrophages and neutrophils to migrate in response to exogenously injected KC in vivo. Notably, in vitro, caspase-11-/- neutrophils displayed random migration in response to a KC gradient when compared to their WT counterparts. This phenotype was associated with altered cofilin phosphorylation. Unlike their wild-type counterparts, caspase-11-/- neutrophils also failed to produce neutrophil extracellular traps (NETs) when treated with MSU. Together, this is the first report demonstrating that caspase-11 promotes neutrophil directional trafficking and function in an acute model of gout. Caspase-11 also governs the production of inflammasome-dependent and -independent cytokines from macrophages. Our results offer new, previously unrecognized functions for caspase-11 in macrophages and neutrophils that may apply to other neutrophil-mediated disease conditions besides gout.


Subject(s)
Actin Depolymerizing Factors/metabolism , Arthritis, Gouty/etiology , Arthritis, Gouty/metabolism , Arthritis, Gouty/pathology , Caspases, Initiator/metabolism , Chemotaxis/immunology , Extracellular Traps/immunology , Neutrophils/immunology , Acute Disease , Animals , Biomarkers , Caspases, Initiator/genetics , Chemotaxis/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Extracellular Traps/metabolism , Gene Expression , Immunohistochemistry , Immunophenotyping , Inflammasomes/metabolism , Inflammation Mediators , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Neutrophils/metabolism , Phosphorylation , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction
7.
Infect Immun ; 87(10)2019 10.
Article in English | MEDLINE | ID: mdl-31308083

ABSTRACT

Bordetella bronchiseptica is an etiologic agent of respiratory diseases in animals and humans. Despite the widespread use of veterinary B. bronchiseptica vaccines, there is limited information on their composition and relative efficacy and on the immune responses that they elicit. Furthermore, human B. bronchiseptica vaccines are not available. We leveraged the dual antigenic and adjuvant functions of Bordetella colonization factor A (BcfA) to develop acellular B. bronchiseptica vaccines in the absence of an additional adjuvant. BALB/c mice immunized with BcfA alone or a trivalent vaccine containing BcfA and the Bordetella antigens FHA and Prn were equally protected against challenge with a prototype B. bronchiseptica strain. The trivalent vaccine protected mice significantly better than the canine vaccine Bronchicine and provided protection against a B. bronchiseptica strain isolated from a dog with kennel cough. Th1/17-polarized immune responses correlate with long-lasting protection against bordetellae and other respiratory pathogens. Notably, BcfA strongly attenuated the Th2 responses elicited by FHA and Prn, resulting in Th1/17-skewed responses in inherently Th2-skewed BALB/c mice. Thus, BcfA functions as both an antigen and an adjuvant, providing protection as a single-component vaccine. BcfA-adjuvanted vaccines may improve the efficacy and durability of vaccines against bordetellae and other pathogens.


Subject(s)
Adhesins, Bacterial/administration & dosage , Adjuvants, Immunologic/administration & dosage , Antigens, Bacterial/administration & dosage , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Vaccines/administration & dosage , Bordetella Infections/prevention & control , Bordetella bronchiseptica/drug effects , Virulence Factors, Bordetella/administration & dosage , Animals , Bordetella Infections/immunology , Bordetella Infections/microbiology , Bordetella bronchiseptica/immunology , Bordetella bronchiseptica/pathogenicity , Dogs , Female , Humans , Immunization , Immunogenicity, Vaccine , Male , Mice , Mice, Inbred BALB C , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/microbiology , Th1-Th2 Balance/drug effects , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/microbiology , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/microbiology
8.
J Vis Exp ; (144)2019 02 22.
Article in English | MEDLINE | ID: mdl-30855568

ABSTRACT

Vaccines are a 20th century medical marvel. They have dramatically reduced the morbidity and mortality caused by infectious diseases and contributed to a striking increase in life expectancy around the globe. Nonetheless, determining vaccine efficacy remains a challenge. Emerging evidence suggests that the current acellular vaccine (aPV) for Bordetella pertussis (B. pertussis) induces suboptimal immunity. Therefore, a major challenge is designing a next-generation vaccine that induces protective immunity without the adverse side effects of a whole-cell vaccine (wPV). Here we describe a protocol that we used to test the efficacy of a promising, novel adjuvant that skews immune responses to a protective Th1/Th17 phenotype and promotes a better clearance of a B. pertussis challenge from the murine respiratory tract. This article describes the protocol for mouse immunization, bacterial inoculation, tissue harvesting, and analysis of immune responses. Using this method, within our model, we have successfully elucidated crucial mechanisms elicited by a promising, next-generation acellular pertussis vaccine. This method can be applied to any infectious disease model in order to determine vaccine efficacy.


Subject(s)
Host-Pathogen Interactions , Pertussis Vaccine/immunology , Animals , Antibodies, Bacterial/immunology , Bordetella pertussis/immunology , Bordetella pertussis/physiology , Female , Male , Mice , Th17 Cells/immunology , Vaccination , Whooping Cough/prevention & control
9.
J Cyst Fibros ; 18(4): 491-500, 2019 07.
Article in English | MEDLINE | ID: mdl-30737168

ABSTRACT

Autophagy is a highly regulated, biological process that provides energy during periods of stress and starvation. This conserved process also acts as a defense mechanism and clears microbes from the host cell. Autophagy is impaired in Cystic Fibrosis (CF) patients and CF mice, as their cells exhibit low expression levels of essential autophagy molecules. The genetic disorder in CF is due to mutations in the cystic fibrosis transmembrane conductance regulator (cftr) gene that encodes for a chloride channel. CF patients are particularly prone to infection by pathogens that are otherwise cleared by autophagy in healthy immune cells including Burkholderia cenocepacia (B. cenocepacia). The objective of this study is to determine the mechanism underlying weak autophagic activity in CF macrophages and find therapeutic targets to correct it. Using reduced representation bisulfite sequencing (RRBS) to determine DNA methylation profile, we found that the promoter regions of Atg12 in CF macrophages are significantly more methylated than in the wild-type (WT) immune cells, accompanied by low protein expression. The natural product epigallocatechin-3-gallate (EGCG) significantly reduced the methylation of Atg12 promoter improving its expression. Accordingly, EGCG restricted B. cenocepacia replication within CF mice and their derived macrophages by improving autophagy and preventing dissemination. In addition, EGCG improved the function of CFTR protein. Altogether, utilizing RRBS for the first time in the CF field revealed a previously unrecognized mechanism for reduced autophagic activity in CF. Our data also offers a mechanism by which EGCG exerts its positive effects in CF.


Subject(s)
Autophagy , Cystic Fibrosis/physiopathology , Macrophages/physiology , Animals , Catechin/analogs & derivatives , Catechin/physiology , Cells, Cultured , Mice , Mice, Inbred C57BL
10.
Autophagy ; 14(11): 1928-1942, 2018.
Article in English | MEDLINE | ID: mdl-30165781

ABSTRACT

CASP4/caspase-11-dependent inflammasome activation is important for the clearance of various Gram-negative bacteria entering the host cytosol. Additionally, CASP4 modulates the actin cytoskeleton to promote the maturation of phagosomes harboring intracellular pathogens such as Legionella pneumophila but not those enclosing nonpathogenic bacteria. Nevertheless, this non-inflammatory role of CASP4 regarding the trafficking of vacuolar bacteria remains poorly understood. Macroautophagy/autophagy, a catabolic process within eukaryotic cells, is also implicated in the elimination of intracellular pathogens such as Burkholderia cenocepacia. Here we show that CASP4-deficient macrophages exhibit a defect in autophagosome formation in response to B. cenocepacia infection. The absence of CASP4 causes an accumulation of the small GTPase RAB7, reduced colocalization of B. cenocepacia with LC3 and acidic compartments accompanied by increased bacterial replication in vitro and in vivo. Together, our data reveal a novel role of CASP4 in regulating autophagy in response to B. cenocepacia infection.


Subject(s)
Autophagosomes/metabolism , Autophagy/genetics , Bacterial Infections/immunology , Burkholderia cenocepacia/immunology , Caspases/physiology , Animals , Autophagosomes/microbiology , Autophagy/immunology , Bacterial Infections/genetics , Bacterial Infections/metabolism , Burkholderia Infections/genetics , Burkholderia Infections/immunology , Burkholderia Infections/metabolism , Burkholderia cenocepacia/metabolism , Caspases/genetics , Caspases, Initiator , Cells, Cultured , Escherichia coli/immunology , Escherichia coli/metabolism , Inflammasomes/genetics , Inflammasomes/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagosomes/genetics , Phagosomes/metabolism , Phagosomes/microbiology , Phagosomes/pathology
11.
J Cyst Fibros ; 17(4): 454-461, 2018 07.
Article in English | MEDLINE | ID: mdl-29241629

ABSTRACT

INTRODUCTION: Cystic fibrosis (CF) is a multi-organ disorder characterized by chronic sino-pulmonary infections and inflammation. Many patients with CF suffer from repeated pulmonary exacerbations that are predictors of worsened long-term morbidity and mortality. There are no reliable markers that associate with the onset or progression of an exacerbation or pulmonary deterioration. Previously, we found that the Mirc1/Mir17-92a cluster which is comprised of 6 microRNAs (Mirs) is highly expressed in CF mice and negatively regulates autophagy which in turn improves CF transmembrane conductance regulator (CFTR) function. Therefore, here we sought to examine the expression of individual Mirs within the Mirc1/Mir17-92 cluster in human cells and biological fluids and determine their role as biomarkers of pulmonary exacerbations and response to treatment. METHODS: Mirc1/Mir17-92 cluster expression was measured in human CF and non-CF plasma, blood-derived neutrophils, and sputum samples. Values were correlated with pulmonary function, exacerbations and use of CFTR modulators. RESULTS: Mirc1/Mir17-92 cluster expression was not significantly elevated in CF neutrophils nor plasma when compared to the non-CF cohort. Cluster expression in CF sputum was significantly higher than its expression in plasma. Elevated CF sputum Mirc1/Mir17-92 cluster expression positively correlated with pulmonary exacerbations and negatively correlated with lung function. Patients with CF undergoing treatment with the CFTR modulator Ivacaftor/Lumacaftor did not demonstrate significant change in the expression Mirc1/Mir17-92 cluster after six months of treatment. CONCLUSIONS: Mirc1/Mir17-92 cluster expression is a promising biomarker of respiratory status in patients with CF including pulmonary exacerbation.


Subject(s)
Aminophenols/administration & dosage , Aminopyridines/administration & dosage , Benzodioxoles/administration & dosage , Cystic Fibrosis , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , Quinolones/administration & dosage , Respiratory System , Adolescent , Adult , Biomarkers/metabolism , Chloride Channel Agonists/administration & dosage , Correlation of Data , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Progression , Drug Combinations , Drug Monitoring/methods , Female , Gene Expression Profiling , Humans , Male , RNA, Long Noncoding , Respiratory Function Tests/methods , Respiratory System/drug effects , Respiratory System/metabolism , Respiratory System/physiopathology , Sputum/metabolism
12.
Autophagy ; 12(11): 2026-2037, 2016 11.
Article in English | MEDLINE | ID: mdl-27541364

ABSTRACT

Cystic fibrosis (CF) is a fatal, genetic disorder that critically affects the lungs and is directly caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, resulting in defective CFTR function. Macroautophagy/autophagy is a highly regulated biological process that provides energy during periods of stress and starvation. Autophagy clears pathogens and dysfunctional protein aggregates within macrophages. However, this process is impaired in CF patients and CF mice, as their macrophages exhibit limited autophagy activity. The study of microRNAs (Mirs), and other noncoding RNAs, continues to offer new therapeutic targets. The objective of this study was to elucidate the role of Mirs in dysregulated autophagy-related genes in CF macrophages, and then target them to restore this host-defense function and improve CFTR channel function. We identified the Mirc1/Mir17-92 cluster as a potential negative regulator of autophagy as CF macrophages exhibit decreased autophagy protein expression and increased cluster expression when compared to wild-type (WT) counterparts. The absence or reduced expression of the cluster increases autophagy protein expression, suggesting the canonical inverse relationship between Mirc1/Mir17-92 and autophagy gene expression. An in silico study for targets of Mirs that comprise the cluster suggested that the majority of the Mirs target autophagy mRNAs. Those targets were validated by luciferase assays. Notably, the ability of macrophages expressing mutant F508del CFTR to transport halide through their membranes is compromised and can be restored by downregulation of these inherently elevated Mirs, via restoration of autophagy. In vivo, downregulation of Mir17 and Mir20a partially restored autophagy expression and hence improved the clearance of Burkholderia cenocepacia. Thus, these data advance our understanding of mechanisms underlying the pathobiology of CF and provide a new therapeutic platform for restoring CFTR function and autophagy in patients with CF.


Subject(s)
Autophagy/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Gene Expression Regulation , Macrophages/metabolism , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Antagomirs/pharmacology , Autophagy/drug effects , Autophagy-Related Proteins/metabolism , Burkholderia cenocepacia/physiology , Cells, Cultured , Cystic Fibrosis/microbiology , Gene Expression Regulation/drug effects , Homozygote , Lung/metabolism , Lung/microbiology , Lung/pathology , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , NIH 3T3 Cells
13.
Epigenetics ; 11(5): 381-8, 2016 05 03.
Article in English | MEDLINE | ID: mdl-26909551

ABSTRACT

Autophagy is a biological process characterized by self-digestion and involves induction of autophagosome formation, leading to degradation of autophagic cargo. Aging is associated with the reduction of autophagy activity leading to neurodegenerative disorders, chronic inflammation, and susceptibility to infection; however, the underlying mechanism is unclear. DNA methylation by DNA methyltransferases reduces the expression of corresponding genes. Since macrophages are major players in inflammation and defense against infection we determined the differences in methylation of autophagy genes in macrophages derived from young and aged mice. We found that promoter regions of Atg5 and LC3B are hypermethylated in macrophages from aged mice and this is accompanied by low gene expression. Treatment of aged mice and their derived macrophages with methyltransferase inhibitor (2)-epigallocatechin-3-gallate (EGCG) or specific DNA methyltransferase 2 (DNMT2) siRNA restored the expression of Atg5 and LC3 in vivo and in vitro. Our study builds a foundation for the development of novel therapeutics aimed to improve autophagy in the elderly population and suggests a role for DNMT2 in DNA methylation activities.


Subject(s)
Aging/genetics , Autophagy-Related Protein 5/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Microtubule-Associated Proteins/genetics , Aging/pathology , Animals , Autophagosomes/drug effects , Autophagy/drug effects , Autophagy/genetics , Catechin/administration & dosage , Catechin/analogs & derivatives , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/drug effects , Enzyme Inhibitors/administration & dosage , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice , RNA, Small Interfering/genetics
14.
Sci Rep ; 5: 18479, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26686473

ABSTRACT

Inflammasomes are multiprotein complexes that include members of the NOD-like receptor family and caspase-1. Caspase-1 is required for the fusion of the Legionella vacuole with lysosomes. Caspase-11, independently of the inflammasome, also promotes phagolysosomal fusion. However, it is unclear how these proteases alter intracellular trafficking. Here, we show that caspase-11 and caspase-1 function in opposing manners to phosphorylate and dephosphorylate cofilin, respectively upon infection with Legionella. Caspase-11 targets cofilin via the RhoA GTPase, whereas caspase-1 engages the Slingshot phosphatase. The absence of either caspase-11 or caspase-1 maintains actin in the polymerized or depolymerized form, respectively and averts the fusion of pathogen-containing vacuoles with lysosomes. Therefore, caspase-11 and caspase-1 converge on the actin machinery with opposing effects to promote vesicular trafficking.


Subject(s)
Actins/metabolism , Caspase 1/genetics , Cofilin 1/genetics , Legionnaires' Disease/genetics , Phosphoprotein Phosphatases/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/genetics , Animals , Caspase 1/metabolism , Cofilin 1/metabolism , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Legionella pneumophila/genetics , Legionella pneumophila/pathogenicity , Legionnaires' Disease/metabolism , Legionnaires' Disease/pathology , Lysosomes/genetics , Lysosomes/metabolism , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphoprotein Phosphatases/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Vacuoles/genetics , Vacuoles/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , rhoA GTP-Binding Protein/genetics
15.
Article in English | MEDLINE | ID: mdl-23750338

ABSTRACT

Legionella pneumophila, the causative agent of Legionnaire's disease, replicates in human alveolar macrophages to establish infection. There is no human-to-human transmission and the main source of infection is L. pneumophila biofilms established in air conditioners, water fountains, and hospital equipments. The biofilm structure provides protection to the organism from disinfectants and antibacterial agents. L. pneumophila infection in humans is characterized by a subtle initial immune response, giving time for the organism to establish infection before the patient succumbs to pneumonia. Planktonic L. pneumophila elicits a strong immune response in murine, but not in human macrophages enabling control of the infection. Interactions between planktonic L. pneumophila and murine or human macrophages have been studied for years, yet the interface between biofilm-derived L. pneumophila and macrophages has not been explored. Here, we demonstrate that biofilm-derived L. pneumophila replicates significantly more in murine macrophages than planktonic bacteria. In contrast to planktonic L. pneumophila, biofilm-derived L. pneumophila lacks flagellin expression, do not activate caspase-1 or -7 and trigger less cell death. In addition, while planktonic L. pneumophila is promptly delivered to lysosomes for degradation, most biofilm-derived bacteria were enclosed in a vacuole that did not fuse with lysosomes in murine macrophages. This study advances our understanding of the innate immune response to biofilm-derived L. pneumophila and closely reproduces the natural mode of infection in human.


Subject(s)
Biofilms/growth & development , Host-Pathogen Interactions , Immune Evasion , Immunity, Innate , Legionella pneumophila/immunology , Macrophages/immunology , Macrophages/microbiology , Animals , Bacterial Load , Legionella pneumophila/growth & development , Legionella pneumophila/isolation & purification , Legionella pneumophila/physiology , Mice , Mice, Inbred C57BL
16.
Eur J Immunol ; 43(5): 1333-44, 2013 May.
Article in English | MEDLINE | ID: mdl-23420491

ABSTRACT

Legionella pneumophila (L. pneumophila) is an intracellular bacterium of human alveolar macrophages that causes Legionnaires' disease. In contrast to humans, most inbred mouse strains are restrictive to L. pneumophila replication. We demonstrate that autophagy targets L. pneumophila vacuoles to lysosomes and that this process requires ubiquitination of L. pneumophila vacuoles and the subsequent binding of the autophagic adaptor p62/SQSTM1 to ubiquitinated vacuoles. The L. pneumophila legA9 encodes for an ankyrin-containing protein with unknown role. We show that the legA9 mutant replicate in WT mice and their bone marrow-derived macrophages. This is the first L. pneumophila mutant to be found to replicate in WT bone marrow-derived macrophages other than the Fla mutant. Less legA9 mutant-containing vacuoles acquired ubiquitin labeling and p62/SQSTM1 staining, evading autophagy uptake and avoiding lysosomal fusion. Thus, we describe a bacterial protein that targets the L. pneumophila-containing vacuole for autophagy uptake.


Subject(s)
Autophagy/immunology , Bacterial Proteins/genetics , Legionella pneumophila/genetics , Macrophages/microbiology , Mutation , Vacuoles/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Bacterial Proteins/immunology , Cells, Cultured , Heat-Shock Proteins/genetics , Heat-Shock Proteins/immunology , Host Specificity , Host-Pathogen Interactions , Humans , Immune Evasion/genetics , Legionella pneumophila/immunology , Lysosomes/immunology , Lysosomes/metabolism , Lysosomes/microbiology , Macrophages/immunology , Mice , Phagosomes/immunology , Phagosomes/metabolism , Phagosomes/microbiology , Protein Binding , Sequestosome-1 Protein , Ubiquitin/metabolism , Ubiquitination , Vacuoles/metabolism , Vacuoles/microbiology
17.
J Biol Chem ; 288(3): 2049-58, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23148214

ABSTRACT

Cystic fibrosis is the most common inherited lethal disease in Caucasians. It is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR), of which the cftr ΔF508 mutation is the most common. ΔF508 macrophages are intrinsically defective in autophagy because of the sequestration of essential autophagy molecules within unprocessed CFTR aggregates. Defective autophagy allows Burkholderia cenocepacia (B. cepacia) to survive and replicate in ΔF508 macrophages. Infection by B. cepacia poses a great risk to cystic fibrosis patients because it causes accelerated lung inflammation and, in some cases, a lethal necrotizing pneumonia. Autophagy is a cell survival mechanism whereby an autophagosome engulfs non-functional organelles and delivers them to the lysosome for degradation. The ubiquitin binding adaptor protein SQSTM1/p62 is required for the delivery of several ubiquitinated cargos to the autophagosome. In WT macrophages, p62 depletion and overexpression lead to increased and decreased bacterial intracellular survival, respectively. In contrast, depletion of p62 in ΔF508 macrophages results in decreased bacterial survival, whereas overexpression of p62 leads to increased B. cepacia intracellular growth. Interestingly, the depletion of p62 from ΔF508 macrophages results in the release of the autophagy molecule beclin1 (BECN1) from the mutant CFTR aggregates and allows its redistribution and recruitment to the B. cepacia vacuole, mediating the acquisition of the autophagy marker LC3 and bacterial clearance via autophagy. These data demonstrate that p62 differentially dictates the fate of B. cepacia infection in WT and ΔF508 macrophages.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autophagy/genetics , Burkholderia Infections/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/genetics , Heat-Shock Proteins/genetics , Macrophages/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biomarkers/metabolism , Burkholderia Infections/complications , Burkholderia Infections/metabolism , Burkholderia Infections/microbiology , Burkholderia cenocepacia/physiology , Cystic Fibrosis/complications , Cystic Fibrosis/metabolism , Cystic Fibrosis/microbiology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gene Expression , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Humans , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Transgenic , Microbial Viability , Microtubule-Associated Proteins/metabolism , Phagosomes/metabolism , Protein Transport , RNA, Small Interfering/genetics , Sequestosome-1 Protein , Transfection , Ubiquitin/genetics , Ubiquitin/metabolism
18.
Immunity ; 37(1): 35-47, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22658523

ABSTRACT

Inflammasomes are multiprotein complexes that include members of the NLR (nucleotide-binding domain leucine-rich repeat containing) family and caspase-1. Once bacterial molecules are sensed within the macrophage, the inflammasome is assembled, mediating the activation of caspase-1. Caspase-11 mediates caspase-1 activation in response to lipopolysaccharide and bacterial toxins, and yet its role during bacterial infection is unknown. Here, we demonstrated that caspase-11 was dispensable for caspase-1 activation in response to Legionella, Salmonella, Francisella, and Listeria. We also determined that active mouse caspase-11 was required for restriction of L. pneumophila infection. Similarly, human caspase-4 and caspase-5, homologs of mouse caspase-11, cooperated to restrict L. pneumophila infection in human macrophages. Caspase-11 promoted the fusion of the L. pneumophila vacuole with lysosomes by modulating actin polymerization through cofilin. However, caspase-11 was dispensable for the fusion of lysosomes with phagosomes containing nonpathogenic bacteria, uncovering a fundamental difference in the trafficking of phagosomes according to their cargo.


Subject(s)
Actins/metabolism , Bacteria/immunology , Caspases/metabolism , Lysosomes/metabolism , Phagosomes/metabolism , Protein Multimerization , Actin Depolymerizing Factors/metabolism , Animals , Bacteria/growth & development , Bacterial Infections/immunology , Bacterial Infections/metabolism , Caspase 1/deficiency , Caspase 1/genetics , Caspase 1/metabolism , Caspases/deficiency , Caspases/genetics , Caspases, Initiator , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagosomes/microbiology , Phosphorylation
19.
Biochem Biophys Res Commun ; 424(2): 221-7, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22728038

ABSTRACT

Cystic fibrosis (CF) is accompanied with heightened inflammation worsened by drug resistant Burkholderia cenocepacia. Human CF macrophage responses to B. cenocepacia are poorly characterized and variable in the literature. Therefore, we examined human macrophage responses to the epidemic B. cenocepacia J2315 strain in order to identify novel anti-inflammatory targets. Peripheral blood monocyte derived macrophages were obtained from 23 CF and 27 non-CF donors. Macrophages were infected with B. cenocepacia J2315 and analyzed for cytokines, cytotoxicity, and microscopy. CF macrophages demonstrated significant increases in IL-1ß, IL-10, MCP-1, and IFN-γ production in comparison to non-CF controls. CF patients on prednisone exhibited globally diminished cytokines compared to controls and other CF patients. CF macrophages also displayed increased bacterial burden and cell death. In conclusion, CF macrophages demonstrate exaggerated IL-1ß, IL-10, MCP-1, and IFN-γ production and cell death during B. cenocepacia infection. Treatment with corticosteroids acutely suppressed cytokine responses.


Subject(s)
Burkholderia Infections/microbiology , Burkholderia cenocepacia , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Macrophages/immunology , Adolescent , Adrenal Cortex Hormones/pharmacology , Adult , Apoptosis , Chemokine CCL2/metabolism , Child , Child, Preschool , Cystic Fibrosis/immunology , Female , Humans , Infant , Inflammation/microbiology , Inflammation/pathology , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Lysosomes/microbiology , Macrophages/drug effects , Macrophages/ultrastructure , Male , Microscopy, Electron , Middle Aged , Young Adult
20.
Autophagy ; 7(11): 1359-70, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21997369

ABSTRACT

Cystic fibrosis (CF) is the most common inherited lethal disease of Caucasians which results in multi organ dysfunction. However, 85% of the deaths are due to pulmonary infections. Infection by Burkholderia cenocepacia (B. cepacia) is a particularly lethal threat to CF patients because it causes severe and persistent lung inflammation and is resistant to nearly all available antibiotics. In CFTR ΔF508 mouse macrophages, B. cepacia persists in vacuoles that do not fuse with the lysosomes and mediates increased production of IL-1ß. It is believed that intracellular bacterial survival contributes to the persistence of the bacterium. Here we show for the first time that in wild-type macrophages but not in ΔF508 macrophages, many B. cepacia reside in autophagosomes that fuse with lysosomes at later stages of infection. Accordingly, association and intracellular survival of B. cepacia are higher in CFTR-ΔF508 (ΔF508) macrophages than in WT macrophages. An autophagosome is a compartment that engulfs non-functional organelles and parts of the cytoplasm then delivers them to the lysosome for degradation to produce nutrients during periods of starvation or stress. Furthermore, we show that B. cepacia downregulates autophagy genes in WT and ΔF508 macrophages. However, autophagy dysfunction is more pronounced in ΔF508 macrophages since they already have compromised autophagy activity. We demonstrate that the autophagy-stimulating agent, rapamycin markedly decreases B. cepacia infection in vitro by enhancing the clearance of B. cepacia via induced autophagy. In vivo, Rapamycin decreases bacterial burden in the lungs of CF mice and drastically reduces signs of lung inflammation. Together, our studies reveal that if efficiently activated, autophagy can control B. cepacia infection and ameliorate the associated inflammation. Therefore, autophagy is a novel target for new drug development for CF patients to control B. cepacia infection and accompanying inflammation.


Subject(s)
Autophagy/drug effects , Burkholderia Infections/drug therapy , Burkholderia cenocepacia/physiology , Cystic Fibrosis/drug therapy , Pneumonia/drug therapy , Sirolimus/pharmacology , Sirolimus/therapeutic use , Animals , Autophagy/genetics , Burkholderia Infections/complications , Burkholderia Infections/microbiology , Burkholderia Infections/pathology , Burkholderia cenocepacia/drug effects , Burkholderia cenocepacia/growth & development , Burkholderia cenocepacia/ultrastructure , Cystic Fibrosis/complications , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Disease Models, Animal , Down-Regulation/genetics , Interleukin-1beta/biosynthesis , Intracellular Space/drug effects , Intracellular Space/microbiology , Lysosomes/drug effects , Lysosomes/microbiology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Microbial Viability/drug effects , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Phagosomes/drug effects , Phagosomes/microbiology , Phagosomes/ultrastructure , Pneumonia/complications , Pneumonia/microbiology , RNA, Small Interfering/metabolism , Vacuoles/drug effects , Vacuoles/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...