Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Anaerobe ; 71: 102399, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34090994

ABSTRACT

Periodontal disease, an inflammatory bone disease of the oral cavity, affects more than 50% of the United States population over the age of 30. The Gram-negative, anaerobic bacterium Porphyromonas gingivalis, the etiological agent of periodontal disease, is known to induce dysbiosis of the oral microbiome while promoting inflammatory bone loss. We have recently reported that P. gingivalis can also alter the gut microbiota of mice prone to develop inflammatory atherosclerosis. However, it is still unknown whether P. gingivalis induces similar changes to the gut microbiome as it does to oral microbiome. In this study, we demonstrate that P. gingivalis infection increases the diversity of the oral microbiome, allowing for colonization of potentially opportunistic species in the oral microbiome and overgrowth of commensal species in both the oral and gut microbiomes. Since periodontal disease treatment in humans typically involves antibiotic treatment, we also examined the combined effect of P. gingivalis infection on mice pretreated with oral antibiotics. By correlating the oral and cecal microbiota of P. gingivalis-infected mice fed a normal chow diet, we identified blooms of the Gram-negative genera Barnesiella and Bacteroides and imbalances of mucin-degrading bacteria. These disrupted community structures were predicted to have increased detrimental functional capacities including increased flavonoid degradation and l-histidine fermentation. Though antibiotic pretreatment (without P. gingivlais) had a dominant impact on the cecal microbiome, P. gingivalis infection of mice with or without antibiotic pretreatment increased the abundance of the phylum Firmicutes and the Porphyromonadaceae family in the cecum. Collectively, our study demonstrates that P. gingivalis oral infection disrupted the oral and cecal microbiomes of otherwise unperturbed mice, altering their community membership and functional potential.


Subject(s)
Gastrointestinal Microbiome , Mouth/microbiology , Periodontal Diseases/microbiology , Porphyromonas gingivalis/physiology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Bacteria/isolation & purification , Dysbiosis/microbiology , Humans , Male , Mice , Mice, Inbred C57BL , Microbiota , Phylogeny , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/isolation & purification
2.
PLoS One ; 14(1): e0210827, 2019.
Article in English | MEDLINE | ID: mdl-30682073

ABSTRACT

Interleukin (IL)-33 is an interleukin-1 like cytokine that enhances Th2 responses and mediates mucosal immunity and allergic inflammation but the mechanism regulating endogenous IL-33 production are still under investigation. In macrophages, lipopolysaccharide (LPS) administration resulted in marked induction of IL-33 mRNA that was blunted in macrophages from glutaredoxin-1 (Glrx) knockout mice and in RAW264.7 macrophages with Glrx knockdown by siRNA. Glutaredoxin-1 is a small cytosolic thioltransferase that controls a reversible protein thiol modification, S-glutationylation (protein-GSH adducts), thereby regulating redox signaling. In this study, we examined the mechanism of Glrx regulation of endogenous IL-33 induction in macrophages. Glrx knockdown resulted in impaired de-glutathionylation of TRAF6, which is required for TRAF6 activation, and inhibited downstream IKKß and NF-κB activation. Inhibitors of NF-κB suppressed IL-33 induction and chromatin IP sequencing data analysis confirmed that IL-33 is an NF-κB-responsive gene. Since TRAF6-NF-κB activation is also essential for IL-33 signaling through its receptor, ST2L, we next tested the involvement of Glrx in exogenous IL-33 responses in RAW264.7 cells. Recombinant IL-33 (rIL-33) administration induced IL-33 mRNA expression in RAW264.7 macrophages, and this was inhibited by Glrx knockdown. Interestingly, rIL-33-induced IL-33 protein was identified as the 20 kDa cleaved form whereas LPS-induced IL-33 protein was identified as full-length IL-33, which may be less active than the cleaved form. In a clinically-relevant mouse model of asthma, intra-tracheal cockroach antigen treatment induced Glrx protein in wild type mouse lungs but Glrx induction was attenuated in IL-33 knockout mouse lungs, suggesting that IL-33 may regulate Glrx induction in vivo in response to allergen challenge. In summary, our data reveal a novel mechanism by which Glrx controls both LPS- and IL-33-mediated NF-κB activation leading to IL-33 production, and paracrine IL-33 can induce Glrx to further regulate inflammatory reactions.


Subject(s)
Glutaredoxins/metabolism , Interleukin-33/biosynthesis , Interleukin-33/genetics , Macrophages/immunology , Macrophages/metabolism , Allergens/administration & dosage , Animals , Asthma/etiology , Asthma/immunology , Asthma/metabolism , Disease Models, Animal , Gene Expression/drug effects , Gene Knockdown Techniques , Glutaredoxins/deficiency , Glutaredoxins/genetics , Glutathione/metabolism , Lipopolysaccharides/pharmacology , Lung/immunology , Lung/metabolism , Macrophages/drug effects , Mice , Mice, Knockout , NF-kappa B/metabolism , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism
3.
Int J Oral Sci ; 10(4): 32, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30420594

ABSTRACT

Oral squamous cell carcinoma (OSCC) is the most prevalent and most commonly studied oral cancer. However, there is a void regarding the role that the oral microbiome may play in OSCC. Although the relationship between microbial community composition and OSCC has been thoroughly investigated, microbial profiles of the human microbiome in cancer are understudied. Here we performed a small pilot study of community-wide metatranscriptome analysis to profile mRNA expression in the entire oral microbiome in OSCC to reveal molecular functions associated with this disease. Fusobacteria showed a statistically significantly higher number of transcripts at tumour sites and tumour-adjacent sites of cancer patients compared to the healthy controls analysed. Regardless of the community composition, specific metabolic signatures were consistently found in disease. Activities such as iron ion transport, tryptophanase activity, peptidase activities and superoxide dismutase were over-represented in tumour and tumour-adjacent samples when compared to the healthy controls. The expression of putative virulence factors in the oral communities associated with OSCC showed that activities related to capsule biosynthesis, flagellum synthesis and assembly, chemotaxis, iron transport, haemolysins and adhesins were upregulated at tumour sites. Moreover, activities associated with protection against reactive nitrogen intermediates, chemotaxis, flagellar and capsule biosynthesis were also upregulated in non-tumour sites of cancer patients. Although they are preliminary, our results further suggest that Fusobacteria may be the leading phylogenetic group responsible for the increase in expression of virulence factors in the oral microbiome of OSCC patients.


Subject(s)
Carcinoma, Squamous Cell/microbiology , Metagenome , Microbiota , Mouth Neoplasms/microbiology , Transcriptome , Virulence Factors/metabolism , Humans , Phylogeny , Pilot Projects , RNA, Messenger/metabolism , Virulence
4.
J Asthma Allergy ; 11: 159-171, 2018.
Article in English | MEDLINE | ID: mdl-30122959

ABSTRACT

BACKGROUND: Purinergic receptors control cell proliferation, apoptosis, migration, inflammation, and cytokine secretion. Increased expression of specific purinergic receptors is reported in asthma. The role of purinergic P2Y6 receptors (P2Y6R) in asthma is controversial. HYPOTHESIS: P2Y6R activation in asthma improves pulmonary function and reduces inflammation and smooth muscle amount. METHODS: Female mice (C57/BL6, age 30 days) were randomly assigned to receive intranasal house dust mite (HDM) antigen (40 or 80 µg) or saline, 5 days/week, for 6 weeks. Randomly selected subgroups received intraperitoneal P2Y6R agonist prodrug (GC021109; 10 or 100 µg/kg weight/dose) simultaneously with HDM. After 6 weeks, lung function was measured. Lung lavage fluid (LLF) was used to measure total cell count, total protein, and cytokines. Immunohistochemistry for alpha smooth muscle actin (α-SMA) was done. Airway wall thickness was measured on micro-computed tomography (micro-CT) images. RESULTS: Pulmonary function testing revealed a HDM dose-dependent airway hyperresponsiveness. Airway resistance was increased 2-fold while compliance was decreased by 50% at the higher HDM dose (P<0.05). GC021109 prevented these changes. HDM-exposed mice had elevated inflammatory cell and total protein levels in LLF which were prevented by GC021109 (P<0.05). HDM mice also had elevated LLF levels of interleukin (IL)-4, IL-5, IL-12, granulocyte colony stimulating factor, chemokine (C-X-C) motif ligand 1, and leukemia inhibitory factor that were reduced by GC021109 with a dose-dependent pattern. HDM mice had increased peribronchial and perivascular inflammatory cell infiltration and increased α-SMA; these changes were absent with GC021109. Airway wall thickness measured on micro-CT images was increased after HDM exposure and significantly reduced by GC021109 treatment. CONCLUSION: The P2Y6R prodrug GC021109 inhibited allergen-induced changes in pulmonary function, inflammatory responses, and airway and vascular smooth muscle mass. P2Y6R activation may be an effective therapeutic maintenance strategy in asthma.

5.
Anaerobe ; 45: 19-30, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28442421

ABSTRACT

Mounting evidence in humans supports an etiological role for the microbiota in inflammatory atherosclerosis. Atherosclerosis is a progressive disease characterized by accumulation of inflammatory cells and lipids in vascular tissue. While retention of lipoprotein into the sub-endothelial vascular layer is believed to be the initiating stimulus leading to the development of atherosclerosis, activation of multiple pathways related to vascular inflammation and endothelial dysfunction sustain the process by stimulating recruitment of leukocytes and immune cells into the sub-endothelial layer. The Gram-negative oral pathogen Porphyromonas gingivalis has been associated with the development and acceleration of atherosclerosis in humans and these observations have been validated in animal models. It has been proposed that common mechanisms of immune signaling link stimulation by lipids and pathogens to vascular inflammation. Despite the common outcome of P. gingivalis and lipid feeding on atherosclerosis progression, we established that these pro-atherogenic stimuli induced distinct gene signatures in the ApoE-/- mouse model of atherosclerosis. In this study, we further defined the distinct roles of dietary lipids and P. gingivalis infection on atherosclerosis progression and the gut microbiota. We demonstrate that diet-induced lipid lowering resulted in less atherosclerotic plaque in ApoE-/- mice compared to ApoE-/- mice continuously fed a Western diet. However, the effect of diet-induced lipid lowering on plaque accumulation was blunted by P. gingivalis infection. Using principal component analysis and hierarchical clustering, we demonstrate that dietary intervention as well as P. gingivalis infection result in distinct bacterial communities in fecal and cecal samples of ApoE-/- mice as compared to ApoE-/- mice continuously fed either a Western diet or a normal chow diet. Collectively, we identified distinct microbiota changes accompanying atherosclerotic plaque, suggesting a future avenue for investigation on the impact of the gut microbiota, diet, and P. gingivalis infection on atherosclerosis.


Subject(s)
Atherosclerosis/physiopathology , Bacterial Infections/complications , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Lipid Metabolism , Porphyromonas gingivalis/pathogenicity , Animals , Disease Models, Animal , Male , Mice, Inbred C57BL
6.
PLoS One ; 10(7): e0131688, 2015.
Article in English | MEDLINE | ID: mdl-26148065

ABSTRACT

INTRODUCTION: Diverse and multi-factorial processes contribute to the progression of cardiovascular disease. These processes affect cells involved in the development of this disease in varying ways, ultimately leading to atherothrombosis. The goal of our study was to compare the differential effects of specific stimuli--two bacterial infections and a Western diet--on platelet responses in ApoE-/- mice, specifically examining inflammatory function and gene expression. Results from murine studies were verified using platelets from participants of the Framingham Heart Study (FHS; n = 1819 participants). METHODS: Blood and spleen samples were collected at weeks 1 and 9 from ApoE-/- mice infected with Porphyromonas gingivalis or Chlamydia pneumoniae and from mice fed a Western diet for 9 weeks. Transcripts based on data from a Western diet in ApoE-/- mice were measured in platelet samples from FHS using high throughput qRT-PCR. RESULTS: At week 1, both bacterial infections increased circulating platelet-neutrophil aggregates. At week 9, these cells individually localized to the spleen, while Western diet resulted in increased platelet-neutrophil aggregates in the spleen only. Microarray analysis of platelet RNA from infected or Western diet-fed mice at week 1 and 9 showed differential profiles. Genes, such as Serpina1a, Ttr, Fgg, Rpl21, and Alb, were uniquely affected by infection and diet. Results were reinforced in platelets obtained from participants of the FHS. CONCLUSION: Using both human studies and animal models, results demonstrate that variable sources of inflammatory stimuli have the ability to influence the platelet phenotype in distinct ways, indicative of the diverse function of platelets in thrombosis, hemostasis, and immunity.


Subject(s)
Blood Platelets/pathology , Diet, Western/adverse effects , Inflammation/pathology , Platelet Aggregation/physiology , Animals , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Blood Platelets/metabolism , Blood Platelets/microbiology , Chlamydophila pneumoniae/pathogenicity , Disease Models, Animal , Humans , Inflammation/metabolism , Inflammation/microbiology , Male , Mice , Neutrophils/microbiology , Neutrophils/pathology , Neutrophils/physiology , Porphyromonas gingivalis/pathogenicity , Thrombosis/metabolism , Thrombosis/pathology
7.
BMC Genomics ; 15: 1176, 2014 Dec 24.
Article in English | MEDLINE | ID: mdl-25540039

ABSTRACT

BACKGROUND: Atherosclerosis is a progressive disease characterized by inflammation and accumulation of lipids in vascular tissue. Porphyromonas gingivalis (Pg) and Chlamydia pneumoniae (Cp) are associated with inflammatory atherosclerosis in humans. Similar to endogenous mediators arising from excessive dietary lipids, these Gram-negative pathogens are pro-atherogenic in animal models, although the specific inflammatory/atherogenic pathways induced by these stimuli are not well defined. In this study, we identified gene expression profiles that characterize P. gingivalis, C. pneumoniae, and Western diet (WD) at acute and chronic time points in aortas of Apolipoprotein E (ApoE-/-) mice. RESULTS: At the chronic time point, we observed that P. gingivalis was associated with a high number of unique differentially expressed genes compared to C. pneumoniae or WD. For the top 500 differentially expressed genes unique to each group, we observed a high percentage (76%) that exhibited decreased expression in P. gingivalis-treated mice in contrast to a high percentage (96%) that exhibited increased expression in WD mice. C. pneumoniae treatment resulted in approximately equal numbers of genes that exhibited increased and decreased expression. Gene Set Enrichment Analysis (GSEA) revealed distinct stimuli-associated phenotypes, including decreased expression of mitochondrion, glucose metabolism, and PPAR pathways in response to P. gingivalis but increased expression of mitochondrion, lipid metabolism, carbohydrate and amino acid metabolism, and PPAR pathways in response to C. pneumoniae; WD was associated with increased expression of immune and inflammatory pathways. DAVID analysis of gene clusters identified by two-way ANOVA at acute and chronic time points revealed a set of core genes that exhibited altered expression during the natural progression of atherosclerosis in ApoE-/- mice; these changes were enhanced in P. gingivalis-treated mice but attenuated in C. pneumoniae-treated mice. Notable differences in the expression of genes associated with unstable plaques were also observed among the three pro-atherogenic stimuli. CONCLUSIONS: Despite the common outcome of P. gingivalis, C. pneumoniae, and WD on the induction of vascular inflammation and atherosclerosis, distinct gene signatures and pathways unique to each pro-atherogenic stimulus were identified. Our results suggest that pathogen exposure results in dysregulated cellular responses that may impact plaque progression and regression pathways.


Subject(s)
Aorta/metabolism , Apolipoproteins E/deficiency , Chlamydophila pneumoniae/physiology , Diet, Western/adverse effects , Gene Expression Profiling , Porphyromonas gingivalis/physiology , Animals , Aorta/pathology , Kinetics , Male , Mice , Mice, Inbred C57BL , Multigene Family/genetics , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/microbiology , Plaque, Atherosclerotic/pathology
8.
J Vis Exp ; (90): e51556, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-25146644

ABSTRACT

Chronic inflammation is a major driver of pathological tissue damage and a unifying characteristic of many chronic diseases in humans including neoplastic, autoimmune, and chronic inflammatory diseases. Emerging evidence implicates pathogen-induced chronic inflammation in the development and progression of chronic diseases with a wide variety of clinical manifestations. Due to the complex and multifactorial etiology of chronic disease, designing experiments for proof of causality and the establishment of mechanistic links is nearly impossible in humans. An advantage of using animal models is that both genetic and environmental factors that may influence the course of a particular disease can be controlled. Thus, designing relevant animal models of infection represents a key step in identifying host and pathogen specific mechanisms that contribute to chronic inflammation. Here we describe a mouse model of pathogen-induced chronic inflammation at local and systemic sites following infection with the oral pathogen Porphyromonas gingivalis, a bacterium closely associated with human periodontal disease. Oral infection of specific-pathogen free mice induces a local inflammatory response resulting in destruction of tooth supporting alveolar bone, a hallmark of periodontal disease. In an established mouse model of atherosclerosis, infection with P. gingivalis accelerates inflammatory plaque deposition within the aortic sinus and innominate artery, accompanied by activation of the vascular endothelium, an increased immune cell infiltrate, and elevated expression of inflammatory mediators within lesions. We detail methodologies for the assessment of inflammation at local and systemic sites. The use of transgenic mice and defined bacterial mutants makes this model particularly suitable for identifying both host and microbial factors involved in the initiation, progression, and outcome of disease. Additionally, the model can be used to screen for novel therapeutic strategies, including vaccination and pharmacological intervention.


Subject(s)
Bacteroidaceae Infections/microbiology , Disease Models, Animal , Inflammation/microbiology , Porphyromonas gingivalis/growth & development , Animals , Male , Mice , Mice, Transgenic , Mouth/microbiology , Porphyromonas gingivalis/genetics
9.
PLoS Pathog ; 10(7): e1004215, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25010102

ABSTRACT

Several successful pathogens have evolved mechanisms to evade host defense, resulting in the establishment of persistent and chronic infections. One such pathogen, Porphyromonas gingivalis, induces chronic low-grade inflammation associated with local inflammatory bone loss and systemic inflammation manifested as atherosclerosis. P. gingivalis expresses an atypical lipopolysaccharide (LPS) structure containing heterogeneous lipid A species, that exhibit Toll-like receptor-4 (TLR4) agonist or antagonist activity, or are non-activating at TLR4. In this study, we utilized a series of P. gingivalis lipid A mutants to demonstrate that antagonistic lipid A structures enable the pathogen to evade TLR4-mediated bactericidal activity in macrophages resulting in systemic inflammation. Production of antagonistic lipid A was associated with the induction of low levels of TLR4-dependent proinflammatory mediators, failed activation of the inflammasome and increased bacterial survival in macrophages. Oral infection of ApoE(-/-) mice with the P. gingivalis strain expressing antagonistic lipid A resulted in vascular inflammation, macrophage accumulation and atherosclerosis progression. In contrast, a P. gingivalis strain producing exclusively agonistic lipid A augmented levels of proinflammatory mediators and activated the inflammasome in a caspase-11-dependent manner, resulting in host cell lysis and decreased bacterial survival. ApoE(-/-) mice infected with this strain exhibited diminished vascular inflammation, macrophage accumulation, and atherosclerosis progression. Notably, the ability of P. gingivalis to induce local inflammatory bone loss was independent of lipid A expression, indicative of distinct mechanisms for induction of local versus systemic inflammation by this pathogen. Collectively, our results point to a pivotal role for activation of the non-canonical inflammasome in P. gingivalis infection and demonstrate that P. gingivalis evades immune detection at TLR4 facilitating chronic inflammation in the vasculature. These studies support the emerging concept that pathogen-mediated chronic inflammatory disorders result from specific pathogen-mediated evasion strategies resulting in low-grade chronic inflammation.


Subject(s)
Bacteroidaceae Infections/immunology , Lipid A/immunology , Porphyromonas gingivalis/immunology , Vasculitis/immunology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/immunology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/microbiology , Atherosclerosis/pathology , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Disease Models, Animal , HEK293 Cells , Humans , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Osteoporosis/genetics , Osteoporosis/immunology , Osteoporosis/microbiology , Osteoporosis/pathology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Vasculitis/genetics , Vasculitis/microbiology , Vasculitis/pathology
10.
Arterioscler Thromb Vasc Biol ; 34(3): 552-64, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24458711

ABSTRACT

OBJECTIVE: Interleukin 1 Receptor 1 (IL1R1) and its ligand, IL1ß, are upregulated in cardiovascular disease, obesity, and infection. Previously, we reported a higher level of IL1R1 transcripts in platelets from obese individuals of the Framingham Heart Study (FHS), but its functional effect in platelets has never been described. Additionally, IL1ß levels are increased in atherosclerotic plaques and in bacterial infections. The aim of this work is to determine whether IL1ß, through IL1R1, can activate platelets and megakaryocytes to promote atherothrombosis. APPROACH AND RESULTS: We found that IL1ß-related genes from platelets, as measured in 1819 FHS participants, were associated with increased body mass index, and a direct relationship was shown in wild-type mice fed a high-fat diet. Mechanistically, IL1ß activated nuclear factor-κB and mitogen-activated protein kinase signaling pathways in megakaryocytes. IL1ß, through IL1R1, increased ploidy of megakaryocytes to 64+ N by 2-fold over control. IL1ß increased agonist-induced platelet aggregation by 1.2-fold with thrombin and 4.2-fold with collagen. IL1ß increased adhesion to both collagen and fibrinogen, and heterotypic aggregation by 1.9-fold over resting. High fat diet-enhanced platelet adhesion was absent in IL1R1(-/-) mice. Wild-type mice infected with Porphyromonas gingivalis had circulating heterotypic aggregates (1.5-fold more than control at 24 hours and 6.2-fold more at 6 weeks) that were absent in infected IL1R1(-/-) and IL1ß(-/-) mice. CONCLUSIONS: In summary, IL1R1- and IL1ß-related transcripts are elevated in the setting of obesity. IL1R1/IL1ß augment both megakaryocyte and platelet functions, thereby promoting a prothrombotic environment during infection and obesity; potentially contributing to the development of atherothrombotic disease.


Subject(s)
Inflammation/pathology , Interleukin-1beta/physiology , Megakaryocytes/cytology , Obesity/blood , Platelet Activation/physiology , Receptors, Interleukin-1 Type I/physiology , Transcription, Genetic/physiology , Animals , Atherosclerosis/etiology , Bacteroidaceae Infections/blood , Bacteroidaceae Infections/pathology , Cell Line , Collagen/pharmacology , Dietary Fats/toxicity , Disease Models, Animal , Gene Expression Profiling , Humans , Imidazoles/pharmacology , Inflammation/etiology , Inflammation/genetics , Interleukin-1beta/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , NF-kappa B/metabolism , Obesity/complications , Obesity/genetics , Phosphorylation/drug effects , Platelet Activation/drug effects , Platelet Adhesiveness/drug effects , Platelet Adhesiveness/physiology , Porphyromonas gingivalis , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/genetics , Thrombin/pharmacology , Transcription, Genetic/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
11.
J Immunol ; 190(3): 1148-57, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23264656

ABSTRACT

Porphyromonas gingivalis is a primary etiological agent of chronic periodontal disease, an infection-driven chronic inflammatory disease that leads to the resorption of tooth-supporting alveolar bone. We previously reported that TLR2 is required for P. gingivalis-induced alveolar bone loss in vivo, and our in vitro work implicated TNF as a key downstream mediator. In this study, we show that TNF-deficient (Tnf(-/-)) mice are resistant to alveolar bone loss following oral infection with P. gingivalis, and thus establish a central role for TNF in experimental periodontal disease. Using bone marrow-derived macrophages (BMDM) from wild-type and gene-specific knockout mice, we demonstrate that the initial inflammatory response to P. gingivalis in naive macrophages is MyD88 dependent and requires cooperative signaling of TLR2 and TLR4. The ability of P. gingivalis to activate cells via TLR2 or TLR4 was confirmed in TLR2- or TLR4-transformed human embryonic kidney cells. Additional studies using bacterial mutants demonstrated a role for fimbriae in the modulation of TLR-mediated activation of NF-κB. Whereas both TLR2 and TLR4 contributed to TNF production in naive macrophages, P. gingivalis preferentially exploited TLR2 in endotoxin-tolerant BMDM to trigger excessive TNF production. We found that TNF induced surface TLR2 expression and augmented TLR-induced cytokine production in P. gingivalis-stimulated BMDM, establishing a previously unidentified TNF-dependent feedback loop. Adoptive transfer of TLR2-expressing macrophages to TLR2-deficient mice restored the ability of P. gingivalis to induce alveolar bone loss in vivo. Collectively, our results identify a TLR2- and TNF-dependent macrophage-specific mechanism underlying pathogen-induced inflammatory bone loss in vivo.


Subject(s)
Alveolar Bone Loss/etiology , Bacteroidaceae Infections/immunology , Gingivitis/physiopathology , Macrophages/physiology , Porphyromonas gingivalis/pathogenicity , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology , Tumor Necrosis Factor-alpha/physiology , Adoptive Transfer , Alveolar Bone Loss/immunology , Alveolar Bone Loss/physiopathology , Animals , Antibiotic Prophylaxis , Bacteroidaceae Infections/microbiology , Fimbriae, Bacterial/physiology , Gene Expression Regulation/immunology , Gingivitis/complications , Gingivitis/immunology , HEK293 Cells , Humans , Lymphocyte Culture Test, Mixed , Macrophage Activation , Macrophages/transplantation , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/physiology , NF-kappa B/metabolism , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/ultrastructure , Signal Transduction , Specific Pathogen-Free Organisms , Tumor Necrosis Factor-alpha/deficiency
13.
J Immunol ; 189(7): 3681-8, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22956579

ABSTRACT

Clinical and epidemiological studies have implicated chronic infections in the development of atherosclerosis. It has been proposed that common mechanisms of signaling via TLRs link stimulation by multiple pathogens to atherosclerosis. However, how pathogen-specific stimulation of TLR4 contributes to atherosclerosis progression remains poorly understood. In this study, atherosclerosis-prone apolipoprotein-E null (ApoE(-/-)) and TLR4-deficient (ApoE(-/-)TLR4(-/-)) mice were orally infected with the periodontal pathogen Porphyromonas gingivalis. ApoE(-/-)TLR4(-/-) mice were markedly more susceptible to atherosclerosis after oral infection with P. gingivalis. Using live animal imaging, we demonstrate that enhanced lesion progression occurs progressively and was increasingly evident with advancing age. Immunohistochemical analysis of lesions from ApoE(-/-)TLR4(-/-) mice revealed an increased inflammatory cell infiltrate composed primarily of macrophages and IL-17 effector T cells (Th17), a subset linked with chronic inflammation. Furthermore, enhanced atherosclerosis in TLR4-deficient mice was associated with impaired development of Th1 immunity and regulatory T cell infiltration. In vitro studies suggest that the mechanism of TLR4-mediated protective immunity may be orchestrated by dendritic cell IL-12 and IL-10, which are prototypic Th1 and regulatory T cell polarizing cytokines. We demonstrate an atheroprotective role for TLR4 in response to infection with the oral pathogen P. gingivalis. Our results point to a role for pathogen-specific TLR signaling in chronic inflammation and atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Bacteroidaceae Infections/immunology , Gingivitis/immunology , Inflammation Mediators/physiology , Porphyromonas gingivalis/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/physiology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/pathology , Disease Progression , Gingivitis/genetics , Gingivitis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Porphyromonas gingivalis/pathogenicity , Signal Transduction/genetics , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
14.
Biochemistry ; 51(32): 6312-9, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22849347

ABSTRACT

NKX2.5 is a homeodomain containing transcription factor regulating cardiac formation and function, and its mutations are linked to congenital heart disease. Here we provide the first report of the crystal structure of the NKX2.5 homeodomain in complex with double-stranded DNA of its endogenous target, locating within the proximal promoter -242 site of the atrial natriuretic factor gene. The crystal structure, determined at 1.8 Å resolution, demonstrates that NKX2.5 homeodomains occupy both DNA binding sites separated by five nucleotides without physical interaction between themselves. The two homeodomains show identical conformation despite the differences in the DNA sequences they bind, and no significant bending of the DNA was observed. Tyr54, absolutely conserved in NK2 family proteins, mediates sequence-specific interaction with the TAAG motif. This high resolution crystal structure of NKX2.5 protein provides a detailed picture of protein and DNA interactions, which allows us to predict DNA binding of mutants identified in human patients.


Subject(s)
Atrial Natriuretic Factor/genetics , DNA/chemistry , Homeodomain Proteins/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Heart Defects, Congenital/genetics , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Promoter Regions, Genetic , Protein Conformation , Thermodynamics , Transcription Factors/genetics
15.
Mol Cell Biol ; 31(22): 4633-45, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21930795

ABSTRACT

Atrial natriuretic factor (ANF) is abundantly expressed in atrial cardiomyocytes throughout ontogeny and in ventricular cardiomyocytes in the developing heart. However, during cardiac failure and hypertrophy, ANF expression can reappear in adult ventricular cardiomyocytes. The transcription factor Nkx2-5 is one of the major transactivators of the ANF gene in the developing heart. We identified Nkx2-5 binding at three 5' regulatory elements (kb -34, -31, and -21) and at the proximal ANF promoter by ChIP assay using neonatal mouse cardiomyocytes. 3C analysis revealed close proximity between the distal elements and the promoter region. A 5.8-kb fragment consisting of these elements transactivated a reporter gene in vivo recapitulating endogenous ANF expression, which was markedly reduced in tamoxifen-inducible Nkx2-5 gene knockout mice. However, expression of a reporter gene was increased and expanded toward the outer compact layer in the absence of the transcription repressor Hey2, similar to endogenous ANF expression. Functional Nkx2-5 and Hey2 binding sites separated by 59 bp were identified in the -34 kb element in neonatal cardiomyocytes. In adult hearts, this fragment did not respond to pressure overload, and ANF was induced in the absence of Nkx2-5. These results demonstrate that Nkx2-5 and its responsive cis-regulatory DNA elements are essential for ANF expression selectively in the developing heart.


Subject(s)
Atrial Natriuretic Factor/genetics , Heart Failure/metabolism , Heart/embryology , Homeodomain Proteins/metabolism , Myocardium/metabolism , Transcription Factors/metabolism , Animals , Atrial Natriuretic Factor/biosynthesis , Atrial Natriuretic Factor/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites/genetics , Cell Differentiation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Heart Failure/genetics , Heart Failure/pathology , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , Repressor Proteins/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcriptional Activation
16.
Free Radic Biol Med ; 48(9): 1182-7, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20132882

ABSTRACT

Myocardial failure is associated with increased oxidative stress and abnormal excitation-contraction coupling characterized by depletion of sarcoplasmic reticulum (SR) Ca(2+) stores and a reduction in Ca(2+)-transient amplitude. Little is known about the mechanisms whereby oxidative stress affects Ca(2+) handling and contractile function; however, reactive thiols may be involved. We used an in vitro cardiomyocyte system to test the hypothesis that short-term oxidative stress induces SR Ca(2+) depletion via redox-mediated regulation of sarcoendoplasmic reticulum Ca(2+)-ATPase (SERCA) and the sodium-Ca(2+) exchanger (NCX) and that this is associated with thiol oxidation. Adult rat ventricular myocytes paced at 5 Hz were superfused with H(2)O(2) (100 microM, 15 min). H(2)O(2) caused a progressive decrease in cell shortening followed by diastolic arrest, which was associated with decreases in SR Ca(2+) content, systolic [Ca(2+)](i), and Ca(2+)-transient amplitude, but no change in diastolic [Ca(2+)](i). H(2)O(2) caused reciprocal effects on the activities of SERCA (decreased) and NCX (increased). Pretreatment with the NCX inhibitor KB-R7943 before H(2)O(2) increased diastolic [Ca(2+)](i) and mimicked the effect of SERCA inhibition with thapsigargin. These functional effects were associated with oxidative modification of thiols on both SERCA and NCX. In conclusion, redox-mediated SR Ca(2+) depletion involves reciprocal regulation of SERCA and NCX, possibly via direct oxidative modification of both proteins.


Subject(s)
Calcium/metabolism , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Cells, Cultured , Hydrogen Peroxide/pharmacology , Male , Myocytes, Cardiac/drug effects , Oxidants/pharmacology , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum/drug effects
17.
Lab Invest ; 89(9): 983-93, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19546853

ABSTRACT

Mutations in homeoprotein NKX2-5 are linked to human congenital heart disease, resulting in various cardiac anomalies, as well as in postnatal progressive conduction defects and occasional left ventricular dysfunction; yet the function of Nkx2-5 in the postnatal period is largely unexplored. In the heart, the majority of cardiomyocytes are believed to complete cell-cycle withdrawal shortly after birth, which is generally accompanied by a re-organization of chromatin structure shown in other tissues. We reasoned that the effects of the loss of Nkx2-5 in mice may be different after cell-cycle withdrawal compared with those of the perinatal loss of Nkx2-5, which results in rapid conduction and contraction defects within 4 days after the deletion of Nkx2-5 alleles (Circ Res. 2008;103:580). In this study, floxed-Nkx2-5 alleles were deleted using tamoxifen-inducible Cre transgene (Cre-ER) beginning at 2 weeks of age. The loss of Nkx2-5 beginning at 2 weeks of age resulted in conduction and contraction defects similar to the perinatal loss of Nkx2-5, however, with a substantially slower disease progression shown by 1 degrees atrioventricular block at 6 weeks of age (4 weeks after tamoxifen injections) and heart enlargement after 12 weeks of age (10 weeks after tamoxifen injections). The phenotypes were accompanied by a slower and smaller degree of reduction of several critical Nkx2-5 downstream targets that were observed in mice with a perinatal loss of Nkx2-5. These results suggest that Nkx2-5 is necessary for proper conduction and contraction after 2 weeks of age, but with a substantially distinct level of necessity at 2 weeks of age compared with that in the perinatal period.


Subject(s)
Cardiomyopathies/physiopathology , Heart Conduction System/physiopathology , Myocardial Contraction/physiology , Myocytes, Cardiac/cytology , Transcription Factors/deficiency , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cell Differentiation/physiology , Down-Regulation , Electrocardiography , Female , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Male , Mice , Mice, Knockout , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Telemetry , Transcription Factors/genetics
18.
Biomark Med ; 3(5): 495-511, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20477519

ABSTRACT

Biomarkers aid in diagnosis by providing important information for the clinical assessment of patients that is not achieved by other means. This article focuses on the use of soluble ST2 as a biomarker in cardiovascular disease. Soluble ST2 is a secreted receptor belonging to the IL-1 receptor family that regulates inflammation and immunity. Recent studies have shown that soluble ST2 is a decoy receptor that disrupts the interaction of IL-33 with the full-length ST2L receptor. The rapidly evolving and expanding literature on ST2 and its ligand reveal emerging roles for this system in the regulation of inflammation in a variety of diseases. Elevated ST2 levels have been detected in cardiovascular diseases and levels provide useful prognostic information in many, but not all, of these diseases, which will be discussed here. Additional studies demonstrating elevated soluble ST2 levels in other human diseases will also be discussed.

19.
J Am Coll Cardiol ; 52(25): 2166-74, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19095135

ABSTRACT

OBJECTIVES: This study was designed to investigate: 1) relationships between serum ST2 levels and hemodynamic/neurohormonal variables; 2) myocardial ST2 production; and the 3) expression of ST2, membrane-anchored ST2L, and its ligand, interleukin (IL)-33, in myocardium, endothelium, and leukocytes from patients with left ventricular (LV) pressure overload and congestive cardiomyopathy. BACKGROUND: Serum levels of ST2 are elevated in heart failure. The relationship of ST2 to hemodynamic variables, source of ST2, and expression of ST2L and IL-33 in the cardiovascular system are unknown. METHODS: Serum ST2 (pg/ml; median [25th, 75th percentile]) was measured in patients with LV hypertrophy (aortic stenosis) (n = 45), congestive cardiomyopathy (n = 53), and controls (n = 23). ST2 was correlated to N-terminal pro-brain natriuretic peptide, C-reactive protein, and hemodynamic variables. Coronary sinus and arterial blood sampling determined myocardial gradient (production) of ST2. The levels of ST2, ST2L, and IL-33 were measured (reverse transcriptase-polymerase chain reaction) in myocardial biopsies and leukocytes. The ST2 protein production was evaluated in human endothelial cells. The IL-33 protein expression was determined (immunohistochemistry) in coronary artery endothelium. RESULTS: The ST2 protein was elevated in aortic stenosis (103 [65, 165] pg/ml, p < 0.05) and congestive cardiomyopathy (194 [69, 551] pg/ml, p < 0.01) versus controls (49 [4, 89] pg/ml) and correlated with B-type natriuretic peptide (r = 0.5, p < 0.05), C-reactive protein (r = 0.6, p < 0.01), and LV end-diastolic pressure (r = 0.38, p < 0.03). The LV ST2 messenger ribonucleic acid was similar in aortic stenosis and congestive cardiomyopathy versus control (p = NS). No myocardial ST2 protein gradient was observed. Endothelial cells secreted ST2. The IL-33 protein was expressed in coronary artery endothelium. Leukocyte ST2L and IL-33 levels were highly correlated (r = 0.97, p < 0.001). CONCLUSIONS: In human hypertrophy and failure, serum ST2 correlates with the diastolic load. Though the heart, endothelium, and leukocytes express components of ST2/ST2L/IL-33 pathway, the source of circulating serum ST2 is extra-myocardial.


Subject(s)
Endothelium, Vascular/metabolism , Heart Failure/metabolism , Hypertrophy, Left Ventricular/metabolism , Interleukins/metabolism , Leukocytes/metabolism , Myocardium/metabolism , Receptors, Cell Surface/metabolism , Aged , C-Reactive Protein/metabolism , Case-Control Studies , Diastole , Endothelium, Vascular/physiopathology , Female , Heart Failure/blood , Heart Failure/physiopathology , Hemodynamics , Humans , Hypertension/metabolism , Hypertrophy, Left Ventricular/blood , Hypertrophy, Left Ventricular/physiopathology , Inflammation/physiopathology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/blood , Male , Middle Aged , Natriuretic Peptide, Brain/metabolism , Prognosis , Receptors, Cell Surface/blood
20.
Circ Res ; 103(6): 580-90, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18689573

ABSTRACT

Homeobox transcription factor Nkx2-5, highly expressed in heart, is a critical factor during early embryonic cardiac development. In this study, using tamoxifen-inducible Nkx2-5 knockout mice, we demonstrate the role of Nkx2-5 in conduction and contraction in neonates within 4 days after perinatal tamoxifen injection. Conduction defect was accompanied by reduction in ventricular expression of the cardiac voltage-gated Na+ channel pore-forming alpha-subunit (Na(v)1.5-alpha), the largest ion channel in the heart responsive for rapid depolarization of the action potential, which leads to increased intracellular Ca2+ for contraction (conduction-contraction coupling). In addition, expression of ryanodine receptor 2, through which Ca2+ is released from sarcoplasmic reticulum, was substantially reduced in Nkx2-5 knockout mice. These results indicate that Nkx2-5 function is critical not only during cardiac development but also in perinatal hearts, by regulating expression of several important gene products involved in conduction and contraction.


Subject(s)
Heart Conduction System/growth & development , Myocardial Contraction/genetics , Transcription Factors/deficiency , Action Potentials/genetics , Animals , Animals, Newborn , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Chickens , Heart Conduction System/physiology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Long QT Syndrome/genetics , Long QT Syndrome/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...