Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Anaerobe ; 71: 102399, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34090994

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Boca/microbiologia , Doenças Periodontais/microbiologia , Porphyromonas gingivalis/fisiologia , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Bactérias/isolamento & purificação , Disbiose/microbiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota , Filogenia , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/isolamento & purificação
2.
Anaerobe ; 45: 19-30, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28442421

RESUMO

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.


Assuntos
Aterosclerose/fisiopatologia , Infecções Bacterianas/complicações , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Metabolismo dos Lipídeos , Porphyromonas gingivalis/patogenicidade , Animais , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL
3.
PLoS Pathog ; 10(7): e1004215, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25010102

RESUMO

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.


Assuntos
Infecções por Bacteroidaceae/imunologia , Lipídeo A/imunologia , Porphyromonas gingivalis/imunologia , Vasculite/imunologia , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/imunologia , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/microbiologia , Aterosclerose/patologia , Infecções por Bacteroidaceae/genética , Infecções por Bacteroidaceae/microbiologia , Infecções por Bacteroidaceae/patologia , Modelos Animais de Doenças , Células HEK293 , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Osteoporose/genética , Osteoporose/imunologia , Osteoporose/microbiologia , Osteoporose/patologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Vasculite/genética , Vasculite/microbiologia , Vasculite/patologia
4.
Arterioscler Thromb Vasc Biol ; 34(3): 552-64, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24458711

RESUMO

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.


Assuntos
Inflamação/patologia , Interleucina-1beta/fisiologia , Megacariócitos/citologia , Obesidade/sangue , Ativação Plaquetária/fisiologia , Receptores Tipo I de Interleucina-1/fisiologia , Transcrição Gênica/fisiologia , Animais , Aterosclerose/etiologia , Infecções por Bacteroidaceae/sangue , Infecções por Bacteroidaceae/patologia , Linhagem Celular , Colágeno/farmacologia , Gorduras na Dieta/toxicidade , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Imidazóis/farmacologia , Inflamação/etiologia , Inflamação/genética , Interleucina-1beta/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Obesidade/complicações , Obesidade/genética , Fosforilação/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Adesividade Plaquetária/fisiologia , Porphyromonas gingivalis , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/farmacologia , Receptores Tipo I de Interleucina-1/deficiência , Receptores Tipo I de Interleucina-1/genética , Trombina/farmacologia , Transcrição Gênica/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
J Immunol ; 190(3): 1148-57, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23264656

RESUMO

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.


Assuntos
Perda do Osso Alveolar/etiologia , Infecções por Bacteroidaceae/imunologia , Gengivite/fisiopatologia , Macrófagos/fisiologia , Porphyromonas gingivalis/patogenicidade , Receptor 2 Toll-Like/fisiologia , Receptor 4 Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Transferência Adotiva , Perda do Osso Alveolar/imunologia , Perda do Osso Alveolar/fisiopatologia , Animais , Antibioticoprofilaxia , Infecções por Bacteroidaceae/microbiologia , Fímbrias Bacterianas/fisiologia , Regulação da Expressão Gênica/imunologia , Gengivite/complicações , Gengivite/imunologia , Células HEK293 , Humanos , Teste de Cultura Mista de Linfócitos , Ativação de Macrófagos , Macrófagos/transplante , Macrófagos Peritoneais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/fisiologia , NF-kappa B/metabolismo , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/imunologia , Porphyromonas gingivalis/ultraestrutura , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Fator de Necrose Tumoral alfa/deficiência
6.
BMC Genomics ; 15: 1176, 2014 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-25540039

RESUMO

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.


Assuntos
Aorta/metabolismo , Apolipoproteínas E/deficiência , Chlamydophila pneumoniae/fisiologia , Dieta Ocidental/efeitos adversos , Perfilação da Expressão Gênica , Porphyromonas gingivalis/fisiologia , Animais , Aorta/patologia , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Família Multigênica/genética , Placa Aterosclerótica/etiologia , Placa Aterosclerótica/genética , Placa Aterosclerótica/microbiologia , Placa Aterosclerótica/patologia
7.
J Immunol ; 189(7): 3681-8, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22956579

RESUMO

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.


Assuntos
Aterosclerose/imunologia , Infecções por Bacteroidaceae/imunologia , Gengivite/imunologia , Mediadores da Inflamação/fisiologia , Porphyromonas gingivalis/imunologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/fisiologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Infecções por Bacteroidaceae/genética , Infecções por Bacteroidaceae/patologia , Progressão da Doença , Gengivite/genética , Gengivite/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Porphyromonas gingivalis/patogenicidade , Transdução de Sinais/genética , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética
8.
Biochemistry ; 51(32): 6312-9, 2012 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-22849347

RESUMO

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.


Assuntos
Fator Natriurético Atrial/genética , DNA/química , Proteínas de Homeodomínio/química , Fatores de Transcrição/química , Sequência de Aminoácidos , Cristalografia por Raios X , Cardiopatias Congênitas/genética , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Regiões Promotoras Genéticas , Conformação Proteica , Termodinâmica , Fatores de Transcrição/genética
9.
Circ Res ; 102(5): 571-80, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18202317

RESUMO

Two myosin light chain (MLC) kinase (MLCK) proteins, smooth muscle (encoded by mylk1 gene) and skeletal (encoded by mylk2 gene) MLCK, have been shown to be expressed in mammals. Even though phosphorylation of its putative substrate, MLC2, is recognized as a key regulator of cardiac contraction, a MLCK that is preferentially expressed in cardiac muscle has not yet been identified. In this study, we characterized a new kinase encoded by a gene homologous to mylk1 and -2, named cardiac MLCK, which is specifically expressed in the heart in both atrium and ventricle. In fact, expression of cardiac MLCK is highly regulated by the cardiac homeobox protein Nkx2-5 in neonatal cardiomyocytes. The overall structure of cardiac MLCK protein is conserved with skeletal and smooth muscle MLCK; however, the amino terminus is quite unique, without significant homology to other known proteins, and its catalytic activity does not appear to be regulated by Ca(2+)/calmodulin in vitro. Cardiac MLCK is phosphorylated and the level of phosphorylation is increased by phenylephrine stimulation accompanied by increased level of MLC2v phosphorylation. Both overexpression and knockdown of cardiac MLCK in cultured cardiomyocytes revealed that cardiac MLCK is likely a new regulator of MLC2 phosphorylation, sarcomere organization, and cardiomyocyte contraction.


Assuntos
Miosinas Cardíacas/metabolismo , Miócitos Cardíacos/metabolismo , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/biossíntese , Animais , Animais Recém-Nascidos , Células Cultivadas , Clonagem Molecular , Sequência Conservada/genética , Átrios do Coração/enzimologia , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/enzimologia , Camundongos , Dados de Sequência Molecular , Contração Miocárdica , Infarto do Miocárdio/complicações , Miócitos Cardíacos/citologia , Quinase de Cadeia Leve de Miosina/genética , Especificidade de Órgãos , Fosforilação , Ratos , Sarcômeros/metabolismo
10.
Circ Res ; 103(6): 580-90, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18689573

RESUMO

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.


Assuntos
Sistema de Condução Cardíaco/crescimento & desenvolvimento , Contração Miocárdica/genética , Fatores de Transcrição/deficiência , Potenciais de Ação/genética , Animais , Animais Recém-Nascidos , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Galinhas , Sistema de Condução Cardíaco/fisiologia , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fatores de Transcrição/genética
11.
Lab Invest ; 89(9): 983-93, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19546853

RESUMO

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.


Assuntos
Cardiomiopatias/fisiopatologia , Sistema de Condução Cardíaco/fisiopatologia , Contração Miocárdica/fisiologia , Miócitos Cardíacos/citologia , Fatores de Transcrição/deficiência , Animais , Cardiomegalia/genética , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Cardiomiopatias/genética , Cardiomiopatias/patologia , Diferenciação Celular/fisiologia , Regulação para Baixo , Eletrocardiografia , Feminino , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Telemetria , Fatores de Transcrição/genética
12.
PLoS One ; 14(1): e0210827, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30682073

RESUMO

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.


Assuntos
Glutarredoxinas/metabolismo , Interleucina-33/biossíntese , Interleucina-33/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Alérgenos/administração & dosagem , Animais , Asma/etiologia , Asma/imunologia , Asma/metabolismo , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Glutarredoxinas/deficiência , Glutarredoxinas/genética , Glutationa/metabolismo , Lipopolissacarídeos/farmacologia , Pulmão/imunologia , Pulmão/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Células RAW 264.7 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/metabolismo
14.
J Asthma Allergy ; 11: 159-171, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30122959

RESUMO

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.

15.
Int J Oral Sci ; 10(4): 32, 2018 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-30420594

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas/microbiologia , Metagenoma , Microbiota , Neoplasias Bucais/microbiologia , Transcriptoma , Fatores de Virulência/metabolismo , Humanos , Filogenia , Projetos Piloto , RNA Mensageiro/metabolismo , Virulência
16.
Physiol Genomics ; 27(1): 86-94, 2006 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-16835353

RESUMO

The pathogenesis of cardiac left ventricular hypertrophy and failure is poorly defined due to the complexity of the disease phenotype. To gain a better understanding of the relationship between gene expression and left ventricular hypertrophy, we employed a quantitative approach to identify genes with expression patterns that correlate in a numerically continuous manner with parameters of cardiac structure and function in a mouse model of left ventricular hypertrophy due to transverse aortic constriction. Several genes showed expression patterns that were significantly correlated (Pearson's correlation coefficient) with measurements of left ventricular weight, left ventricular wall thickness, and diastolic dimension. We validated our findings in two independent data sets and in a small subset of genes by real-time RT-PCR. Of genes with significant correlations to numerically continuous measurements of hypertrophy, we found enrichment for genes encoding extracellular matrix, growth-related and secreted proteins in the directly correlated subset, and for genes encoding mitochondria and metabolic/fatty acid oxidation proteins in the inversely correlated subset. The results of this filtering strategy suggest that this subset of transcripts with quantitative relationships between gene expression and left ventricular hypertrophy represents potentially important pathways that contribute to the progression to heart failure and are thus candidates for follow-up and functional analysis.


Assuntos
Expressão Gênica , Hipertrofia Ventricular Esquerda/genética , Animais , Bases de Dados Genéticas , Feminino , Perfilação da Expressão Gênica , Genômica , Ventrículos do Coração/metabolismo , Hipertrofia Ventricular Esquerda/diagnóstico por imagem , Hipertrofia Ventricular Esquerda/metabolismo , Masculino , Metanálise como Assunto , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Ultrassonografia
17.
Circulation ; 107(5): 721-6, 2003 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-12578875

RESUMO

BACKGROUND: Using genomic technology, we previously identified an interleukin-1 receptor family member, ST2, as a gene markedly induced by mechanical strain in cardiac myocytes. The soluble receptor form of ST2 is secreted and detectable in human serum. This study tested the hypothesis that soluble ST2 levels in the serum of patients with severe chronic heart failure are increased in patients with neurohormonal activation. METHODS AND RESULTS: Serum samples, clinical variables, and neurohormone levels from the PRAISE-2 heart failure trial (NYHA functional class III-IV; end point, mortality or transplantation) were analyzed. ST2 serum measurements were performed with ELISA on samples from 161 patients obtained at trial enrollment and from 139 of the same patients obtained 2 weeks after trial enrollment. Baseline ST2 levels were correlated with baseline B-type natriuretic peptide (BNP) levels (r=0.36, P<0.0001), baseline proatrial natriuretic peptide (ProANP) levels (r=0.36, P<0.0001), and baseline norepinephrine levels (r=0.39, P<0.0001). The change in ST2 was significant as a univariate predictor of subsequent mortality or transplantation (P=0.048), as was baseline BNP (P<0.0001) and baseline ProANP (P<0.0001). In multivariate models including BNP and ProANP, the change in ST2 remained significant as a predictor of mortality or transplantation independent of BNP and ProANP. CONCLUSIONS: Serum soluble ST2 is a novel biomarker for neurohormonal activation in patients with heart failure. In patients with severe chronic NYHA class III to IV heart failure, the change in ST2 levels is an independent predictor of subsequent mortality or transplantation.


Assuntos
Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/diagnóstico , Proteínas de Membrana/sangue , Adolescente , Adulto , Idoso , Anlodipino/uso terapêutico , Fator Natriurético Atrial/sangue , Biomarcadores/sangue , Bloqueadores dos Canais de Cálcio/uso terapêutico , Criança , Pré-Escolar , Método Duplo-Cego , Feminino , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1 , Masculino , Pessoa de Meia-Idade , Modelos Estatísticos , Análise Multivariada , Peptídeo Natriurético Encefálico , Neurotransmissores/sangue , Estresse Oxidativo , Valor Preditivo dos Testes , Estudos Prospectivos , Grupos Raciais , Receptores de Superfície Celular , Fatores Sexuais
18.
Circulation ; 109(18): 2186-90, 2004 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-15117853

RESUMO

BACKGROUND: Mechanically overloaded cardiomyocytes secrete a soluble interleukin-1 receptor family member called ST2. Serum levels of ST2 are associated with prognosis in nonischemic heart failure, but the predictive value of ST2 in patients with acute myocardial infarction is unknown. METHODS AND RESULTS: ST2 levels were measured in serum from 810 patients with acute myocardial infarction in the Thrombolysis In Myocardial Infarction (TIMI) 14 (362 patients) and Enoxaparin and TNK-tPA With or Without GPIIb/IIIa Inhibitor as Reperfusion Strategy in STEMI (ENTIRE)-TIMI 23 (448 patients) clinical trials. Baseline levels of ST2 were significantly higher in those patients who died (0.379 versus 0.233 ng/mL, P=0.0001) or developed new congestive heart failure (0.287 versus 0.233 ng/mL, P=0.009) by 30 days. In an analysis of outcomes at 30 days by ST2 quartiles, both death (P=0.001) and the combined death/heart failure end point (P=0.001) showed a significant graded association with levels of ST2; furthermore, in-hospital death (P=0.003) and death/heart failure (P=0.004) were also significantly associated with higher ST2 levels. In a logistic regression analysis that controlled for important clinical factors, increasing levels of ST2 remained associated with death at 30 days (P=0.047). ST2 levels rose during the first day after infarction and were maximal at 12 hours; ST2 levels at 12 hours were also independently associated with death at 30 days (P<0.001). CONCLUSIONS: Serum levels of the interleukin-1 receptor family member ST2 predict mortality and heart failure in patients with acute myocardial infarction. These data suggest that ST2 may be a useful biomarker and that this novel inflammatory receptor may play a role in cardiac pathophysiology.


Assuntos
Insuficiência Cardíaca/epidemiologia , Proteínas de Membrana/sangue , Infarto do Miocárdio/sangue , Abciximab , Idoso , Anticorpos Monoclonais/uso terapêutico , Anticoagulantes/uso terapêutico , Ensaios Clínicos como Assunto , Comorbidade , Quimioterapia Combinada , Enoxaparina/uso terapêutico , Feminino , Fibrinolíticos/uso terapêutico , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/etiologia , Mortalidade Hospitalar , Humanos , Fragmentos Fab das Imunoglobulinas/uso terapêutico , Incidência , Proteína 1 Semelhante a Receptor de Interleucina-1 , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/complicações , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/mortalidade , Inibidores da Agregação Plaquetária/uso terapêutico , Prognóstico , Receptores de Superfície Celular , Fatores de Risco , Análise de Sobrevida , Terapia Trombolítica , Resultado do Tratamento
19.
Circulation ; 106(23): 2961-6, 2002 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-12460879

RESUMO

BACKGROUND: We identified an interleukin-1 receptor family member, ST2, as a gene markedly induced by mechanical strain in cardiac myocytes and hypothesized that ST2 participates in the acute myocardial response to stress and injury. METHODS AND RESULTS: ST2 mRNA was induced in cardiac myocytes by mechanical strain (4.7+/-0.9-fold) and interleukin-1beta (2.0+/-0.2-fold). Promoter analysis revealed that the proximal and not the distal promoter of ST2 is responsible for transcriptional activation in cardiac myocytes by strain and interleukin-1beta. In mice subjected to coronary artery ligation, serum ST2 was transiently increased compared with unoperated controls (20.8+/-4.4 versus 0.8+/-0.8 ng/mL, P<0.05). Soluble ST2 levels were increased in the serum of human patients (N=69) 1 day after myocardial infarction and correlated positively with creatine kinase (r=0.41, P<0.001) and negatively with ejection fraction (P=0.02). CONCLUSIONS: These data identify ST2 release in response to myocardial infarction and suggest a role for this innate immune receptor in myocardial injury.


Assuntos
Proteínas de Membrana/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Receptores de Interleucina-1/metabolismo , Angiotensina II/farmacologia , Antagonistas de Receptores de Angiotensina , Animais , Animais Recém-Nascidos , Biomarcadores/sangue , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Imidazóis/farmacologia , Interleucina-1/farmacologia , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-4/farmacologia , Lipopolissacarídeos/farmacologia , Proteínas de Membrana/sangue , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/sangue , Miocárdio/citologia , Ensaios de Proteção de Nucleases , Ésteres de Forbol/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/fisiologia , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina , Receptores de Superfície Celular , Receptores de Interleucina , Receptores de Interleucina-1/sangue , Receptores de Interleucina-1/genética , Estresse Mecânico , Volume Sistólico
20.
PLoS One ; 10(7): e0131688, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26148065

RESUMO

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.


Assuntos
Plaquetas/patologia , Dieta Ocidental/efeitos adversos , Inflamação/patologia , Agregação Plaquetária/fisiologia , Animais , Apolipoproteínas E/metabolismo , Aterosclerose/metabolismo , Aterosclerose/patologia , Plaquetas/metabolismo , Plaquetas/microbiologia , Chlamydophila pneumoniae/patogenicidade , Modelos Animais de Doenças , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Masculino , Camundongos , Neutrófilos/microbiologia , Neutrófilos/patologia , Neutrófilos/fisiologia , Porphyromonas gingivalis/patogenicidade , Trombose/metabolismo , Trombose/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA