Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cell ; 163(7): 1585-95, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26687352

RESUMO

Trimethylamine (TMA) N-oxide (TMAO), a gut-microbiota-dependent metabolite, both enhances atherosclerosis in animal models and is associated with cardiovascular risks in clinical studies. Here, we investigate the impact of targeted inhibition of the first step in TMAO generation, commensal microbial TMA production, on diet-induced atherosclerosis. A structural analog of choline, 3,3-dimethyl-1-butanol (DMB), is shown to non-lethally inhibit TMA formation from cultured microbes, to inhibit distinct microbial TMA lyases, and to both inhibit TMA production from physiologic polymicrobial cultures (e.g., intestinal contents, human feces) and reduce TMAO levels in mice fed a high-choline or L-carnitine diet. DMB inhibited choline diet-enhanced endogenous macrophage foam cell formation and atherosclerotic lesion development in apolipoprotein e(-/-) mice without alterations in circulating cholesterol levels. The present studies suggest that targeting gut microbial production of TMA specifically and non-lethal microbial inhibitors in general may serve as a potential therapeutic approach for the treatment of cardiometabolic diseases.


Assuntos
Aterosclerose/tratamento farmacológico , Colina/análogos & derivados , Trato Gastrointestinal/microbiologia , Hexanóis/administração & dosagem , Liases/antagonistas & inibidores , Metilaminas/metabolismo , Animais , Apolipoproteínas E/genética , Aterosclerose/metabolismo , Colesterol/metabolismo , Colina/metabolismo , Dieta , Fezes/química , Células Espumosas/metabolismo , Humanos , Liases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microbiota
2.
Int J Mol Sci ; 22(24)2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34948275

RESUMO

L-alpha glycerylphosphorylcholine (GPC), a nutritional supplement, has been demonstrated to improve neurological function. However, a new study suggests that GPC supplementation increases incident stroke risk thus its potential adverse effects warrant further investigation. Here we show that GPC promotes atherosclerosis in hyperlipidemic Apoe-/- mice. GPC can be metabolized to trimethylamine N-oxide, a pro-atherogenic agent, suggesting a potential molecular mechanism underlying the observed atherosclerosis progression. GPC supplementation shifted the gut microbial community structure, characterized by increased abundance of Parabacteroides, Ruminococcus, and Bacteroides and decreased abundance of Akkermansia, Lactobacillus, and Roseburia, as determined by 16S rRNA gene sequencing. These data are consistent with a reduction in fecal and cecal short chain fatty acids in GPC-fed mice. Additionally, we found that GPC supplementation led to an increased relative abundance of choline trimethylamine lyase (cutC)-encoding bacteria via qPCR. Interrogation of host inflammatory signaling showed that GPC supplementation increased expression of the proinflammatory effectors CXCL13 and TIMP-1 and activated NF-κB and MAPK signaling pathways in human coronary artery endothelial cells. Finally, targeted and untargeted metabolomic analysis of murine plasma revealed additional metabolites associated with GPC supplementation and atherosclerosis. In summary, our results show GPC promotes atherosclerosis through multiple mechanisms and that caution should be applied when using GPC as a nutritional supplement.


Assuntos
Aterosclerose/etiologia , Glicerilfosforilcolina/efeitos adversos , Glicerilfosforilcolina/metabolismo , Animais , Apolipoproteínas E/genética , Aterosclerose/induzido quimicamente , Aterosclerose/metabolismo , Ceco/metabolismo , Ceco/microbiologia , Linhagem Celular , Suplementos Nutricionais/efeitos adversos , Células Endoteliais/metabolismo , Ácidos Graxos Voláteis/metabolismo , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Glicerilfosforilcolina/farmacologia , Humanos , Masculino , Metilaminas/efeitos adversos , Metilaminas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo
3.
Anal Biochem ; 455: 35-40, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24704102

RESUMO

Trimethylamine-N-oxide (TMAO) levels in blood predict future risk for major adverse cardiac events including myocardial infarction, stroke, and death. Thus, the rapid determination of circulating TMAO concentration is of clinical interest. Here we report a method to measure TMAO in biological matrices by stable isotope dilution liquid chromatography tandem mass spectrometry (LC/MS/MS) with lower and upper limits of quantification of 0.05 and >200µM, respectively. Spike and recovery studies demonstrate an accuracy at low (0.5µM), mid (5µM), and high (100µM) levels of 98.2, 97.3, and 101.6%, respectively. Additional assay performance metrics include intraday and interday coefficients of variance of <6.4 and <9.9%, respectively, across the range of TMAO levels. Stability studies reveal that TMAO in plasma is stable both during storage at -80°C for 5years and to multiple freeze thaw cycles. Fasting plasma normal range studies among apparently healthy subjects (n=349) show a range of 0.73-126µM, median (interquartile range) levels of 3.45 (2.25-5.79)µM, and increasing values with age. The LC/MS/MS-based assay reported should be of value for further studies evaluating TMAO as a risk marker and for examining the effect of dietary, pharmacologic, and environmental factors on TMAO levels.


Assuntos
Cromatografia Líquida/métodos , Metilaminas/sangue , Espectrometria de Massas em Tandem/métodos , Adulto , Idoso , Deutério , Jejum , Feminino , Humanos , Técnicas de Diluição do Indicador , Limite de Detecção , Masculino , Pessoa de Meia-Idade , Valores de Referência , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização por Electrospray/métodos
4.
Sci Transl Med ; 15(678): eabn8134, 2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36630484

RESUMO

The antibiotic-resistant bacterium Acinetobacter baumannii is a leading cause of hospital-associated infections. Despite surveillance and infection control efforts, new A. baumannii strains are regularly isolated from health care facilities worldwide. In a mouse model of urinary tract infection, we found that mice infected with A. baumannii displayed high bacterial burdens in urine for several weeks. Two months after the resolution of A. baumannii infection, inserting a catheter into the bladder of mice with resolved infection led to the resurgence of a same-strain urinary tract infection in ~53% of the mice within 24 hours. We identified intracellular A. baumannii bacteria in the bladder epithelial cells of mice with resolved infection, which we propose could act as a host reservoir that was activated upon insertion of a catheter, leading to a resurgent secondary infection.


Assuntos
Acinetobacter baumannii , Infecção Hospitalar , Infecções Urinárias , Animais , Camundongos , Bexiga Urinária , Cateterismo , Infecções Urinárias/microbiologia , Antibacterianos/farmacologia , Infecção Hospitalar/microbiologia , Farmacorresistência Bacteriana Múltipla
5.
Sci Rep ; 9(1): 11128, 2019 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-31366948

RESUMO

Miltefosine is an FDA approved oral drug for treating cutaneous and visceral leishmaniasis. Leishmania is a flagellated protozoa, which infects and differentiates in macrophages. Here, we studied the effects of Miltefosine on macrophage's lipid homeostasis, autophagy, and NLRP3 inflammasome assembly/activity. Miltefosine treatment conferred multiple effects on macrophage lipid homeostasis leading to increased cholesterol release from cells, increased lipid-raft disruption, decreased phosphatidylserine (PS) flip from the cell-surface, and redistribution of phosphatidylinositol 4,5-bisphosphate (PIP2) from the plasma membrane to actin rich regions in the cells. Enhanced basal autophagy, lipophagy and mitophagy was observed in cells treated with Miltefosine vs. control. Miltefosine treated cells showed marked increased in phosphorylation of kinases involved in autophagy induction such as; Adenosine monophosphate-activated protein kinase (AMPK) and Unc-51 like autophagy activating kinase (ULK1). The Toll like receptor (TLR) signaling pathway was blunted by Miltefosine treatment, resulting in decreased TLR4 recruitment to cell-surface and ~75% reduction in LPS induced pro-IL-1ß mRNA levels. Miltefosine reduced endotoxin-mediated mitochondrial reactive oxygen species and protected the mitochondrial membrane potential. Miltefosine treatment induced mitophagy and dampened NLRP3 inflammasome assembly. Collectively, our data shows that Miltefosine induced ABCA1 mediated cholesterol release, induced AMPK phosphorylation and mitophagy, while dampening NLRP3 inflammasome assembly and IL-1ß release.


Assuntos
Colesterol/metabolismo , Inflamassomos/efeitos dos fármacos , Interleucina-1beta/metabolismo , Macrófagos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fosforilcolina/análogos & derivados , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular , Células HEK293 , Humanos , Inflamassomos/metabolismo , Macrófagos/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilcolina/farmacologia , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Elife ; 72018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29345620

RESUMO

Uropathogenic E. coli (UPEC), which cause urinary tract infections (UTI), utilize type 1 pili, a chaperone usher pathway (CUP) pilus, to cause UTI and colonize the gut. The pilus rod, comprised of repeating FimA subunits, provides a structural scaffold for displaying the tip adhesin, FimH. We solved the 4.2 Å resolution structure of the type 1 pilus rod using cryo-electron microscopy. Residues forming the interactive surfaces that determine the mechanical properties of the rod were maintained by selection based on a global alignment of fimA sequences. We identified mutations that did not alter pilus production in vitro but reduced the force required to unwind the rod. UPEC expressing these mutant pili were significantly attenuated in bladder infection and intestinal colonization in mice. This study elucidates an unappreciated functional role for the molecular spring-like property of type 1 pilus rods in host-pathogen interactions and carries important implications for other pilus-mediated diseases.


Assuntos
Adesinas Bacterianas/metabolismo , Adesinas Bacterianas/ultraestrutura , Aderência Bacteriana , Infecções por Escherichia coli/microbiologia , Fímbrias Bacterianas/metabolismo , Interações Hospedeiro-Patógeno , Escherichia coli Uropatogênica/ultraestrutura , Animais , Microscopia Crioeletrônica , Modelos Animais de Doenças , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/ultraestrutura , Camundongos , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica/fisiologia
7.
J Am Coll Cardiol ; 64(18): 1908-14, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25444145

RESUMO

BACKGROUND: Altered intestinal function is prevalent in patients with heart failure (HF), but its role in adverse outcomes is unclear. OBJECTIVES: This study investigated the potential pathophysiological contributions of intestinal microbiota in HF. METHODS: We examined the relationship between fasting plasma trimethylamine-N-oxide (TMAO) and all-cause mortality over a 5-year follow-up in 720 patients with stable HF. RESULTS: The median TMAO level was 5.0 µM, which was higher than in subjects without HF (3.5 µM; p < 0.001). There was modest but significant correlation between TMAO concentrations and B-type natriuretic peptide (BNP) levels (r = 0.23; p < 0.001). Higher plasma TMAO levels were associated with a 3.4-fold increased mortality risk. Following adjustments for traditional risk factors and BNP levels, elevated TMAO levels remained predictive of 5-year mortality risk (hazard ratio [HR]: 2.2; 95% CI: 1.42 to 3.43; p < 0.001), as well as following the addition of estimated glomerular filtration rate to the model (HR: 1.75; 95% CI: 1.07 to 2.86; p < 0.001). CONCLUSIONS: High TMAO levels were observed in patients with HF, and elevated TMAO levels portended higher long-term mortality risk independent of traditional risk factors and cardiorenal indexes.


Assuntos
Insuficiência Cardíaca/mortalidade , Mucosa Intestinal/metabolismo , Metilaminas/sangue , Microbiota , Medição de Risco/métodos , Idoso , Biomarcadores/sangue , Feminino , Seguimentos , Insuficiência Cardíaca/sangue , Humanos , Intestinos/microbiologia , Estimativa de Kaplan-Meier , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Ohio/epidemiologia , Prognóstico , Estudos Prospectivos , Taxa de Sobrevida/tendências , Fatores de Tempo
8.
Nat Med ; 20(2): 193-203, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24464187

RESUMO

Recent studies have indicated that high-density lipoproteins (HDLs) and their major structural protein, apolipoprotein A1 (apoA1), recovered from human atheroma are dysfunctional and are extensively oxidized by myeloperoxidase (MPO). In vitro oxidation of either apoA1 or HDL particles by MPO impairs their cholesterol acceptor function. Here, using phage display affinity maturation, we developed a high-affinity monoclonal antibody that specifically recognizes both apoA1 and HDL that have been modified by the MPO-H2O2-Cl(-) system. An oxindolyl alanine (2-OH-Trp) moiety at Trp72 of apoA1 is the immunogenic epitope. Mutagenesis studies confirmed a critical role for apoA1 Trp72 in MPO-mediated inhibition of the ATP-binding cassette transporter A1 (ABCA1)-dependent cholesterol acceptor activity of apoA1 in vitro and in vivo. ApoA1 containing a 2-OH-Trp72 group (oxTrp72-apoA1) is in low abundance within the circulation but accounts for 20% of the apoA1 in atherosclerosis-laden arteries. OxTrp72-apoA1 recovered from human atheroma or plasma is lipid poor, virtually devoid of cholesterol acceptor activity and demonstrated both a potent proinflammatory activity on endothelial cells and an impaired HDL biogenesis activity in vivo. Elevated oxTrp72-apoA1 levels in subjects presenting to a cardiology clinic (n = 627) were associated with increased cardiovascular disease risk. Circulating oxTrp72-apoA1 levels may serve as a way to monitor a proatherogenic process in the artery wall.


Assuntos
Apolipoproteína A-I/metabolismo , Doenças Cardiovasculares/genética , Lipoproteínas HDL/metabolismo , Peroxidase/metabolismo , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Alanina/análogos & derivados , Alanina/genética , Anticorpos Monoclonais , Apolipoproteína A-I/genética , Apolipoproteína A-I/imunologia , Técnicas de Visualização da Superfície Celular , Cromatografia Líquida , Ensaio de Imunoadsorção Enzimática , Epitopos/genética , Imunofluorescência , Humanos , Imuno-Histoquímica , Lipoproteínas HDL/imunologia , Mutagênese , Razão de Chances , Oxirredução , Oxindóis , Espectrometria de Massas em Tandem , Molécula 1 de Adesão de Célula Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA