Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.286
Filtrar
1.
Nat Commun ; 15(1): 2526, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38514666

RESUMO

ß-Cell dysfunction and ß-cell loss are hallmarks of type 2 diabetes (T2D). Here, we found that trimethylamine N-oxide (TMAO) at a similar concentration to that found in diabetes could directly decrease glucose-stimulated insulin secretion (GSIS) in MIN6 cells and primary islets from mice or humans. Elevation of TMAO levels impairs GSIS, ß-cell proportion, and glucose tolerance in male C57BL/6 J mice. TMAO inhibits calcium transients through NLRP3 inflammasome-related cytokines and induced Serca2 loss, and a Serca2 agonist reversed the effect of TMAO on ß-cell function in vitro and in vivo. Additionally, long-term TMAO exposure promotes ß-cell ER stress, dedifferentiation, and apoptosis and inhibits ß-cell transcriptional identity. Inhibition of TMAO production improves ß-cell GSIS, ß-cell proportion, and glucose tolerance in both male db/db and choline diet-fed mice. These observations identify a role for TMAO in ß-cell dysfunction and maintenance, and inhibition of TMAO could be an approach for the treatment of T2D.


Assuntos
Diabetes Mellitus Tipo 2 , Humanos , Masculino , Animais , Camundongos , Camundongos Endogâmicos C57BL , Glucose/farmacologia , Metilaminas/farmacologia , Transdução de Sinais , Insulina/farmacologia
2.
Int Immunopharmacol ; 130: 111778, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38432147

RESUMO

OBJECTIVE: To investigate the mechanism of action of fatty acid receptors, FFAR1 and FFAR4, on ulcerative colitis (UC) through fatty acid metabolism and macrophage polarization. METHODS: Dextran sulfate sodium (DSS)-induced mouse model of UC mice was used to evaluate the efficacy of FFAR1 (GW9508) and FFAR4 (GSK137647) agonists by analyzing body weight, colon length, disease activity index (DAI), and histological scores. Real-time PCR and immunofluorescence analysis were performed to quantify the levels of fatty acid metabolizing enzymes and macrophage makers. FFA-induced lipid accumulation in RAW264.7 cells was visualized by Oil Red O staining analysis, and cells were collected to detect macrophage polarization by flow cytometry. RESULTS: The combination of GW9508 and GSK137647 significantly improved DSS-induced UC symptoms, caused recovery in colon length, and decreased histological injury. GW9508 + GSK137647 treatment upregulated the expressions of CD206, lipid oxidation enzyme (CPT-1α) and anti-inflammatory cytokines (IL-4, IL-10, IL-13) but downregulated those of CD86, lipogenic enzymes (ACC1, FASN, SCD1), and pro-inflammatory cytokines (IL-1ß, IL-6, TNF-α). Combining the two agonists decreased FFA-induced lipid accumulation and increased CD206 expression in cell-based experiments. CONCLUSION: Activated FFAR1 and FFAR4 ameliorates DSS-induced UC by promoting fatty acid metabolism to reduce lipid accumulation and mediate M2 macrophage polarization.


Assuntos
Colite Ulcerativa , Ácidos Graxos não Esterificados , Macrófagos , Receptores Acoplados a Proteínas G , Animais , Camundongos , Compostos de Anilina/farmacologia , Compostos de Anilina/uso terapêutico , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/tratamento farmacológico , Colo/patologia , Citocinas/metabolismo , Sulfato de Dextrana , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Metilaminas/farmacologia , Metilaminas/uso terapêutico , Camundongos Endogâmicos C57BL , Propionatos/farmacologia , Propionatos/uso terapêutico , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Receptores Acoplados a Proteínas G/agonistas
3.
Molecules ; 29(6)2024 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-38542959

RESUMO

Previous studies have revealed the microbial metabolism of dietary choline in the gut, leading to its conversion into trimethylamine (TMA). Polymethoxyflavones (PMFs), exemplified by tangeretin, have shown efficacy in mitigating choline-induced cardiovascular inflammation. However, the specific mechanism by which these compounds exert their effects, particularly in modulating the gut microbiota, remains uncertain. This investigation focused on tangeretin, a representative PMFs, to explore its influence on the gut microbiota and the choline-TMA conversion process. Experimental results showed that tangeretin treatment significantly attenuated the population of CutC-active bacteria, particularly Clostridiaceae and Lactobacillus, induced by choline chloride in rat models. This inhibition led to a decreased efficiency in choline conversion to TMA, thereby ameliorating cardiovascular inflammation resulting from prolonged choline consumption. In conclusion, tangeretin's preventive effect against cardiovascular inflammation is intricately linked to its targeted modulation of TMA-producing bacterial activity.


Assuntos
Arterite , Flavonas , Microbioma Gastrointestinal , Ratos , Animais , Colina/metabolismo , Metilaminas/farmacologia , Metilaminas/metabolismo , Bactérias/metabolismo , Inflamação/tratamento farmacológico
4.
Biochem Biophys Res Commun ; 703: 149667, 2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38382362

RESUMO

Trimethylamine N-oxide (TMAO) is a novel risk factor for atherosclerosis, and its underlying regulatory mechanisms are under intensive investigation. Inflammation-related vascular endothelial damage is the major driver in atherogenic process. Pyroptosis, a type of proinflammatory programmed cell death, has been proved to promote the initiation and progression of atherosclerosis. In our study, we found that TMAO triggered endothelial cells excessive mitophagy, thereby facilitating pyroptosis. This process is mediated by the upexpression of phosphatidylethanolamine acyltransferase (LPEAT). These findings provide insights into TMAO-induced vascular endothelial cell damage and suggest that LPEAT may be a valuable target for the prevention and treatment of atherosclerosis.


Assuntos
Aterosclerose , Células Endoteliais , Humanos , Células Endoteliais/metabolismo , Piroptose , Mitofagia , Metilaminas/farmacologia , Metilaminas/metabolismo , Aterosclerose/metabolismo
5.
Nanoscale ; 16(5): 2540-2551, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38214221

RESUMO

Osmolytes are well known to protect the protein structure against different chemical and physical denaturants. Since their actions with protein surfaces are mechanistically complicated and context dependent, the underlying molecular mechanism is not fully understood. Here, we combined single-molecule magnetic tweezers and molecular dynamics (MD) simulation to explore the mechanical role of osmolytes from two different classes, trimethylamine N-oxide (TMAO) and trehalose, as mechanical stabilizers of protein structure. We observed that these osmolytes increase the protein L mechanical stability by decreasing unfolding kinetics while accelerating the refolding kinetics under force, eventually shifting the energy landscape toward the folded state. These osmolytes mechanically stabilize the protein L and plausibly guide them to more thermodynamically robust states. Finally, we observed that osmolyte-modulated protein folding increases mechanical work output up to twofold, allowing the protein to fold under a higher force regime and providing a significant implication for folding-induced structural stability in proteins.


Assuntos
Dobramento de Proteína , Proteínas , Proteínas/química , Simulação de Dinâmica Molecular , Estabilidade Proteica , Metilaminas/química , Metilaminas/farmacologia , Termodinâmica
6.
Int J Mol Sci ; 24(24)2023 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-38139384

RESUMO

In this study, the plausible role of trimethylamine N-oxide (TMAO), a microbiota metabolite, was investigated as a link between peripheral inflammation and the inflammation of the central nervous system using different cell lines. TMAO treatment favored the differentiation of adipocytes from preadipocytes (3T3-L1 cell line). In macrophages (RAW 264.7 cell line), which infiltrate adipose tissue in obesity, TMAO increased the expression of pro-inflammatory cytokines. The treatment with 200 µM of TMAO seemed to disrupt the blood-brain barrier as it induced a significant decrease in the expression of occludin in hCMECs. TMAO also increased the expression of pro-inflammatory cytokines in primary neuronal cultures, induced a pro-inflammatory state in primary microglial cultures, and promoted phagocytosis. Data obtained from this project suggest that microbial dysbiosis and increased TMAO secretion could be a key link between peripheral and central inflammation. Thus, TMAO-decreasing compounds may be a promising therapeutic strategy for neurodegenerative diseases.


Assuntos
Inflamação , Metilaminas , Humanos , Inflamação/metabolismo , Metilaminas/farmacologia , Metilaminas/metabolismo , Citocinas , Projetos de Pesquisa
7.
Biochem Biophys Res Commun ; 669: 134-142, 2023 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-37271025

RESUMO

Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide but still lacks specific treatment modalities. The gut microbiota and its metabolites have been shown to be intimately involved in NAFLD development, participating in and regulating disease progression. Trimethylamine N-oxide (TMAO), a metabolite highly dependent on the gut microbiota, has been shown to play deleterious regulatory roles in cardiovascular disease, but the relationship between it and NAFLD lacks validation from basic experiments. This research applied TMAO intervention by constructing fatty liver cell models in vitro to observe its effect on fatty liver cells and potential key genes and performed siRNA interference on the gene to verify the action. The results showed that TMAO intervention promoted the appearance of more red-stained lipid droplets in Oil-red O staining results, increased triglyceride (TG) levels and increased mRNA levels of liver fibrosis-related genes, and also identified one of the key genes, keratin17 (KRT17) via transcriptomics. Following the reduction in its expression level, under the same treatment, there were decreased red-stained lipid droplets, decreased TG levels, decreased indicators of impaired liver function as well as decreased mRNA levels of liver fibrosis-related genes. In conclusion, the gut microbiota metabolite TMAO could promote lipid deposition and fibrosis process via the KRT17 gene in fatty liver cells in vitro.


Assuntos
Microbioma Gastrointestinal , Hepatopatia Gordurosa não Alcoólica , Humanos , Fibrose , Metilaminas/farmacologia , Metilaminas/metabolismo , Cirrose Hepática , Lipídeos
8.
J Obstet Gynaecol Res ; 49(7): 1736-1742, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37045561

RESUMO

BACKGROUND: The aim of this study is to investigate the effect of trimethylamine (TMA) and trimethylamine-n-oxide (TMAO) on the contractility of human umbilical artery and the possible mechanisms involved. METHODS: Vasoactive responses to TMA and TMAO on human umbilical artery rings were measured in isolated organ baths. Cumulative dose-response curves for TMA and TMAO were obtained before and after incubation with atropine, yohimbine, prazosin, indomethacin, verapamil, and Ca+2 -free Krebs-Henselite solution. RESULTS: Administration of cumulative TMA and TMAO resulted in dose-dependent contraction at concentrations ranging from 10 to 100 mM on human umbilical artery rings. TMA-induced contractions were more potent than TMAO-induced contractions (TMA: -logEC50 = 1.00 ± 0.02, TMAO: -logEC50 = 0.57 ± 0.02). Contraction responses to TMA were significantly lower in the presence of verapamil and in the absence of external Ca+2 (p < 0.001, p < 0.05, respectively). CONCLUSION: Our results showed that TMA and TMAO caused vasoconstriction in isolated human umbilical artery rings. Our findings also indicated that TMA but not TMAO-induced vasoconstriction was partially dependent on extracellular Ca2+ and calcium influx through L-type Ca2+ channels. Our results suggest that TMA and TMAO may have the potential to contribute to cardiovascular diseases through their direct effect on vascular contractility in human arteries.


Assuntos
Metilaminas , Artérias Umbilicais , Humanos , Metilaminas/administração & dosagem , Metilaminas/farmacologia , Óxidos , Artérias Umbilicais/efeitos dos fármacos
9.
J Biomol Struct Dyn ; 41(8): 3659-3666, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-35315738

RESUMO

Trimethylamine N-oxide (TMAO) is generally accumulated by organisms and cells to cope with denaturing effects of urea/hydrodynamic pressure on proteins and can even reverse misfolded or aggregated proteins so as to sustain proteostasis. However, most of the work regarding this urea-TMAO counteraction has been performed on folded proteins. Compelling evidence of aggregation of intrinsically disordered proteins (IDPs) like tau, α-synuclein, amyloid ß etc., by TMAO and its potential to impact various protein processes in absence of stressing agents (such as urea) suggests that the contrary feature of interaction profiles of urea and TMAO maximizes their chances of offsetting the perturbing effects of each other. Recently, our lab observed that TMAO induces aggregation of α-casein, a model IDP. In this context, the present study, for the first time, evaluated urea for its potential to counteract the TMAO-induced aggregation of α-casein. It was observed that, at the biologically relevant ratios of 2:1 or 3:1 (urea:TMAO), urea was able to inhibit TMAO-induced aggregation of α-casein. However, urea did not reverse the effects of TMAO on α-casein. In addition to this, α-casein in presence of 1:1 and 2:1 urea:TMAO working ratios show aggregation-induced cytotoxic effect on HEK-293, Neuro2A and HCT-116 cell lines but not in presence of 3:1 working ratio, as there was no aggregation at all. The study infers that the accumulation of TMAO alone in the cells, in absence of stress (such as urea), might result in loss of conformational flexibility and aggregation of IDPs in TMAO accumulating organisms.Communicated by Ramaswamy H. Sarma.


Assuntos
Proteínas Intrinsicamente Desordenadas , Humanos , Caseínas , Peptídeos beta-Amiloides , Ureia/farmacologia , Células HEK293 , Metilaminas/farmacologia
10.
Biol Res Nurs ; 25(3): 353-366, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36444640

RESUMO

BACKGROUND: Impaired wound healing is a health problem around the world, and the search for a novel product to repair wounded skin is a major topic in the field. GW9508 is a synthetic molecule described as a selective agonist of free fatty acid receptors (FFARs) 1 and 4, and there is evidence of its anti-inflammatory effects on several organs of the body. PURPOSE: Here, we aimed to evaluate the effects of topical GW9508 on wound healing in mice. RESEARCH DESIGN: First, we used bioinformatic methods to determine the expression of FFAR1 and FFAR4 mRNA in the skin from a human cell atlas assembled with single-cell transcriptomes. Next, we employed 6-week-old C57BL6J mice with 2 wounds inflicted in the back. The mice were randomly divided into 2 groups, a control group, which received topical vehicle, and a treatment group, which received GW9508, for 12 days. The wound was monitored by photographic documentation every 2 days, and samples were collected at day 6 and 12 post injury for RT-PCR, western blot and histology analyses. RESULTS: FFAR1 and FFAR4 mRNA are expressed in skin cells in similar amounts to those in other tissues. Topical GW9508 accelerated wound healing and decreased gene expression of IL-10 and metalloproteinase 9 on days 6 and 12 post injury. It increased the quantity of Collagen I and improved the organization of collagen fibres. Conclusions: Our results show that GW9508 could be an attractive drug treatment for wounded skin. Future studies need to be performed to assess the impact of GW9508 in chronic wound models.


Assuntos
Cicatriz , Ácidos Graxos não Esterificados , Camundongos , Humanos , Animais , Metilaminas/farmacologia , Pele , Colágeno
11.
BMC Oral Health ; 22(1): 457, 2022 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-36309721

RESUMO

BACKGROUND: The present study aimed to develop a novel protein-repellent and antibacterial polymethyl methacrylate (PMMA) dental resin with 2-methacryloyloxyethyl phosphorylcholine (MPC) and quaternary ammonium dimethylaminohexadecyl methacrylate (DMAHDM), and to investigate the effects of water-aging for 6 months on the mechanical properties, protein adsorption, and antibacterial activity of the dental resin. METHODS: Four groups were tested: PMMA control; PMMA + 3% MPC; PMMA + 1.5% DMAHDM; and PMMA + 3% MPC + 1.5% DMADDM in acrylic resin powder. Specimens were water-aged for 1 d, 3 months, and 6 months at 37 ℃. Their mechanical properties were then measured using a three-point flexure test. Protein adsorption was measured using a micro bicinchoninic acid (BCA) method. A human saliva microcosm model was used to inoculate bacteria on water-aged specimens and to investigate the live/dead staining, metabolic activity of biofilms, and colony-forming units (CFUs). RESULTS: The flexural strength and elastic modulus showed a significant loss after 6 months of water-ageing for the PMMA control (mean ± SD; n = 10); in contrast, the new protein repellent and antibacterial PMMA resin showed no strength loss. The PMMA-MPC-DMAHDM-containing resin imparted a strong antibacterial effect by greatly reducing biofilm viability and metabolic activity. The biofilm CFU count was reduced by about two orders of magnitude (p < 0.05) compared with that of the PMMA resin control. The protein adsorption was 20% that of a commercial composite (p < 0.05). Furthermore, the PMMA-MPC-DMAHDM-containing resin exhibited a long-term antibacterial performance, with no significant difference between 1 d, 3 months and 6 months (p > 0.05). CONCLUSIONS: The flexural strength and elastic modulus of the PMMA-MPC-DMAHDM-containing resin were superior to those of the PMMA control after 6 months of water-ageing. The novel PMMA resin incorporating MPC and DMAHDM exhibited potent and lasting protein-repellent and antibacterial properties.


Assuntos
Polimetil Metacrilato , Água , Humanos , Antibacterianos/farmacologia , Biofilmes , Metacrilatos/farmacologia , Metilaminas/farmacologia , Polimetil Metacrilato/farmacologia , Proteínas , Água/farmacologia , Fatores de Tempo
12.
Biomolecules ; 12(9)2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-36139126

RESUMO

The gut microbial metabolite trimethylamine N-oxide (TMAO) has received increased attention due to its close relationship with cardiovascular disease and type 2 diabetes. In previous studies, TMAO has shown both harmful and beneficial effects on various tissues, but the underlying molecular mechanisms remain to be clarified. Here, we explored the effects of TMAO treatment on H2O2-impaired C2C12 myoblasts, analyzed metabolic changes and identified significantly altered metabolic pathways through nuclear magnetic resonance-based (NMR-based) metabolomic profiling. The results exhibit that TMAO treatment partly alleviated the H2O2-induced oxidative stress damage of cells and protected C2C12 myoblasts by improving cell viability, increasing cellular total superoxide dismutase capacity, improving the protein expression of catalase, and reducing the level of malondialdehyde. We further showed that H2O2 treatment decreased levels of branched-chain amino acids (isoleucine, leucine and valine) and several amino acids including alanine, glycine, threonine, phenylalanine and histidine, and increased the level of phosphocholine related to cell membrane structure, while the TMAO treatment partially reversed the changing trends of these metabolite levels by improving the integrity of the cell membranes. This study indicates that the TMAO treatment may be a promising strategy to alleviate oxidative stress damage in skeletal muscle.


Assuntos
Diabetes Mellitus Tipo 2 , Peróxido de Hidrogênio , Alanina/farmacologia , Catalase/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glicina/metabolismo , Histidina/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Isoleucina , Leucina/metabolismo , Espectroscopia de Ressonância Magnética , Malondialdeído/metabolismo , Metilaminas/metabolismo , Metilaminas/farmacologia , Mioblastos , Estresse Oxidativo , Fenilalanina/metabolismo , Fosforilcolina/farmacologia , Superóxido Dismutase/metabolismo , Treonina , Valina
13.
Cancer Genomics Proteomics ; 19(5): 624-635, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35985690

RESUMO

BACKGROUND/AIM: Metastasis negatively affects the survival of lung cancer patients, however, relatively few compounds have potential in metastasis suppression. This study investigated the molecular targets of N,N-bis (5-ethyl-2-hydroxybenzyl) methylamine (EMD) for metastatic inhibition. MATERIALS AND METHODS: Proteins were analyzed by proteomic and bioinformatic analyses. Protein-protein interaction (PPI) networks were created with the Search Tool for the Retrieval of Interacting Genes. The Kyoto Encyclopedia of Genes and Genomes database and hub genes were used to determine dominant pathways. Immunofluorescence and western blot analyses validated the proteomic results and investigated signaling pathways in NCI-H23 lung cancer cells. RESULTS: A total of 1,751 proteins were common to the control, EMD and N,N-bis(5-methoxy-2-hydroxybenzyl) methylamine (MeMD) groups; 1,980 different proteins were categorized using metastatic capacity category and analyzed for unique proteins affected by EMD. Fifteen proteins were associated with cell adhesion and six with cell migration. Nectin cell adhesion molecule 2 (NECTIN2) was expressed in the control and MeMD-treated groups but not the EMD-treated group, suggesting NECTIN2 as an EMD target. PPI network showed association of NECTIN2 with proteins regulating cancer metastasis. Kyoto Encyclopedia of Genes and Genomes pathways revealed that NECTIN2 is an upstream target of cytoskeletal regulation via SRC signaling. Western blot and immunofluorescence analyses confirmed that EMD suppressed NECTIN2, and its downstream targets, including p-SRC (Y146 and Y527) and the epithelial-to-mesenchymal transition markers tight junction protein 1, vimentin, ß-catenin, snail family transcriptional repressor 1 (SNAI1), and SNAI2, while increasing E-cadherin. CONCLUSION: EMD suppressed NECTIN2-induced activation of EMT signaling. These data support the development of EMD to prevent metastasis of lung cancer.


Assuntos
Neoplasias Pulmonares , Nectinas , Humanos , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Metilaminas/farmacologia , Nectinas/efeitos dos fármacos , Nectinas/metabolismo , Proteômica
14.
J Dent ; 124: 104218, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35817225

RESUMO

OBJECTIVES: Recent studies developed low-shrinkage-stress composite with remineralizing and antibacterial properties to combat secondary caries and increase restoration longevity. However, their long-term durability in thermal cycling is unclear. The objectives of this study were to develop an antibacterial, remineralizing and low-shrinkage-stress composite, and to investigate its durability in thermal cycling for 20,000 cycles, equivalent to two years of clinical life. METHODS: The resin consisted of urethane dimethacrylate (UDMA) and triethylene glycol divinylbenzyl ether (TEG-DVBE). Composites were made with 5% dimethylaminohexadecyl methacrylate (DMAHDM) and 20% of nanoparticles of amorphous calcium phosphate (NACP). Composites were thermal cycled at 5°C and 55°C for 20,000 cycles. A human salivary biofilm model was used to evaluate antibiofilm activity before and after thermal cycling. RESULTS: After 20,000 cycles, the flexural strength of bioactive low-shrinkage-stress composite matched commercial control with no antibacterial activity (p > 0.05). Surface roughness was clinically acceptable at less than 0.2 µm. UV+NACP+DMAHDM composite reduced the total microorganisms, total streptococci, and mutans streptococci by 2-5 logs, compared to commercial composite. Biofilm lactic acid production was reduced by 11 folds. The antibacterial performance was maintained after thermal cycling, with no decrease after 20,000 cycles. CONCLUSIONS: Bioactive low-shrinkage-stress composite possessed good mechanical properties that matched commercial composite both before and after thermal cycling. The new composite had potent antibacterial activity, which was maintained and did not decrease after thermal cycling. CLINICAL SIGNIFICANCE: The new bioactive low-shrinkage-stress composite could reduce polymerization shrinkage stress and release calcium and phosphate ions, with good mechanical properties and strong antibacterial function that were durable after thermal cycling. These properties indicate great potential for inhibiting recurrent caries and increasing the restoration longevity.


Assuntos
Cárie Dentária , Nanocompostos , Antibacterianos/farmacologia , Biofilmes , Fosfatos de Cálcio/farmacologia , Cárie Dentária/prevenção & controle , Humanos , Metacrilatos/farmacologia , Metilaminas/farmacologia , Streptococcus mutans
15.
Cell Death Dis ; 13(4): 299, 2022 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-35379776

RESUMO

A high plasma level of the choline-derived metabolite trimethylamine N-oxide (TMAO) is closely related to the development of cardiovascular disease. However, the underlying mechanism remains unclear. In the present study, we demonstrated that a positive correlation of protein arginine methyltransferase 5 (PRMT5) expression and TMAO-induced vascular inflammation, with upregulated vascular cell adhesion molecule-1 (VCAM-1) expression in primary rat and human vascular smooth muscle cells (VSMC) in vitro. Knockdown of PRMT5 suppressed VCAM-1 expression and the adhesion of primary bone marrow-derived macrophages to TMAO-stimulated VSMC. VSMC-specific PRMT5 knockout inhibited vascular inflammation with decreased expression of VCAM-1 in mice. We further identified that PRMT5 promoted VCAM-1 expression via symmetrical demethylation of Nuclear factor-κB p65 on arginine 30 (R30). Finally, we found that TMAO markedly induced the expression of nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) and production of reactive oxygen species, which contributed to PRMT5 expression and subsequent VCAM-1 expression. Collectively, our data provide novel evidence to establish a Nox4-PRMT5-VCAM-1 in mediating TMAO-induced VSMC inflammation. PRMT5 may be a potential target for the treatment of TMAO-induced vascular diseases.


Assuntos
Músculo Liso Vascular , Molécula 1 de Adesão de Célula Vascular , Animais , Inflamação/genética , Inflamação/metabolismo , Metilaminas/metabolismo , Metilaminas/farmacologia , Camundongos , Músculo Liso Vascular/metabolismo , Ratos , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
16.
Biomolecules ; 11(12)2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34944536

RESUMO

Serum accumulation of the gut microbial metabolite trimethylamine N-oxide (TMAO) is associated with high caloric intake and type 2 diabetes (T2D). Impaired pancreatic ß-cell function is a hallmark of diet-induced T2D, which is linked to hyperglycemia and hyperlipidemia. While TMAO production via the gut microbiome-liver axis is well defined, its molecular effects on metabolic tissues are unclear, since studies in various tissues show deleterious and beneficial TMAO effects. We investigated the molecular effects of TMAO on functional ß-cell mass. We hypothesized that TMAO may damage functional ß-cell mass by inhibiting ß-cell viability, survival, proliferation, or function to promote T2D pathogenesis. We treated INS-1 832/13 ß-cells and primary rat islets with physiological TMAO concentrations and compared functional ß-cell mass under healthy standard cell culture (SCC) and T2D-like glucolipotoxic (GLT) conditions. GLT significantly impeded ß-cell mass and function by inducing oxidative and endoplasmic reticulum (ER) stress. TMAO normalized GLT-mediated damage in ß-cells and primary islet function. Acute 40µM TMAO recovered insulin production, insulin granule formation, and insulin secretion by upregulating the IRE1α unfolded protein response to GLT-induced ER and oxidative stress. These novel results demonstrate that TMAO protects ß-cell function and suggest that TMAO may play a beneficial molecular role in diet-induced T2D conditions.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Endorribonucleases/metabolismo , Células Secretoras de Insulina/citologia , Metilaminas/farmacologia , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Tipo 2/prevenção & controle , Estresse do Retículo Endoplasmático , Feminino , Microbioma Gastrointestinal , Regulação da Expressão Gênica/efeitos dos fármacos , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Modelos Biológicos , Estresse Oxidativo , Cultura Primária de Células , Ratos
17.
Int J Mol Sci ; 22(20)2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34681805

RESUMO

Diet is a well-known risk factor of cardiovascular diseases (CVDs). Some microRNAs (miRNAs) have been described to regulate molecular pathways related to CVDs. Diet can modulate miRNAs and their target genes. Choline, betaine, and l-carnitine, nutrients found in animal products, are metabolized into trimethylamine n-oxide (TMAO), which has been associated with CVD risk. The aim of this study was to investigate TMAO regulation of CVD-related miRNAs and their target genes in cellular models of liver and macrophages. We treated HEPG-2, THP-1, mouse liver organoids, and primary human macrophages with 6 µM TMAO at different timepoints (4, 8, and 24 h for HEPG-2 and mouse liver organoids, 12 and 24 h for THP-1, and 12 h for primary human macrophages) and analyzed the expression of a selected panel of CVD-related miRNAs and their target genes and proteins by real-time PCR and Western blot, respectively. HEPG-2 cells were transfected with anti-miR-30c and syn-miR-30c. TMAO increased the expression of miR-21-5p and miR-30c-5p. PER2, a target gene of both, decreased its expression with TMAO in HEPG-2 and mice liver organoids but increased its mRNA expression with syn-miR-30c. We concluded that TMAO modulates the expression of miRNAs related to CVDs, and that such modulation affects their target genes.


Assuntos
Doenças Cardiovasculares/genética , Metilaminas/farmacologia , MicroRNAs/efeitos dos fármacos , Animais , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/fisiologia , Proteínas Circadianas Period/efeitos dos fármacos , Proteínas Circadianas Period/genética , Células THP-1
18.
Diabetes ; 70(10): 2250-2263, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34315726

RESUMO

Choline metabolite trimethylamine N-oxide (TMAO) has been recognized as a risk factor of gestational diabetes mellitus (GDM), but its exact role in GDM has not been reported. In this study, we focused on the placenta development to reveal the role of TMAO in GDM. We found that the TMAO levels in peripheral and cord plasma were increased in women with GDM and that TMAO levels were positively correlated with newborn weight and placental thickness. Neutrophil extracellular traps (NETs) in the peripheral and cord plasma and the myeloperoxidase expression in the placenta of women with GDM also increased. NETs could inhibit the proliferation, migration, invasion, and angiogenesis of HTR-8/Svneo cells. However, TMAO not only could inhibit the formation of NETs but also could enhance the biological function of HTR-8/Svneo cells. With induction of GDM in NETs-deficient PAD4-/- and wild-type mice, the placental weight of PAD4-/- mice increased significantly. TMAO feeding also inhibited the formation of NETs and further increased the weight of the placenta and fetuses, and this increase did not affect the placental structure. Our data indicate that higher TMAO levels and the formation of abnormal NETs were associated with GDM. TMAO not only could promote the development of the placenta and fetuses but also could inhibit the formation of NETs.


Assuntos
Diabetes Gestacional/fisiopatologia , Armadilhas Extracelulares/efeitos dos fármacos , Metilaminas/farmacologia , Placentação/efeitos dos fármacos , Adulto , Animais , Estudos de Casos e Controles , Células Cultivadas , Colina/metabolismo , Diabetes Gestacional/patologia , Armadilhas Extracelulares/metabolismo , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Desenvolvimento Fetal/genética , Humanos , Recém-Nascido , Metilaminas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Proteína-Arginina Desiminase do Tipo 4/genética , Adulto Jovem
19.
Int J Neuropsychopharmacol ; 24(9): 749-757, 2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34228806

RESUMO

BACKGROUND: Though drugs binding to serotonergic 5-HT2A receptors have long been claimed to influence human anxiety, it remains unclear if this receptor subtype is best described as anxiety promoting or anxiety dampening. Whereas conditioned fear expressed as freezing in rats is modified by application of 5-HT2A-acting drugs locally into different brain regions, reports on the effect of systemic administration of 5-HT2A receptor agonists and 5-HT2A antagonists or inverse agonists on this behavior remain sparse. METHODS: We assessed the possible impact of systemic administration of 5-HT2A receptor agonists, 5-HT2A receptor inverse agonists, and a selective serotonin reuptake inhibitor (SSRI)-per se or in combination-on the freezing displayed by male rats when re-exposed to a conditioning chamber in which they received foot shocks 7 days earlier. RESULTS: The 5-HT2A receptor agonists psilocybin and 25CN-NBOH induced a reduction in conditioned fear that was countered by pretreatment with 5-HT2A receptor inverse agonist MDL 100907. While both MDL 100907 and another 5-HT2A receptor inverse agonist, pimavanserin, failed to impact freezing per se, both compounds unmasked a robust fear-reducing effect of an SSRI, escitalopram, which by itself exerted no such effect. CONCLUSIONS: The results indicate that 5-HT2A receptor activation is not a prerequisite for normal conditioned freezing in rats but that this receptor subtype, when selectively over-activated prior to expression, exerts a marked fear-reducing influence. However, in the presence of an SSRI, the 5-HT2A receptor, on the contrary, appears to counter an anti-freezing effect of the enhanced extracellular serotonin levels following reuptake inhibition.


Assuntos
Comportamento Animal/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Medo/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Agonistas do Receptor 5-HT2 de Serotonina/farmacologia , Antagonistas do Receptor 5-HT2 de Serotonina/farmacologia , Animais , Compostos Bicíclicos com Pontes/farmacologia , Fluorbenzenos/farmacologia , Ligantes , Masculino , Metilaminas/farmacologia , Piperidinas/farmacologia , Psilocibina/farmacologia , Ratos , Ratos Sprague-Dawley , Ureia/análogos & derivados , Ureia/farmacologia
20.
Circ Heart Fail ; 14(6): e007909, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34129361

RESUMO

BACKGROUND: Trimethylamine N-oxide (TMAO)-a gut-derived metabolite-is elevated in heart failure (HF) and linked to poor prognosis. We investigated variations in TMAO in HF, left ventricular assist device (LVAD), and heart transplant (HT) and assessed its relation with inflammation, endotoxemia, oxidative stress, and gut dysbiosis. METHODS: We enrolled 341 patients. TMAO, CRP (C-reactive protein), IL (interleukin)-6, TNF-α (tumor necrosis factor alpha), ET-1 (endothelin-1), adiponectin, lipopolysaccharide, soluble CD14, and isoprostane were measured in 611 blood samples in HF (New York Heart Association class I-IV) and at multiple time points post-LVAD and post-HT. Gut microbiota were assessed via 16S rRNA sequencing among 327 stool samples. Multivariable regression models were used to assess the relationship between TMAO and (1) New York Heart Association class; (2) pre- versus post-LVAD or post-HT; (3) biomarkers of inflammation, endotoxemia, oxidative stress, and microbial diversity. RESULTS: ln-TMAO was lower among HF New York Heart Association class I (1.23 [95% CI, 0.52-1.94] µM) versus either class II, III, or IV (1.99 [95% CI, 1.68-2.30], 1.97 [95% CI, 1.71-2.24], and 2.09 [95% CI, 1.83-2.34] µM, respectively; all P<0.05). In comparison to class II-IV, ln-TMAO was lower 1 month post-LVAD (1.58 [95% CI, 1.32-1.83] µM) and 1 week and 1 month post-HT (0.97 [95% CI, 0.60-1.35] and 1.36 [95% CI, 1.01-1.70] µM). ln-TMAO levels in long-term LVAD (>6 months: 1.99 [95% CI, 1.76-2.22] µM) and HT (>6 months: 1.86 [95% CI, 1.66-2.05] µM) were not different from symptomatic HF. After multivariable adjustments, TMAO was not associated with biomarkers of inflammation, endotoxemia, oxidative stress, or microbial diversity. CONCLUSIONS: TMAO levels are increased in symptomatic HF patients and remain elevated long term after LVAD and HT. TMAO levels were independent from measures of inflammation, endotoxemia, oxidative stress, and gut dysbiosis.


Assuntos
Disbiose/tratamento farmacológico , Insuficiência Cardíaca/tratamento farmacológico , Inflamação/tratamento farmacológico , Metilaminas/farmacologia , Tempo , Idoso , Idoso de 80 Anos ou mais , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Insuficiência Cardíaca/fisiopatologia , Transplante de Coração/métodos , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Coração Auxiliar , Humanos , Masculino , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...