Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Metabolomics ; 18(10): 76, 2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-36180605

RESUMO

INTRODUCTION: Pro-inflammatory cytokines are responsible for initiating an effective defense against exogenous pathogens, and their regulation has a vital role in maintaining physiological homeostasis. The involvement of pro-inflammatory cytokines in pathological conditions have been explored in great detail, however, studies investigating metabolic pathways associated with these cytokines under normal homeostatic conditions are scarce. OBJECTIVES: The aim of the current study was to identify metabolites and metabolic pathways associated with circulating pro-inflammatory cytokines under homeostatic conditions using a metabolomics approach. METHODS: The study participants (n = 133) were derived from the Newfoundland Osteoarthritis Study (NFOAS) and the Complex Diseases in the Newfoundland population: Environment and Genetics (CODING) study. Plasma concentrations of cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-1 beta (IL-1ß), and macrophage migration inhibitory factor (MIF) were assessed by enzyme-linked immunosorbent assay. Targeted metabolomic profiling on fasting plasma samples was performed using Biocrates MxP® Quant 500 kit which measures a total of 630 metabolites. Associations between natural log-transformed metabolite concentrations and metabolite sums/ratios and cytokine levels were assessed using linear regression with adjustment for age, sex, body mass index (BMI), and osteoarthritis status. RESULTS: Seven metabolites and 11 metabolite sums/ratios were found to be significantly associated with TNF-α, IL-1ß, and MIF (all p ≤ 5.13 × 10- 5) after controlling multiple testing with Bonferroni method, indicating the association between glutathione (GSH), polyamine, and lysophosphatidylcholine (lysoPC) synthesis pathways and these pro-inflammatory cytokines. CONCLUSION: GSH, polyamine, and lysoPC synthesis pathways were positively associated with circulating TNF-α, IL-1ß, and MIF levels under homeostatic conditions.


Assuntos
Fatores Inibidores da Migração de Macrófagos , Osteoartrite , Glutationa , Humanos , Interleucina-1beta , Interleucina-6 , Lisofosfatidilcolinas , Metabolômica , Poliaminas , Fator de Necrose Tumoral alfa
2.
J Cell Biochem ; 119(3): 2806-2817, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29058761

RESUMO

The excessive recruitment and improper activation of polymorphonuclear neutrophils (PMNs) often induces serious injury of host tissues, leading to inflammatory disorders. Therefore, to understand the molecular mechanism on neutrophil recruitment possesses essential pathological and physiological importance. In this study, we found that physiological shear stress induces c-Abl kinase activation in neutrophils, and c-Abl kinase inhibitor impaired neutrophil crawling behavior on ICAM-1. We further identified Vav1 was a downstream effector phosphorylated at Y174 and Y267. Once activated, c-Abl kinase regulated the activity of Vav1, which further affected Rac1/PAK1/LIMK1/cofilin signaling pathway. Here, we demonstrate a novel signaling function and critical role of c-Abl kinase during neutrophil crawling under physiological shear by regulating Vav1. These findings provide a promising treatment strategy for inflammation-related disease by inactivation of c-Abl kinase to restrict neutrophil recruitment.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Movimento Celular , Quinases Lim/metabolismo , Neutrófilos/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Resistência ao Cisalhamento , Transdução de Sinais , Quinases Ativadas por p21/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Feminino , Células HEK293 , Humanos , Masculino , Neutrófilos/citologia
3.
Biochem Biophys Res Commun ; 498(1): 214-220, 2018 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-29501746

RESUMO

Recent work has found that complex I is the sole source of reactive oxygen species (ROS) during myocardial ischemia-reperfusion (IR) injury. However, it has also been reported that heart mitochondria can also generate ROS from other sources in the respiratory chain and Krebs cycle. This study examined the impact of partial complex I deficiency due to selective loss of the Ndufs4 gene on IR injury to heart tissue. Mice heterozygous for NDUFS4 (NDUFS4+/-) did not display any significant changes in overall body or organ weight when compared to wild-type (WT) littermates. There were no changes in superoxide (O2●-)/hydrogen peroxide (H2O2) release from cardiac or liver mitochondria isolated from NDUFS4 ±â€¯mice. Using selective ROS release inhibitors, we found that complex III is a major source of ROS in WT and NDUFS4 ±â€¯cardiac mitochondria respiring under state 4 conditions. Subjecting hearts from NDUFS4 ±â€¯mice to reperfusion injury revealed that the partial loss of complex I decreases contractile recovery and increases myocardial infarct size. These results correlated with a significant increase in O2●-/H2O2 release rates in mitochondria isolated from NDUFS4 ±â€¯hearts subjected to an IR challenge. Taken together, these results demonstrate that the partial absence of complex I sensitizes the myocardium towards IR injury and that the main source of ROS following reperfusion is complex III.


Assuntos
Complexo I de Transporte de Elétrons/deficiência , Peróxido de Hidrogênio/metabolismo , Mitocôndrias Cardíacas/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Superóxidos/metabolismo , Animais , Antioxidantes/metabolismo , Peso Corporal , Complexo I de Transporte de Elétrons/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Deleção de Genes , Masculino , Camundongos , Miocárdio/metabolismo , Miocárdio/patologia , Tamanho do Órgão
4.
J Mol Cell Cardiol ; 91: 104-13, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26746142

RESUMO

AMP-activated kinase (AMPK) is a stress responsive kinase that regulates cellular metabolism and protects against cardiomyocyte injury during ischemia-reperfusion (IR). Mitochondria play an important role in cell survival, but the specific actions of activated AMPK in maintaining mitochondrial integrity and function during reperfusion are unknown. Thus, we assessed the consequences of AMPK inactivation on heart mitochondrial function during reperfusion. Mouse hearts expressing wild type (WT) or kinase-dead (KD) AMPK were studied. Mitochondria isolated from KD hearts during reperfusion had intact membrane integrity, but demonstrated reduced oxidative capacity, increased hydrogen peroxide production and decreased resistance to mitochondrial permeability transition pore opening compared to WT. KD hearts showed increased activation of the mitogen activated protein kinase kinase 4 (MKK4) and downstream c-Jun terminal kinase (JNK) and greater necrosis during reperfusion after coronary occlusion. Transgenic expression of mitochondrial catalase (MCAT) prevented the excessive cardiac JNK activation and attenuated the increased myocardial necrosis observed during reperfusion in KD mice. Inhibition of JNK increased the resistance of KD hearts to mPTP opening, contractile dysfunction and necrosis during IR. Thus, intrinsic activation of AMPK is critical to prevent excess mitochondrial reactive oxygen production and consequent JNK signaling during reperfusion, thereby protecting against mPTP opening, irreversible mitochondrial damage and myocardial injury.


Assuntos
MAP Quinase Quinase 4/genética , Infarto do Miocárdio/genética , Necrose/genética , Animais , Apoptose/efeitos dos fármacos , Catalase/genética , Catalase/metabolismo , Regulação da Expressão Gênica , Peróxido de Hidrogênio/antagonistas & inibidores , Peróxido de Hidrogênio/metabolismo , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Reperfusão Miocárdica , Miocárdio/metabolismo , Miocárdio/patologia , Necrose/metabolismo , Necrose/patologia , Necrose/prevenção & controle , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Transgenes
5.
Proc Natl Acad Sci U S A ; 110(40): 16133-8, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24043794

RESUMO

Urocortin 2 (Ucn2), a peptide of the corticotropin-releasing factor (CRF) family, binds with high affinity to type 2 CRF receptors (CRFR2) on cardiomyocytes and confers protection against ischemia/reperfusion. The mechanisms by which the Ucn2-CRFR2 axis mitigates against ischemia/reperfusion injury remain incompletely delineated. Activation of AMP-activated protein kinase (AMPK) also limits cardiac damage during ischemia/reperfusion. AMPK is classically activated by alterations in cellular energetics; however, hormones, cytokines, and additional autocrine/paracrine factors also modulate its activity. We examined the effects of both the endogenous cardiac Ucn2 autocrine/paracrine pathway and Ucn2 treatment on AMPK regulation. Ucn2 treatment increased AMPK activation and downstream acetyl-CoA carboxylase phosphorylation and glucose uptake in isolated heart muscles. These actions were blocked by the CRFR2 antagonist anti-sauvagine-30 and by a PKCε translocation-inhibitor peptide (εV1-2). Hypoxia-induced AMPK activation was also blunted in heart muscles by preincubation with either anti-sauvagine-30, a neutralizing anti-Ucn2 antibody, or εV1-2. Treatment with Ucn2 in vivo augmented ischemic AMPK activation and reduced myocardial injury and cardiac contractile dysfunction after regional ischemia/reperfusion in mice. Ucn2 also directly activated AMPK in ex vivo-perfused mouse hearts and diminished injury and contractile dysfunction during ischemia/reperfusion. Thus, both Ucn2 treatment and the endogenous cardiac Ucn2 autocrine/paracrine pathway activate AMPK signaling pathway, via a PKCε-dependent mechanism, defining a Ucn2-CRFR2-PKCε-AMPK pathway that mitigates against ischemia/reperfusion injury.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Miocárdio/enzimologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/fisiologia , Urocortinas/farmacologia , Acetil-CoA Carboxilase/metabolismo , Análise de Variância , Animais , Anticorpos Neutralizantes/farmacologia , Hormônio Liberador da Corticotropina/sangue , Hormônio Liberador da Corticotropina/metabolismo , Ativação Enzimática/efeitos dos fármacos , Immunoblotting , Imuno-Histoquímica , Camundongos , Fragmentos de Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Urocortinas/sangue , Urocortinas/metabolismo
6.
Transl Psychiatry ; 14(1): 210, 2024 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-38802393

RESUMO

Atypical antipsychotics (AAPs) are primary medications for schizophrenia (SZ). However, their use is frequently associated with the development of adverse metabolic effects, and the mechanisms behind these negative effects remain inadequately elucidated. To investigate the role of macrophage migration inhibitory factor (MIF) in regulating antipsychotic-induced metabolic abnormalities, between 2017 and 2020, a cross-sectional study was conducted, involving 142 healthy individuals and 388 SZ patients undergoing treatment with either typical antipsychotic (TAP) or AAP medications. Symptoms of SZ patients were evaluated using the Positive and Negative Syndrome Scale (PANSS), and measurements of metabolic indices and plasma MIF levels were performed on all individuals. A significant increase in plasma MIF levels was observed in groups receiving five major AAP monotherapies in comparison to healthy controls (all p < 0.0001). There was no such increase shown in the group receiving TAP treatment (p > 0.05). Elevated plasma MIF levels displayed a notable correlation with insulin resistance (ß = 0.024, p = 0.020), as well as with the levels of triglycerides (ß = 0.019, p = 0.001) and total cholesterol (ß = 0.012, p = 0.038) in the groups receiving AAPs. However, while the TAP group also displayed certain metabolic dysfunction compared to healthy controls, no significant association was evident with plasma MIF levels (all p > 0.05). In conclusion, plasma MIF levels exhibit a distinctive correlation with metabolic abnormalities triggered by AAPs. Hence, there is potential for further development of MIF as a distinctive marker for monitoring adverse metabolic effects induced by AAPs in clinical settings.


Assuntos
Antipsicóticos , Oxirredutases Intramoleculares , Fatores Inibidores da Migração de Macrófagos , Esquizofrenia , Humanos , Fatores Inibidores da Migração de Macrófagos/sangue , Masculino , Antipsicóticos/efeitos adversos , Antipsicóticos/uso terapêutico , Feminino , Adulto , Esquizofrenia/tratamento farmacológico , Esquizofrenia/sangue , Estudos Transversais , Oxirredutases Intramoleculares/sangue , Pessoa de Meia-Idade , Resistência à Insulina , Estudos de Casos e Controles , Triglicerídeos/sangue
7.
Mol Metab ; 79: 101834, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37935315

RESUMO

Attenuation of adipose hormone sensitive lipase (HSL) may impair lipolysis and exacerbate obesity. We investigate the role of cytokine, macrophage migration inhibitory factor (MIF) in regulating adipose HSL and adipocyte hypertrophy. Extracellular MIF downregulates HSL in an autocrine fashion, by activating the AMPK/JNK signaling pathway upon binding to its membrane receptor, CD74. WT mice fed high fat diet (HFD), as well as mice overexpressing MIF, both had high circulating MIF levels and showed suppression of HSL during the development of obesity. Blocking the extracellular action of MIF by a neutralizing MIF antibody significantly reduced obesity in HFD mice. Interestingly, intracellular MIF binds with COP9 signalosome subunit 5 (Csn5) and JNK, which leads to an opposing effect to inhibit JNK phosphorylation. With global MIF deletion, adipocyte JNK phosphorylation increased, resulting in decreased HSL expression, suggesting that the loss of MIF's intracellular inhibitory action on JNK was dominant in Mif-/- mice. Adipose tissue from Mif-/- mice also exhibited higher Akt and lower PKA phosphorylation following HFD feeding compared with WT, which may contribute to the downregulation of HSL activation during more severe obesity. Both intracellular and extracellular MIF have opposing effects to regulate HSL, but extracellular actions predominate to downregulate HSL and exacerbate the development of obesity during HFD.


Assuntos
Fatores Inibidores da Migração de Macrófagos , Animais , Camundongos , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Fatores Inibidores da Migração de Macrófagos/genética , Fatores Inibidores da Migração de Macrófagos/metabolismo , Obesidade/metabolismo , Esterol Esterase/metabolismo
8.
J Control Release ; 358: 78-97, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37076016

RESUMO

The establishment of in vitro models of the BBB is significant for the evaluation of the mechanism and permeability of drugs and their sustained-release formulations across the BBB. Among the different models, the immortalized human cell line hCMEC/D3 has the potential to be used for a standardized in vitro BBB model due to its high throughput, reproducibility, homology and low cost. The high permeability of the paracellular pathway and the low expression of both certain transporters and metabolic enzymes in this model lead to low physiological levels of physical, transport and metabolic barriers, thus limiting the application of these cells. The barrier properties of this model have been improved in different studies by various means. However, no systematic review has been conducted on the optimization of model-building conditions or on the regulation and expression of transporters in the models. Some existing reviews focus on the overall description of the entire field of blood-brain barrier in vitro models, lacking in-depth and systematic reviews on the experimental details and model evaluation methods based on hCMEC/D3.This paper deals with a detailed review on the optimization of multiple aspects and modalities of the hCMEC/D3 cell culture process, including initial medium, optimal serum levels, Transwell membrane materials, supra-membrane supports, inoculum density, endogenous growth factor, exogenous drug levels, co-culture and transfection methods, to provide references for the establishment and evaluation of hCMEC/D3 cell models.


Assuntos
Barreira Hematoencefálica , Células Endoteliais , Humanos , Barreira Hematoencefálica/metabolismo , Reprodutibilidade dos Testes , Células Endoteliais/metabolismo , Transporte Biológico , Linhagem Celular
9.
Environ Sci Pollut Res Int ; 30(7): 19304-19312, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36227495

RESUMO

Eighty 24-week-old laying hens were divided into eight groups, seven given a single oral dose per chicken with 7 dosing levels from 13.6 to 137 mg/kg body weight (bw) and one serving as sham control. The hens were observed for 28 days for clinical abnormalities, egg yield, and body weight. Egg samples from groups of low-to-medium doses were analyzed for residues of fipronil and its metabolites by LC-MS/MS. Blood and organ samples from hens of the group receiving 63.3 mg/kg bw were collected for hematochemical and histopathological analysis. We found that the median lethal dose (LD50) of fipronil was 74 mg/kg bw for laying hens. No death occurred, and there were no obvious changes in body weight and egg production in the hens receiving doses at or below 20 mg/kg bw. In the hens that survived exposure to the dose at 63.3 mg/kg bw, there was significant reduction in body weight and egg yield; histopathological changes in the liver and kidney; and increased levels of creatine, urea, glutamate oxaloacetate transferase, and glutamate pyruvic transaminase. Fipronil-sulfone was the residual marker in eggs with significantly higher concentrations and longer withdrawal periods than its maternal compound. We conclude that fipronil is efficiently transformed into fipronil-sulfone in the body with subsequent excretion into eggs. More attention should be paid to the potential food safety risk of fipronil-sulfone because of its persistence in eggs.


Assuntos
Galinhas , Espectrometria de Massas em Tandem , Animais , Feminino , Cromatografia Líquida , Galinhas/metabolismo , Ovos/análise , Peso Corporal , Sulfonas
10.
Sci Rep ; 13(1): 13068, 2023 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-37567977

RESUMO

Switching to normal diet (ND) is the regular therapy for high-fat diet (HFD)-induced nonalcoholic fatty liver disease (NAFLD). Intermittent fasting (IF) is a unique treatment which may exhibits better therapeutic efficacy. Thus, we aim to investigate the therapeutic effects of these treatments and exploring the mechanisms. In the present study, NAFLD mouse model was induced by a 10-week HFD. Thereafter, mice adopted continued HFD, ND, or IF for the next 12 weeks. Finally, the liver was then harvested to assess lipid deposition, lipid metabolism, apoptosis, and autophagy, while blood was collected to determine blood glucose and insulin. The results showed that IF and ND treatment improved lipid deposition and metabolic disorder of NAFLD mice; the increasing body weight, liver weight, and HOMA-IR index of HFD mice were also alleviated by IF and ND. Furthermore, IF and ND treatment activated the macrophage migration inhibitory factor (MIF)/AMPK pathway and regulated its downstream autophagy and apoptosis. However, the efficacy of IF was better than ND. Both IF and ND activates MIF signaling and alleviate the lipotoxicity of NAFLD while IF therapy is more effective than ND. The different MIF up-regulation might be the underlying mechanism of why IF benefits more than ND.


Assuntos
Fatores Inibidores da Migração de Macrófagos , Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Dieta Hiperlipídica/efeitos adversos , Jejum Intermitente , Metabolismo dos Lipídeos , Lipídeos , Fígado/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/terapia , Hepatopatia Gordurosa não Alcoólica/metabolismo
11.
iScience ; 26(6): 106923, 2023 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-37283810

RESUMO

While insulin resistance (IR) is associated with inflammation in white adipose tissue, we report a non-inflammatory adipose mechanism of high fat-induced IR mediated by loss of Pref-1. Pref-1, released from adipose Pref-1+ cells with characteristics of M2 macrophages, endothelial cells or progenitors, inhibits MIF release from both Pref-1+ cells and adipocytes by binding with integrin ß1 and inhibiting the mobilization of p115. High palmitic acid induces PAR2 expression in Pref-1+ cells, downregulating Pref-1 expression and release in an AMPK-dependent manner. The loss of Pref-1 increases adipose MIF secretion contributing to non-inflammatory IR in obesity. Treatment with Pref-1 blunts the increase in circulating plasma MIF levels and subsequent IR induced by a high palmitic acid diet. Thus, high levels of fatty acids suppress Pref-1 expression and secretion, through increased activation of PAR2, resulting in an increase in MIF secretion and a non-inflammatory adipose mechanism of IR.

12.
Am J Physiol Heart Circ Physiol ; 303(6): H732-42, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22842069

RESUMO

Toll-like receptor (TLR)4 regulates inflammation and metabolism and has been linked to the pathogenesis of heart disease. TLR4 is upregulated in diabetic cardiomyocytes, and we examined the role of TLR4 in modulating cardiac fatty acid (FA) metabolism and the pathogenesis of diabetic heart disease in nonobese diabetic (NOD) mice. Both wild-type (WT) NOD and TLR4-deficient NOD animals had increased plasma triglyceride levels after the onset of diabetes. However, by comparison, TLR4-deficient NOD mouse hearts had lower triglyceride accumulation in the early stages of diabetes, which was associated with a reduction in myeloid differentiation primary response gene (88) (MyD88), phosphorylation of p38 MAPK (phospho-p38), lipoprotein lipase (LPL), and JNK levels but increased phospho-AMP-activated protein kinase (AMPK). Oleic acid treatment in H9C2 cardiomyocytes also led to cellular lipid accumulation, which was attenuated by TLR4 small interfering RNA. TLR4 deficiency in the cells decreased FA-induced augmentation of MyD88, phospho-p38, and LPL, suggesting that TLR4 may modulate FA-induced lipid metabolism in cardiomyocytes. In addition, although cardiac function was impaired in both diabetic WT NOD and TLR4-deficient NOD animals compared with control nondiabetic mice, this deficit was less in the diabetic TLR4-deficient NOD mice, which had greater ejection fraction, greater fractional shortening, and increased left ventricular developed pressure in the early stages after the development of diabetes compared with their diabetic WT NOD counterparts. Thus, we conclude that TLR4 plays a role in regulating lipid accumulation in cardiac muscle after the onset of type 1 diabetes, which may contribute to cardiac dysfunction.


Assuntos
Diabetes Mellitus Tipo 1/complicações , Cardiomiopatias Diabéticas/etiologia , Metabolismo dos Lipídeos , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Receptor 4 Toll-Like/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Glicemia/metabolismo , Linhagem Celular , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Cardiomiopatias Diabéticas/sangue , Cardiomiopatias Diabéticas/genética , Cardiomiopatias Diabéticas/patologia , Modelos Animais de Doenças , Ácidos Graxos não Esterificados/sangue , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipase Lipoproteica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , Ácido Oleico/metabolismo , Fosforilação , Interferência de RNA , Ratos , Fatores de Tempo , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética , Triglicerídeos/sangue , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Artigo em Inglês | MEDLINE | ID: mdl-34358623

RESUMO

Inflammation is involved in the pathogenesis of psychiatric disorders. Many previous studies have defined the important roles of inflammatory factors in the pathogenesis, diagnosis, and treatment outcomes of psychiatric disorders. Macrophage migration inhibitory factor (MIF), a pro-inflammatory factor, has been gradually recognized to be involved in the development of neurological diseases in recent years. Our current review focuses on discussing the potential beneficial and detrimental roles of MIF in psychiatric disorders. We will provide new mechanistic insights for the development of potential diagnostic and therapeutic biomarkers based on MIF for psychiatric diseases.


Assuntos
Biomarcadores , Inflamação , Fatores Inibidores da Migração de Macrófagos/imunologia , Transtornos Mentais/imunologia , Doença de Alzheimer/imunologia , Animais , Depressão/imunologia , Humanos , Doenças do Sistema Nervoso , Esquizofrenia/imunologia
14.
Curr Drug Deliv ; 19(4): 508-517, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34238184

RESUMO

BACKGROUND: The research and development of drugs for the treatment of central nervous system diseases faces many challenges at present. One of the most important questions to be answered is, how does the drug cross the blood-brain barrier to get to the target site for pharmacological action. Fluoxetine is widely used in clinical antidepressant therapy. However, the mechanism by which fluoxetine passes through the BBB also remains unclear. Under physiological pH conditions, fluoxetine is an organic cation with a relatively small molecular weight (<500), which is in line with the substrate characteristics of organic cation transporters (OCTs). Therefore, this study aimed to investigate the interaction of fluoxetine with OCTs at the BBB and BBB-associated efflux transporters. This is of great significance for fluoxetine to better treat depression. Moreover, it can provide a theoretical basis for clinical drug combination. METHODS: In vitro BBB model was developed using human brain microvascular endothelial cells (hCMEC/D3), and the cellular accumulation was tested in the presence or absence of transporter inhibitors. In addition, an in vivo trial was performed in rats to investigate the effect of OCTs on the distribution of fluoxetine in the brain tissue. Fluoxetine concentration was determined by a validated UPLC-MS/MS method. RESULTS: The results showed that amantadine (an OCT1/2 inhibitor) and prazosin (an OCT1/3 inhibitor) significantly decreased the cellular accumulation of fluoxetine (P <.001). Moreover, we found that N-methylnicotinamide (an OCT2 inhibitor) significantly inhibited the cellular uptake of 100 and 500 ng/mL fluoxetine (P <.01 and P <.05 respectively). In contrast, corticosterone (an OCT3 inhibitor) only significantly inhibited the cellular uptake of 1000 ng/mL fluoxetine (P <.05). The P-glycoprotein (P-gp) inhibitor, verapamil, and the multidrug resistance associated proteins (MRPs) inhibitor, MK571, significantly decreased the cellular uptake of fluoxetine. However, intracellular accumulation of fluoxetine was not significantly changed when fluoxetine was incubated with the breast cancer resistance protein (BCRP) inhibitor Ko143. Furthermore, in vivo experiments proved that corticosterone and prazosin significantly inhibited the brain-plasma ratio of fluoxetine at 5.5 h and 12 h, respectively. CONCLUSION: OCTs might play a significant role in the transport of fluoxetine across the BBB. In addition, P-gp, BCRP, and MRPs seemed not to mediate the efflux transport of fluoxetine.


Assuntos
Barreira Hematoencefálica , Fluoxetina , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Cromatografia Líquida , Corticosterona/metabolismo , Corticosterona/farmacologia , Células Endoteliais/metabolismo , Fluoxetina/metabolismo , Fluoxetina/farmacologia , Humanos , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/farmacologia , Prazosina/metabolismo , Prazosina/farmacologia , Ratos , Espectrometria de Massas em Tandem
15.
Redox Biol ; 57: 102481, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36148770

RESUMO

Metformin is widely used to surmount insulin resistance (IR) and type 2 diabetes. Accumulating evidence suggests that metformin may improve IR through regulating gut microbiota and bile acids. However, the underlying mechanisms remain unclear. Our metabolomic analysis showed that metformin significantly increased the accumulation of tauroursodeoxycholic acid (TUDCA) in intestine and liver from high-fat diet (HFD)-induced IR mice. TUDCA also alleviated IR, and reduced oxidative stress and intestinal inflammation in ob/ob mice. TUDCA blocked KEAP1 to bind with Nrf2, resulting in Nrf2 translocation into nuclear and initiating the transcription of antioxidant genes, which eventually reduced intracellular ROS accumulation and improved insulin signaling. Analysis of gut microbiota further revealed that metformin reduced the relative abundance of Bifidobacterium, which produces bile salt hydrolase (BSH). The reduction in BSH was probably crucial for the accumulation of TUDCA. Metformin also increased the proportion of Akkermanisia muciniphlia in gut microbiota of ob/ob mice via TUDCA. These beneficial effects of metformin in remodeling gut microbiota, reducing oxidative stress and improving insulin sensitivity were partly due to the accumulation of TUDCA, suggesting that TUDCA may be a potential therapy for metabolic syndrome.

16.
Metabolites ; 12(4)2022 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-35448521

RESUMO

Obesity is a global pandemic, but there is yet no effective measure to control it. Recent metabolomics studies have identified a signature of altered amino acid profiles to be associated with obesity, but it is unclear whether these findings have actionable clinical potential. The aims of this study were to reveal the metabolic alterations of obesity and to explore potential strategies to mitigate obesity. We performed targeted metabolomic profiling of the plasma/serum samples collected from six independent cohorts and conducted an individual data meta-analysis of metabolomics for body mass index (BMI) and obesity. Based on the findings, we hypothesized that restriction of branched-chain amino acids (BCAAs), phenylalanine, or tryptophan may prevent obesity and tested our hypothesis in a dietary restriction trial with eight groups of 4-week-old male C57BL/6J mice (n = 5/group) on eight different types of diets, respectively, for 16 weeks. A total of 3397 individuals were included in the meta-analysis. The mean BMI was 30.7 ± 6.1 kg/m2, and 49% of participants were obese. Fifty-eight metabolites were associated with BMI and obesity (all p ≤ 2.58 × 10-4), linked to alterations of the BCAA, phenylalanine, tryptophan, and phospholipid metabolic pathways. The restriction of BCAAs within a high-fat diet (HFD) maintained the mice's weight, fat and lean volume, subcutaneous and visceral adipose tissue weight, and serum glucose and insulin at levels similar to those in the standard chow group, and prevented obesity, adipocyte hypertrophy, adipose inflammation, and insulin resistance induced by HFD. Our data suggest that four metabolic pathways, BCAA, phenylalanine, tryptophan, and phospholipid metabolic pathways, are altered in obesity and restriction of BCAAs within a HFD can prevent the development of obesity and insulin resistance in mice, providing a promising strategy to potentially mitigate diet-induced obesity.

17.
J Mol Cell Cardiol ; 51(1): 24-32, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21402077

RESUMO

AMP-activated protein kinase (AMPK) is a stress signaling enzyme that orchestrates the regulation of energy-generating and -consuming pathways. Intrinsic AMPK activation protects the heart against ischemic injury and apoptosis, but whether pharmacologic AMPK stimulation mitigates ischemia-reperfusion damage is unknown. The aims of this study were to determine whether direct stimulation of AMPK using a small molecule activator, A-769662, attenuates myocardial ischemia-reperfusion injury and to examine its cardioprotective mechanisms. Isolated mouse hearts pre-treated with A-769662 had better recovery of left ventricular contractile function (55% vs. 29% of baseline rate-pressure product; p=0.03) and less myocardial necrosis (56% reduction in infarct size; p<0.01) during post-ischemic reperfusion compared to control hearts. Pre-treatment with A-769662 in vivo attenuated infarct size in C57Bl/6 mice undergoing left coronary artery occlusion and reperfusion compared to vehicle (36% vs. 18%, p=0.025). Mouse hearts with genetically inactivated AMPK were not protected by A-769662, indicating the specificity of this compound. Pre-treatment with A-769662 increased the phosphorylation and inactivation of eukaryotic elongation factor 2 (eEF2), preserved energy charge during ischemia, delayed the development of ischemic contracture, and reduced myocardial apoptosis and necrosis. A-769662 also augmented endothelial nitric oxide synthase (eNOS) activation during ischemia, which partially attenuated myocardial stunning, but did not prevent necrosis. AMPK is a therapeutic target that can be stimulated by a direct-acting small molecule in order to prevent injury during ischemia-reperfusion. The use of AMPK activators may represent a novel strategy to protect the heart and other solid organs against ischemia.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Cardiotônicos/farmacologia , Ativadores de Enzimas/farmacologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Pironas/farmacologia , Tiofenos/farmacologia , Proteínas Quinases Ativadas por AMP/genética , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Compostos de Bifenilo , Coração/fisiopatologia , Precondicionamento Isquêmico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Necrose , Óxido Nítrico Sintase Tipo III/metabolismo , Fator 2 de Elongação de Peptídeos/metabolismo
18.
J Immunol ; 183(6): 3712-9, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19710451

RESUMO

TLR3 is known to respond to dsRNA from viruses, apoptotic cells, and/or necrotic cells. Dying cells are a rich source of ligands that can activate TLRs, such as TLR3. TLR3 expressed in the liver is likely to be a mediator of innate activation and inflammation in the liver. The importance of this function of TLR3 during acute hepatitis has not previously been fully explored. We used the mouse model of Con A-induced hepatitis and observed a novel role for TLR3 in hepatocyte damage in the absence of an exogenous viral stimulus. Interestingly, TLR3 expression in liver mononuclear cells and sinus endothelial cells was up-regulated after Con A injection and TLR3(-/-) mice were protected from Con A-induced hepatitis. Moreover, splenocytes from TLR3(-/-) mice proliferated less to Con A stimulation in the presence of RNA derived from damaged liver tissue compared with wild-type (WT) mice. To determine the relative contribution of TLR3 expression by hematopoietic cells or nonhematopoietic to liver damage during Con A-induced hepatitis, we generated bone marrow chimeric mice. TLR3(-/-) mice engrafted with WT hematopoietic cells were protected in a similar manner to WT mice reconstituted with TLR3(-/-) bone marrow, indicating that TLR3 signaling in both nonhematopoietic and hematopoietic cells plays an important role in mediating liver damage. In summary, our data suggest that TLR3 signaling is necessary for Con A-induced liver damage in vivo and that TLR3 regulates inflammation and the adaptive T cell immune response in the absence of viral infection.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/patologia , Inflamação/etiologia , Receptor 3 Toll-Like/fisiologia , Doença Aguda , Animais , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Concanavalina A , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Hepatócitos/patologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Linfócitos T/imunologia , Receptor 3 Toll-Like/administração & dosagem , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/genética
19.
Oxid Med Cell Longev ; 2021: 6657944, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33791072

RESUMO

Long noncoding RNAs (lncRNAs) have attracted extensive attention due to their regulatory role in various cellular processes. Emerging studies have indicated that lncRNAs are expressed to varying degrees after the growth and development of the nervous system as well as injury and degeneration, thus affecting various physiological processes of the nervous system. In this review, we have compiled various reported lncRNAs related to the growth and development of central and peripheral nerves and pathophysiology (including advanced nerve centers, spinal cord, and peripheral nervous system) and explained how these lncRNAs play regulatory roles through their interactions with target-coding genes. We believe that a full understanding of the regulatory function of lncRNAs in the nervous system will contribute to understand the molecular mechanism of changes after nerve injury and will contribute to discover new diagnostic markers and therapeutic targets for nerve injury diseases.


Assuntos
Sistema Nervoso/metabolismo , RNA Longo não Codificante/genética , Animais , Diferenciação Celular/genética , Humanos , Regeneração Nervosa/genética , Sistema Nervoso/crescimento & desenvolvimento , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/patologia , Células-Tronco Neurais/metabolismo , RNA Longo não Codificante/metabolismo
20.
Pain ; 162(2): 600-608, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32833795

RESUMO

ABSTRACT: Musculoskeletal pain often occurs simultaneously at multiple anatomical sites. The aim of the study was to identify metabolic biomarkers for multisite musculoskeletal pain (MSMP) by metabolomics with an extreme phenotype sampling strategy. The study participants (n = 610) were derived from the Newfoundland Osteoarthritis Study. Musculoskeletal pain was assessed using a self-reported pain questionnaire where painful sites were circled on a manikin by participants and the total number of painful sites were calculated. Targeted metabolomic profiling on fasting plasma samples was performed using the Biocrates AbsoluteIDQ p180 kit. Plasma cytokine concentrations including tumor necrosis factor-α, interleukin-6, interleukin-1ß, and macrophage migration inhibitory factor were assessed by enzyme-linked immunosorbent assay. Data on blood cholesterol profiles were retrieved from participants' medical records. Demographic, anthropological, and clinical information was self-reported. The number of reported painful sites ranged between 0 and 21. Two hundred and five participants were included in the analysis comprising 83 who had ≥7 painful sites and 122 who had ≤1 painful site. Women and younger people were more likely to have MSMP (P ≤ 0.02). Multisite musculoskeletal pain was associated with a higher risk of having incontinence, worse functional status and longer period of pain, and higher levels of low-density lipoprotein and non-high-density lipoprotein cholesterol (all P ≤ 0.03). Among the 186 metabolites measured, 2 lysophosphatidylcholines, 1 with 26 carbons with no double bond and 1 with 28 carbons with 1 double bond, were significantly and positively associated with MSMP after adjusting for multiple testing with the Bonferroni method (P ≤ 0.0001) and could be considered as novel metabolic markers for MSMP.


Assuntos
Dor Musculoesquelética , Feminino , Humanos , Lisofosfatidilcolinas , Metabolômica , Medição da Dor , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA