Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 391
Filtrar
1.
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
2.
EMBO Mol Med ; 15(12): e17836, 2023 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-37766669

RESUMO

The epithelial-to-mesenchymal transition (EMT) plays a central role in the development of cancer metastasis and resistance to chemotherapy. However, its pharmacological treatment remains challenging. Here, we used an EMT-focused integrative functional genomic approach and identified an inverse association between short-chain fatty acids (propionate and butanoate) and EMT in non-small cell lung cancer (NSCLC) patients. Remarkably, treatment with propionate in vitro reinforced the epithelial transcriptional program promoting cell-to-cell contact and cell adhesion, while reducing the aggressive and chemo-resistant EMT phenotype in lung cancer cell lines. Propionate treatment also decreased the metastatic potential and limited lymph node spread in both nude mice and a genetic NSCLC mouse model. Further analysis revealed that chromatin remodeling through H3K27 acetylation (mediated by p300) is the mechanism underlying the shift toward an epithelial state upon propionate treatment. The results suggest that propionate administration has therapeutic potential in reducing NSCLC aggressiveness and warrants further clinical testing.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Animais , Camundongos , Neoplasias Pulmonares/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Propionatos/farmacologia , Propionatos/uso terapêutico , Camundongos Nus , Linhagem Celular Tumoral , Pulmão/metabolismo , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Movimento Celular
3.
Biomed Pharmacother ; 165: 115276, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37542852

RESUMO

Short-chain fatty acids (SCFAs) derived from the fermentation of carbohydrates by gut microbiota play a crucial role in regulating host physiology. Among them, acetate, propionate, and butyrate are key players in various biological processes. Recent research has revealed their significant functions in immune and inflammatory responses. For instance, butyrate reduces the development of interferon-gamma (IFN-γ) generating cells while promoting the development of regulatory T (Treg) cells. Propionate inhibits the initiation of a Th2 immune response by dendritic cells (DCs). Notably, SCFAs have an inhibitory impact on the polarization of M2 macrophages, emphasizing their immunomodulatory properties and potential for therapeutics. In animal models of asthma, both butyrate and propionate suppress the M2 polarization pathway, thus reducing allergic airway inflammation. Moreover, dysbiosis of gut microbiota leading to altered SCFA production has been implicated in prostate cancer progression. SCFAs trigger autophagy in cancer cells and promote M2 polarization in macrophages, accelerating tumor advancement. Manipulating microbiota- producing SCFAs holds promise for cancer treatment. Additionally, SCFAs enhance the expression of hypoxia-inducible factor 1 (HIF-1) by blocking histone deacetylase, resulting in increased production of antibacterial effectors and improved macrophage-mediated elimination of microorganisms. This highlights the antimicrobial potential of SCFAs and their role in host defense mechanisms. This comprehensive review provides an in-depth analysis of the latest research on the functional aspects and underlying mechanisms of SCFAs in relation to macrophage activities in a wide range of diseases, including infectious diseases and cancers. By elucidating the intricate interplay between SCFAs and macrophage functions, this review aims to contribute to the understanding of their therapeutic potential and pave the way for future interventions targeting SCFAs in disease management.


Assuntos
Microbioma Gastrointestinal , Propionatos , Masculino , Animais , Propionatos/uso terapêutico , Ácidos Graxos Voláteis/metabolismo , Butiratos/farmacologia , Butiratos/uso terapêutico , Inflamação/tratamento farmacológico , Microbioma Gastrointestinal/fisiologia , Macrófagos/metabolismo
4.
Microbes Infect ; 25(7): 105167, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37271368

RESUMO

Eradication of Helicobacter pylori, the class 1 carcinogen, faces several obstacles, which demand alternative options to conventional drug development methods. Alanine racemase (Alr) was proposed as H. pylori drug target, inhibited by propanoic acid (PA), in a previous in silico study. We investigated the possible treatment of H. pylori infection through Alr inhibition. A new model of H. pylori Alr was built, validated, and the binding of PA to the active site was modelled via molecular docking with a good docking score. PA minimum inhibitory concentration (MIC) against H. pylori ATCC 43504 and six H. pylori clinical isolates ranged from 312.5 to 416.7 ± 180 µg/ml and remained unchanged after 14 serial passages in increasing PA concentrations. The minimum bactericidal concentration of PA was 625 µg/ml. Selective Alr inhibition was confirmed by a significant PA MIC increase with increasing d-alanine concentrations. Similar PA MIC in other tested pathogens was recorded (312.5-625 µg/ml). PA lacked cytotoxicity in tested cell lines and efficiently eradicated H. pylori in a rat infection model. In conclusion, Alr is a promising broad-spectrum drug target, inhibited by PA without resistance development by repeated exposure for 14 serial passages.


Assuntos
Alanina Racemase , Infecções por Helicobacter , Helicobacter pylori , Ratos , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Alanina Racemase/química , Simulação de Acoplamento Molecular , Propionatos/uso terapêutico , Infecções por Helicobacter/tratamento farmacológico , Testes de Sensibilidade Microbiana
5.
J Allergy Clin Immunol ; 151(2): 447-457.e5, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36216081

RESUMO

BACKGROUND: Microbiota are recognized to play a major role in regulation of immunity through release of immunomodulatory metabolites such as short-chain fatty acids (SCFAs). Rhinoviruses (RVs) induce upper respiratory tract illnesses and precipitate exacerbations of asthma and chronic obstructive pulmonary disease through poorly understood mechanisms. Local interactions between SCFAs and antiviral immune responses in the respiratory tract have not been previously investigated. OBJECTIVE: We sought to investigate whether pulmonary metabolite manipulation through lung-delivered administration of SCFAs can modulate antiviral immunity to RV infection. METHODS: We studied the effects of intranasal administration of the SCFAs acetate, butyrate, and propionate on basal expression of antiviral signatures, and of acetate in a mouse model of RV infection and in RV-infected lung epithelial cell lines. We additionally assessed the effects of acetate, butyrate, and propionate on RV infection in differentiated human primary bronchial epithelial cells. RESULTS: Intranasal acetate administration induced basal upregulation of IFN-ß, an effect not observed with other SCFAs. Butyrate induced RIG-I expression. Intranasal acetate treatment of mice increased interferon-stimulated gene and IFN-λ expression during RV infection and reduced lung virus loads at 8 hours postinfection. Acetate ameliorated virus-induced proinflammatory responses with attenuated pulmonary mucin and IL-6 expression observed at day 4 and 6 postinfection. This interferon-enhancing effect of acetate was confirmed in human bronchial and alveolar epithelial cell lines. In differentiated primary bronchial epithelial cells, butyrate treatment better modulated IFN-ß and IFN-λ gene expression during RV infection. CONCLUSIONS: SCFAs augment antiviral immunity and reduce virus load and proinflammatory responses during RV infection.


Assuntos
Infecções por Enterovirus , Infecções por Picornaviridae , Humanos , Camundongos , Animais , Antivirais/uso terapêutico , Rhinovirus , Propionatos/farmacologia , Propionatos/uso terapêutico , Interferons , Brônquios , Células Epiteliais , Acetatos/farmacologia , Acetatos/uso terapêutico , Butiratos/farmacologia , Butiratos/uso terapêutico
6.
Acta Trop ; 234: 106571, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35752205

RESUMO

Neurocysticercosis (NCC) is an endemic public health disease of the central nervous system highly related to epilepsy and seizures. Taenia crassiceps is an experimental model used for NCC and biochemical studies of the host-parasite relationship. For the past 50 years the NCC therapeutic treatment is performed with albendazole (ABZ) and praziquantel which opens a gap for new therapies due to parasitic resistance and other adverse effects of the drugs. Oxfendazole (OXF) is an albendazole derivative with efficacy against tissue cestodes of veterinary importance. The aim of this study was to determine the metabolic impact of OXF on T. crassiceps cysticerci intracranially inoculated in Balb/C mice. The animals were intracranially inoculated with T. crassiceps cysticerci and 30 days later received single dose oral treatment of OXF, ABZ and NaCl 0.9% (control group). The metabolic impact was quantified through the detection of metabolites from glycolysis, anaerobic fermentation of lactate and propionate, tricarboxylic acid cycle, protein catabolism, fatty acids oxidation. The differences observed in the concentrations of metabolites from the OXF treated group showed that the drug induced gluconeogenesis, increase in protein catabolism, fatty acids oxidation and propionate fermentation in comparison to the ABZ and control treated groups. In conclusion, OXF induced greater metabolic impact in T. crassiceps cysticerci than the standard NCC treatment, ABZ, showing that it may represent an alternative drug for its treatment.


Assuntos
Anti-Helmínticos , Neurocisticercose , Taenia , Albendazol/uso terapêutico , Animais , Anti-Helmínticos/uso terapêutico , Benzimidazóis , Cysticercus , Gluconeogênese , Camundongos , Camundongos Endogâmicos BALB C , Neurocisticercose/tratamento farmacológico , Neurocisticercose/parasitologia , Propionatos/metabolismo , Propionatos/farmacologia , Propionatos/uso terapêutico
7.
Biol Pharm Bull ; 44(9): 1247-1253, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34471053

RESUMO

The rate of glycolysis in cancer cells is higher than that of normal cells owing to high energy demands, which results in the production of excess lactate. Monocarboxylate transporters (MCTs), especially MCT1 and MCT4, play a critical role in maintaining an appropriate pH environment through lactate transport, and their high expression is associated with poor prognosis in breast cancer. Thus, we hypothesized that inhibition of MCTs is a promising therapeutic target for adjuvant breast cancer treatment. We investigated the effect of MCT inhibition in combination with 4-hydroxytamoxifen (4-OHT), an active metabolite of tamoxifen, using two estrogen receptor (ER)-positive breast cancer cell lines, MCF-7 and T47D. Lactate transport was investigated in cellular uptake studies. The cytotoxicity of 4-OHT was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. In both cell lines evaluated, MCT1 and MCT4 were constitutively expressed at the mRNA and protein levels. [14C]-L-lactate uptake by both cells was significantly inhibited by bindarit, a selective MCT4 inhibitor, but weakly affected by 5-oxoploline (5-OP), a selective MCT1 inhibitor. The results of the MTT assay showed that combination with bindarit, but not 5-OP, decreased 4-OHT sensitivity. Bindarit significantly increased the levels of hypoxia-inducible factor-1α (HIF-1α) in MCF-7 cells. Moreover, HIF-1α knockdown significantly increased 4-OHT sensitivity, whereas induction of HIF-1α by hypoxia decreased 4-OHT sensitivity in MCF-7 cells. In conclusion, pharmacological MCT4 inhibition confers resistance to 4-OHT rather than sensitivity, by increasing HIF-1α protein levels. In addition, HIF-1α inhibition represents a potential therapeutic strategy for enhancing 4-OHT sensitivity.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Transportadores de Ácidos Monocarboxílicos/antagonistas & inibidores , Proteínas Musculares/antagonistas & inibidores , Tamoxifeno/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Indazóis/farmacologia , Indazóis/uso terapêutico , Células MCF-7 , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Propionatos/farmacologia , Propionatos/uso terapêutico , Receptores de Estrogênio/análise , Receptores de Estrogênio/metabolismo , Tamoxifeno/farmacologia , Tamoxifeno/uso terapêutico
8.
J Med Chem ; 64(18): 13410-13428, 2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34499493

RESUMO

Retinoic acid receptor-related orphan receptor γ (RORc, RORγ, or NR1F3) is the nuclear receptor master transcription factor that drives the function and development of IL-17-producing T helper cells (Th17), cytotoxic T cells (Tc17), and subsets of innate lymphoid cells. Activation of RORγ+ T cells in the tumor microenvironment is hypothesized to render immune infiltrates more effective at countering tumor growth. To test this hypothesis, a family of benzoxazines was optimized to provide LYC-55716 (37c), a potent, selective, and orally bioavailable small-molecule RORγ agonist. LYC-55716 decreases tumor growth and enhances survival in preclinical tumor models and was nominated as a clinical development candidate for evaluation in patients with solid tumors.


Assuntos
Antineoplásicos/uso terapêutico , Benzoxazinas/uso terapêutico , Neoplasias/tratamento farmacológico , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/agonistas , Propionatos/uso terapêutico , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacocinética , Benzoxazinas/síntese química , Benzoxazinas/farmacocinética , Feminino , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , Estrutura Molecular , Propionatos/síntese química , Propionatos/farmacocinética , Ratos Sprague-Dawley , Relação Estrutura-Atividade
9.
Curr Pain Headache Rep ; 25(5): 29, 2021 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-33761014

RESUMO

PURPOSE OF REVIEW: Dual enkephalinase inhibitors (DENKIs) are pain medications that indirectly activate opioid receptors and can be used as an alternative to traditional opioids. Understanding the physiology of enkephalins and their inhibitors and the pharmacology of these drugs will allow for proper clinical application for chronic pain patients in the future. RECENT FINDINGS: DENKIs can be used as an alternative mode of analgesia for patients suffering from chronic pain by preventing the degradation of endogenous opioid ligands. By inhibiting the two major enkephalin-degrading enzymes (neprilysin and aminopeptidase N), DENKIs can provide analgesia with less adverse effects than nonendogenous opioids. The purpose of this paper is to review the current literature investigating DENKIs and explore their contribution to chronic pain management.


Assuntos
Antígenos CD13/antagonistas & inibidores , Dor Crônica/tratamento farmacológico , Dissulfetos/uso terapêutico , Encefalinas/metabolismo , Inibidores Enzimáticos/uso terapêutico , Neprilisina/antagonistas & inibidores , Propionatos/uso terapêutico , Propilaminas/uso terapêutico , Dipeptídeos/uso terapêutico , Humanos , Fenilalanina/análogos & derivados , Fenilalanina/uso terapêutico
10.
Medicine (Baltimore) ; 100(12): e24884, 2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33761646

RESUMO

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is a highly prevalent chronic liver disease characterized by excess accumulation of fat in hepatocytes. Because no drug has been approved for NAFLD treatment, this work analyzed the effects of agents resulting from 2 research hotspots, metabolic target agents, and natural plant drugs, on NAFLD with network meta-analysis. METHODS: Public databases were searched through August 14, 2020. Randomized controlled trials that compared obeticholic acid, elafibranor, cenicriviroc, selonsertib, curcumin, silymarin, and resveratrol to placebo were included. Liver pathology improvement, hepatic biochemical indicators, and lipid metabolism indicators were analyzed. RESULTS: Thirty-five studies were included in the meta-analysis. Obeticholic acid was found to significantly increase the frequency of liver biopsy improvement compared to placebo (OR: 2.10; 95% CI: 1.60, 2.77). The ranking results among the hepatic biochemical indicators showed that obeticholic acid (94.9%) and elafibranor (86.3%) have a relative advantage in reducing alanine aminotransferase (ALT) levels, and obeticholic acid also had an advantage (95.4%) in reducing aspartate aminotransferase (AST) levels. Considering lipid metabolic indicators, elafibranor (expSMD: 0.01; 95% CI: 0.00, 0.05; SUCRA: 100%), and obeticholic acid (expSMD: 0.48; 95% CI: 0.28,0.84; SUCRA: 75.6%) significantly reduced triglyceride (TG) levels compared with placebo; moreover, obeticholic acid, but not elafibranor, caused a serious increase in total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) levels and a decrease in high-density lipoprotein cholesterol (HDL-C) levels. CONCLUSIONS: Novel metabolic targeted agents generally have better effects than natural plant drugs, especially obeticholic acid, and elafibranor. However, obeticholic acid showed serious adverse effects such as increasing LDL-C levels and decreasing HDL-C levels. Curcumin showed potential advantages for NAFLD but lacked statistical significance.


Assuntos
Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Preparações de Plantas/uso terapêutico , Alanina Transaminase/sangue , Aspartato Aminotransferases/sangue , Chalconas/uso terapêutico , Ácido Quenodesoxicólico/efeitos adversos , Ácido Quenodesoxicólico/análogos & derivados , Ácido Quenodesoxicólico/uso terapêutico , HDL-Colesterol/sangue , HDL-Colesterol/efeitos dos fármacos , LDL-Colesterol/sangue , LDL-Colesterol/efeitos dos fármacos , Curcumina/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Humanos , Metanálise em Rede , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/enzimologia , Propionatos/uso terapêutico , Triglicerídeos/sangue
11.
J Pharmacokinet Pharmacodyn ; 48(4): 525-541, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33728547

RESUMO

Verinurad, a uric acid transporter 1 (URAT1) inhibitor, lowers serum uric acid by promoting its urinary excretion. Co-administration with a xanthine oxidase inhibitor (XOI) to simultaneously reduce uric acid production rate reduces the potential for renal tubular precipitation of uric acid, which can lead to acute kidney injury. The combination is currently in development for chronic kidney disease and heart failure. The aim of this work was to apply and extend a previously developed semi-mechanistic exposure-response model for uric acid kinetics to include between-subject variability to verinurad and its combinations with XOIs, and to provide predictions to support future treatment strategies. The model was developed using data from 12 clinical studies from a total of 434 individuals, including healthy volunteers, patients with hyperuricemia, and renally impaired subjects. The model described the data well, taking into account the impact of various patient characteristics such as renal function, baseline fractional excretion of uric acid, and race. The potencies (EC50s) of verinurad (reducing uric acid reuptake), febuxostat (reducing uric acid production), and oxypurinol (reducing uric acid production) were: 29, 128, and 13,030 ng/mL, respectively. For verinurad, symptomatic hyperuricemic (gout) subjects showed a higher EC50 compared with healthy volunteers (37 ng/mL versus 29 ng/mL); while no significant difference was found for asymptomatic hyperuricemic patients. Simulations based on the uric acid model were performed to assess dose-response of verinurad in combination with XOI, and to investigate the impact of covariates. The simulations demonstrated application of the model to support dose selection for verinurad.


Assuntos
Hiperuricemia/tratamento farmacológico , Naftalenos/uso terapêutico , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Propionatos/uso terapêutico , Piridinas/uso terapêutico , Ácido Úrico/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Quimioterapia Combinada , Feminino , Humanos , Hiperuricemia/sangue , Hiperuricemia/urina , Rim/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Modelos Estatísticos , Naftalenos/administração & dosagem , Naftalenos/farmacologia , Propionatos/administração & dosagem , Propionatos/farmacologia , Piridinas/administração & dosagem , Piridinas/farmacologia , Ácido Úrico/urina , Xantina Oxidase/antagonistas & inibidores , Adulto Jovem
12.
Nutrients ; 13(2)2021 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-33572661

RESUMO

We are currently facing an obesity pandemic, with worldwide obesity rates having tripled since 1975. Obesity is one of the main risk factors for the development of non-communicable diseases, which are now the leading cause of death worldwide. This calls for urgent action towards understanding the underlying mechanisms behind the development of obesity as well as developing more effective treatments and interventions. Appetite is carefully regulated in humans via the interaction between the central nervous system and peripheral hormones. This involves a delicate balance in external stimuli, circulating satiating and appetite stimulating hormones, and correct functioning of neuronal signals. Any changes in this equilibrium can lead to an imbalance in energy intake versus expenditure, which often leads to overeating, and potentially weight gain resulting in overweight or obesity. Several lines of research have shown imbalances in gut hormones are found in those who are overweight or obese, which may be contributing to their condition. Therefore, this review examines the evidence for targeting gut hormones in the treatment of obesity by discussing how their dysregulation influences food intake, the potential possibility of altering the circulating levels of these hormones for treating obesity, as well as the role of short chain fatty acids and protein as novel treatments.


Assuntos
Regulação do Apetite/fisiologia , Ácidos Graxos Voláteis/uso terapêutico , Hormônios Gastrointestinais/metabolismo , Obesidade/terapia , Ácido Acético/uso terapêutico , Animais , Apetite/fisiologia , Butiratos/uso terapêutico , Sistema Nervoso Central/fisiologia , Colecistocinina/metabolismo , Dipeptídeos/metabolismo , Dipeptídeos/uso terapêutico , Ingestão de Energia/fisiologia , Metabolismo Energético/fisiologia , Hormônios Gastrointestinais/sangue , Trato Gastrointestinal/fisiologia , Grelina/metabolismo , Peptídeo 1 Semelhante ao Glucagon/agonistas , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/uso terapêutico , Humanos , Hiperfagia/etiologia , Camundongos , Neuropeptídeo Y/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Sobrepeso/etiologia , Sobrepeso/metabolismo , Oxintomodulina/metabolismo , Oxintomodulina/uso terapêutico , Polipeptídeo Pancreático/metabolismo , Propionatos/uso terapêutico , Saciação/fisiologia
13.
Medicina (Kaunas) ; 57(1)2021 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-33435164

RESUMO

This article aims to critically review the evidence on the available therapeutic strategies for the treatment of hyperuricemia. For this reason, several papers were reviewed. Xanthine oxidase inhibitors are the safest and most effective uric acid lowering drugs for the management of chronic hyperuricemia, while the efficacy of uricosuric agents is strongly modulated by pharmacogenetics. Emergent drugs (lesinurad, peglotidase) were found to be more effective for the acute management of refractory hyperuricemia, but their use is supported by a relatively small number of clinical trials so that further well-designed clinical research is needed to deepen their efficacy and safety profile.


Assuntos
Hiperuricemia/tratamento farmacológico , Uricosúricos/uso terapêutico , Xantina Oxidase/antagonistas & inibidores , Acetamidas/uso terapêutico , Alopurinol/uso terapêutico , Benzobromarona/uso terapêutico , Doença Crônica , Medicina Baseada em Evidências , Febuxostat/uso terapêutico , Supressores da Gota/uso terapêutico , Humanos , Naftalenos/uso terapêutico , Nitrilas/uso terapêutico , Fenilacetatos/uso terapêutico , Polietilenoglicóis/uso terapêutico , Probenecid/uso terapêutico , Propionatos/uso terapêutico , Piridinas/uso terapêutico , Tioglicolatos/uso terapêutico , Triazóis/uso terapêutico , Urato Oxidase/uso terapêutico
14.
Acta Pharmacol Sin ; 42(8): 1311-1323, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32973326

RESUMO

Propionate is a short-chain fatty acid (SCFA) mainly produced from carbohydrates by gut microbiota. Sodium propionate (SP) has shown to suppress the invasion in G protein-coupled receptor 41 (GPR41) and GPR43-overexpressing breast cancer cells. In this study we investigated the effects of SP on the proliferation, apoptosis, autophagy, and antioxidant production of breast cancer cells. We showed that SP (5-20 mM) dose-dependently inhibited proliferation and induced apoptosis in breast cancer cell lines JIMT-1 (ER-negative and HER2-expressing) and MCF7 (ER-positive type), and this effect was not affected by PTX, thus not mediated by the GPR41 or GPR43 SCFA receptors. Meanwhile, we demonstrated that SP treatment increased autophagic and antioxidant activity in JIMT-1 and MCF7 breast cancer cells, which might be a compensatory mechanism to overcome SP-induced apoptosis, but were not sufficient to overcome SP-mediated suppression of proliferation and induction of apoptosis. We revealed that the anticancer effect of SP was mediated by inhibiting JAK2/STAT3 signaling which led to cell-cycle arrest at G0/G1 phase, and increasing levels of ROS and phosphorylation of p38 MAPK which induced apoptosis. In nude mice bearing JIMT-1 and MCF7 cells xenograft, administration of SP (20 mg/mL in drinking water) significantly suppressed tumor growth by regulating STAT3 and p38 in tumor tissues. These results suggest that SP suppresses proliferation and induces apoptosis in breast cancer cells by inhibiting STAT3, increasing the ROS level and activating p38. Therefore, SP is a candidate therapeutic agent for breast cancer.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Propionatos/uso terapêutico , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Humanos , Camundongos Nus , Propionatos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Antiviral Res ; 185: 104985, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33271272

RESUMO

APE1 is a multi-functional protein with a redox function in its N-terminal domain and an apurinic/apyrimidinic endonuclease activity in the C-terminal domain. APE1 redox function plays an important role in regulating cell proliferation and survival through activating specific transcriptional activators. APE1 redox function is also found to be associated with some cancer occurrence. In this study, we demonstrated that APE1 redox function is essential for Epstein-Barr virus (EBV) lytic replication as the silencing of APE1 expression or treatment with APE1 redox inhibitors C10 and E3330 can inhibit EBV lytic replication and virion production. Furthermore, C10 and E3330 also inhibit MHV-68 replication in vitro and in vivo. C10 and E3330 were able to significantly reduce the loss of pulmonary alveoli and thickening of alveolar septa in mice caused by MHV-68 infection. Altogether, (i) APE1 redox function is validated as a new antiviral target; (ii) APE1 redox inhibitors, especially C10, have potentials to be used for the treatment of γ-herpesvirus infection and associated diseases; (iii) MHV-68 is validated to be a surrogate for the study of the pathogenesis and therapy of EBV and KSHV infection in vivo.


Assuntos
DNA Liase (Sítios Apurínicos ou Apirimidínicos)/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Herpesviridae/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Benzoquinonas/farmacologia , Benzoquinonas/uso terapêutico , Linhagem Celular Tumoral , Feminino , Células HEK293 , Herpesviridae/classificação , Herpesviridae/patogenicidade , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/virologia , Humanos , Pulmão/efeitos dos fármacos , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Oxirredução , Propionatos/farmacologia , Propionatos/uso terapêutico
16.
Mol Hum Reprod ; 26(6): 452-468, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32236411

RESUMO

Spontaneous preterm birth is a global health issue affecting up to 20% of pregnancies and leaves a legacy of neurodevelopmental complications. Inflammation has been implicated in a significant proportion of preterm births, where pro-inflammatory insults trigger production of additional pro-inflammatory and pro-labor mediators. Thus, novel therapeutics that can target inflammation may be a novel avenue for preventing preterm birth and improving adverse fetal outcomes. Short-chain fatty acids (SCFAs), such as butyrate and propionate, are dietary metabolites produced by bacterial fermentation of fiber in the gut. SCFAs are known to possess anti-inflammatory properties and have been found to function through G-coupled-receptors and histone deacetylases. Therefore, this study aimed to investigate the effect of SCFAs on pro-inflammatory and pro-labor mediators in an in vitro model of preterm birth. Primary human cells isolated from myometrium and fetal membranes (decidua, amnion mesenchymal and amnion epithelial cells) were stimulated with the pro-inflammatory cytokines tumor necrosis factor alpha (TNF) or interleukin 1B (IL1B). The SCFAs butyrate and propionate suppressed inflammation-induced expression of pro-inflammatory cytokines and chemokines, adhesion molecules, the uterotonic prostaglandin PGF2alpha and enzymes involved in remodeling of myometrium and degradation of the fetal membranes. Notably, propionate and butyrate also suppressed inflammation-induced prostaglandin signaling and myometrial cell contraction. These effects appear to be mediated through suppression of nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) activation. These results suggest that the SCFAs may be able to prevent myometrial contractions and rupture of membranes. Further in vivo studies are warranted to identify the efficacy of SCFAs as a novel anti-inflammatory therapeutic to prevent inflammation-induced spontaneous preterm birth.


Assuntos
Butiratos/farmacologia , Butiratos/uso terapêutico , Inflamação/tratamento farmacológico , Miométrio/metabolismo , Propionatos/farmacologia , Propionatos/uso terapêutico , Animais , Ácidos Graxos Voláteis/metabolismo , Feminino , Histona Desacetilases/metabolismo , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miométrio/efeitos dos fármacos , NF-kappa B/metabolismo , Gravidez , Nascimento Prematuro
17.
Radiology ; 296(1): 67-75, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32343209

RESUMO

Background Liver biopsy is the reference standard to diagnose nonalcoholic steatohepatitis (NASH) but is invasive with potential complications. Purpose To evaluate molecular MRI with type 1 collagen-specific probe EP-3533 and allysine-targeted fibrogenesis probe Gd-Hyd, MR elastography, and native T1 to characterize fibrosis and to assess treatment response in a rat model of NASH. Materials and Methods MRI was performed prospectively (June-November 2018) in six groups of male Wistar rats (a) age- and (b) weight-matched animals received standard chow (n = 12 per group); (c) received choline-deficient, l-amino acid-defined, high-fat diet (CDAHFD) for 6 weeks or (d) 9 weeks (n = 8 per group); (e) were fed 6 weeks of CDAHFD and switched to standard chow for 3 weeks (n = 12); (f) were fed CDAHFD for 9 weeks with daily treatment of elafibranor beginning at week 6 (n = 14). Differences in imaging measurements and tissue analyses among groups were tested with one-way analysis of variance. The ability of each imaging measurement to stage fibrosis was quantified by using area under the receiver operating characteristic curve (AUC) with quantitative digital pathology (collagen proportionate area [CPA]) as reference standard. Optimal cutoff values for distinguishing advanced fibrosis were used to assess treatment response. Results AUC for distinguishing fibrotic (CPA >4.8%) from nonfibrotic (CPA ≤4.8%) livers was 0.95 (95% confidence interval [CI]: 0.91, 1.00) for EP-3533, followed by native T1, Gd-Hyd, and MR elastography with AUCs of 0.90 (95% CI: 0.83, 0.98), 0.84 (95% CI: 0.74, 0.95), and 0.65 (95% CI: 0.51, 0.79), respectively. AUCs for discriminating advanced fibrosis (CPA >10.3%) were 0.86 (95% CI: 0.76, 0.97), 0.96 (95% CI: 0.90, 1.01), 0.84 (95% CI: 0.70, 0.98), and 0.74 (95% CI: 0.63, 0.86) for EP-3533, Gd-Hyd, MR elastography, and native T1, respectively. Gd-Hyd MRI had the highest accuracy (24 of 26, 92%; 95% CI: 75%, 99%) in identifying responders and nonresponders in the treated groups compared with MR elastography (23 of 26, 88%; 95% CI: 70%, 98%), EP-3533 (20 of 26, 77%; 95% CI: 56%, 91%), and native T1 (14 of 26, 54%; 95% CI: 33%, 73%). Conclusion Collagen-targeted molecular MRI most accurately detected early onset of fibrosis, whereas the fibrogenesis probe Gd-Hyd proved most accurate for detecting treatment response. © RSNA, 2020 Online supplemental material is available for this article.


Assuntos
Cirrose Hepática/diagnóstico por imagem , Cirrose Hepática/terapia , Imageamento por Ressonância Magnética/métodos , Hepatopatia Gordurosa não Alcoólica/diagnóstico por imagem , Hepatopatia Gordurosa não Alcoólica/terapia , Animais , Chalconas/uso terapêutico , Dieta/métodos , Modelos Animais de Doenças , Fígado/diagnóstico por imagem , Cirrose Hepática/etiologia , Masculino , Hepatopatia Gordurosa não Alcoólica/complicações , Propionatos/uso terapêutico , Estudos Prospectivos , Ratos , Ratos Wistar
18.
J Endocrinol ; 245(3): 425-437, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32302970

RESUMO

This study aimed to assess the effects of three major SCFAs (acetate, propionate, and butyrate) on NASH phenotype in mice. C57BL/6 mice were fed a methionine- and choline-deficient (MCD) diet and treated with sodium acetate, sodium propionate, or sodium butyrate during the 6-week feeding period. SCFA treatment significantly reduced serum levels of alanine aminotransferase and aspartate transaminase, the numbers of lipid droplets, and the levels of triglycerides and cholesterols in livers of the mice compared with control treatment. SCFAs also reduced MCD-induced hepatic aggregation of macrophages and proinflammatory responses. Among the three SCFAs, sodium acetate (NaA) revealed the best efficacy at alleviating MCD-induced hepatic steatosis and inflammation. Additionally, NaA increased AMP-activated protein kinase activation in the liver and induced the expression of fatty acid oxidation gene in both the liver and cultured hepatocytes. In vitro, NaA decreased MCD-mimicking media-induced proinflammatory responses in macrophages to a greater extent than in hepatocytes. These results indicated that NaA alleviates steatosis in a manner involving AMPK activation. Also, NaA alleviation of hepatic inflammation appears to be due to, in large part, suppression of macrophage proinflammatory activation. SCFAs may represent as a novel and viable approach for alleviating NASH.


Assuntos
Acetatos/uso terapêutico , Butiratos/uso terapêutico , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Propionatos/uso terapêutico , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Ácido Butírico/uso terapêutico , Ácidos Graxos Voláteis/metabolismo , Fígado Gorduroso/sangue , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Inflamação/sangue , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/metabolismo , Acetato de Sódio/uso terapêutico
19.
Breast Cancer ; 27(4): 696-705, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32095987

RESUMO

INTRODUCTION: Short-chain fatty acids (SCFAs) are ubiquitous lipids produced as a result of bacterial fermentation of dietary fiber. While their role in colorectal cancer is well known, the effect of SCFAs in breast cancer is poorly defined. OBJECTIVE: To understand the various effects of SCFAs on breast carcinogenesis, we investigated the effect of sodium butyrate (NaB) and sodium propionate (NaP) in MCF-7 cell line. MATERIALS AND METHODS: Cells were incubated with different concentrations of NaB or NaP for 24, 48, 72 or 96 h. Cell proliferation was assayed using MTT kit. Cell cycle was performed using propidium iodide staining then analyzed with a flow cytometer. Apoptosis was assessed by Hoechst technique and cell-cycle sub-G1 phase. RESULTS: NaB and NaP inhibited MCF-7 cell proliferation in a dose-dependent manner with respective IC50 of 1.26 mM and 4.5 mM, thus indicating that NaB is more potent than NaP. Low and medium levels of both SCFAs induced morphology changes which are characteristic of a differentiated phenotype. Flow cytometry analysis revealed a blockage in G1 growth phase. Interestingly, removing NaB or NaP from culture media after few days of treatment showed a reversible effect on cell morphology and proliferation where cells reentered the cycle after 24 h of drug wash-out. Finally, treatment with medium levels of these molecules induced low MCF-7 apoptosis, while higher doses led to massive apoptosis. CONCLUSION: Our results show that SCFAs may be considered as an interesting inhibitor for breast cancer progression.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Ácido Butírico/farmacologia , Carcinogênese/efeitos dos fármacos , Propionatos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Ácido Butírico/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Progressão da Doença , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Células MCF-7 , Propionatos/uso terapêutico
20.
Neuropharmacology ; 166: 107977, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32004548

RESUMO

Sodium propionate (SP) is one of the main short chain fatty acids (SCFA) that can be produced naturally through host metabolic pathways. SP have been documented and include the reduction of pro-inflammatory mediators in an in vivo model of colitis. The aim of this study is to evaluate the neuroprotective effects of SP in reducing inflammatory process associated to neurological disorders. We performed both in vitro model of Alzheimer's disease, induced by oligomeric Aß1-42 stimulation, and in in vivo model of spinal cord injury (SCI) in which neuroinflammation plays a crucial role. For in vitro model, the human neuroblastoma SH-SY5Y cell line was first differentiated with retinoic acid (100 µM) for 24 h and then stimulated by oligomeric Aß1-42 (1 µg/ml) and treated with SP at 0.1- 1-10 µM concentrations for another 24 h. Instead, the in vivo model of SCI was induced by extradural compression of the spinal cord at T6-T8 levels, and animals were treated with SP (10-30-100 mg/kg o.s) 1 and 6 h after SCI. Our results demonstrated that both in in vitro neuroinflammatory model and in vivo model of SCI the treatment with SP significantly reduced NF-κB nuclear translocation and IκBα degradation, as well as decreases COX-2 and iNOS expressions evaluated by Western blot analysis. Moreover, we showed that SP treatment significantly ameliorated histopathology changes and improved motor recovery in a dose-dependent manner. In conclusion, our results demonstrated that SP possesses neuroprotective effects, suggesting it could represent a target for therapeutic intervention in neuroinflammatory disorders.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Fármacos Neuroprotetores/uso terapêutico , Fragmentos de Peptídeos/toxicidade , Propionatos/uso terapêutico , Traumatismos da Medula Espinal/induzido quimicamente , Traumatismos da Medula Espinal/prevenção & controle , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Masculino , Camundongos , Distribuição Aleatória , Traumatismos da Medula Espinal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA