Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Molecules ; 28(18)2023 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-37764501

RESUMO

(1) Background: Shikonin, the main ingredient in Chinese herbal medicine, is described as a novel angiogenesis inhibitor, and its anticancer effects have already been studied. Shikonin and its derivatives induce apoptosis and suppress metastasis, which further enhance the effectiveness of chemotherapy. However, their mechanism of function has not been completely elucidated on human renal cancer cells. (2) Methods: In our study, CAKI-2 and A-498 cells were treated with increasing concentrations (2.5-40 µM) of shikonin, when colony formation ability and cytotoxic activity were tested. The changes in the expression of the main targets of apoptotic pathways were measured by RT-qPCR and Western blot. The intracellular levels of miR-21 and miR-155 were quantified by RT-qPCR. (3) Results: Shikonin exerted a dose-dependent effect on the proliferation of the cell lines examined. In 5 µM concentration of shikonin in vitro elevated caspase-3 and -7 levels, the proteins of the Ras/MAPK and PI3K/AKT pathways were activated. However, no significant changes were detected in the miR-21 and miR-155 expressions. (4) Conclusions: Our findings indicated that shikonin causes apoptosis of renal cancer cells by activating the Ras/MAPK and PI3K/AKT pathways. These effects of shikonin on renal cancer cells may bear important potential therapeutic implications for the treatment of renal cancer.

2.
Int J Obes (Lond) ; 46(5): 969-976, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35058572

RESUMO

BACKGROUND: Long non-coding RNAs (lncRNAs) have emerged as a rapidly expanding area of interest in chronic diseases. They are mostly unknown for roles in metabolic regulation. Sirtuins, an epigenetic modulator class, regulate metabolic pathways. However, how sirtuins are regulated via lncRNA is unknown. We hypothesized that a high-fat high-fructose diet (HFD-HF) during pregnancy would increase the risk for obesity via lncRNA-Sirtuin pathways. METHODS: Female C57Bl/6 mice (F0) were fed either chow diet (CD) or HFD-HF for 6 weeks till birth. The pups (F1) were fed either CD or HFD-HF for 20 weeks. Expression of Dleu2, sirtuins, mitochondrial respiratory complexes, and oxidative stress were investigated in the F1 livers. Fasting blood glucose, insulin sensitivity, glucose tolerance, body and tissues weight were measured. A mechanistic interaction was then carried out using a DLEU2 knockdown experiment in the HepG2 cell. RESULTS: Dleu2 and sirtuins were both significantly decreased in the livers of HFD-HF fed male F1 whose mothers were either fed CD or HFD-HF during reproductive and pregnancy windows. Confirming this connection, upon silencing DLEU2, transcription levels of SIRT1 through 6 and translational levels of SIRT1, 3, 5, and 6 were significantly downregulated. Knockdown of DLEU2 significantly decreased the protein level of cytochrome-c oxidase (complex IV, MTCO1) without altering other mitochondrial complexes, decreased mitochondrial membrane potential, decreased ATP, and increased reactive oxygen species. Interestingly, in F1 livers, the protein level of MTCO1 was also significantly decreased under an HFD-HF diet or even under chow diet if the mother was exposed to HFD-HF. CONCLUSION: Our findings reveal for the first time that one lncRNA can regulate sirtuins and a specific mitochondrial complex. Furthermore, diet or maternal diet can modulate Dleu2 and its downstream regulators in offspring, suggesting a potential role of DLEU2 in metabolic disorders over one or more generations.


Assuntos
RNA Longo não Codificante , Sirtuínas , Animais , Dieta Hiperlipídica , Transporte de Elétrons , Feminino , Frutose , Masculino , Camundongos , Obesidade/metabolismo , Gravidez , RNA Longo não Codificante/genética , Sirtuína 1/metabolismo , Sirtuínas/metabolismo , Transferases
3.
Biochem J ; 478(7): 1359-1375, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33861844

RESUMO

Atherosclerosis is a chronic inflammatory disease associated with lipid metabolism disorder. Autophagy is a catabolic process and contributes to maintaining cellular homeostasis. Substantial evidence suggests that defective autophagy is implicated in several diseases, including atherosclerosis, while increased autophagy mitigates atherosclerosis development. Thus, understanding the mechanisms of autophagy regulation and its association with atherosclerosis is vital to develop new therapies against atherosclerosis. Dietary bioactive compounds are non-nutrient natural compounds that include phenolics, flavonoids, and carotenoids. Importantly, these bioactive compounds possess anti-inflammatory, antioxidant, and antibacterial properties that may alleviate various chronic diseases. Recently, examining the effects of bioactive compounds on autophagy activity in atherogenesis has drawn considerable attention. The current review discusses the role of macrophage autophagy in the development and progression of atherosclerosis. We also summarize our current knowledge of the therapeutic potential of bioactive compounds on atherosclerosis and autophagy.


Assuntos
Aterosclerose/tratamento farmacológico , Autofagia , Produtos Biológicos/farmacologia , Macrófagos/patologia , Animais , Aterosclerose/patologia , Humanos
4.
Chem Res Toxicol ; 34(1): 54-62, 2021 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-33395283

RESUMO

Exposure to endocrine-disrupting chemicals used in plastic manufacturing may contribute to the current obesity and diabetes epidemic. Our previous study demonstrated that benzyl butyl phthalate (BBP) induced adipogenesis in the C3H10T1/2 stem cell line. Here we investigated if BBP deregulated long noncoding RNA H19 and its downstream pathway and whether BBP plays a role in the insulin signaling pathway during adipocyte diiferentiation. Cells treated with an 8 day BBP regimen showed that H19 expression was decreased at day 2 with 50 µM BBP exposure (p < 0.05). However, no significant changes were observed from day 4 to day 8. Expression of miRNA-103/107, H19 regulated miRNAs, was upregulated at day 2 (p < 0.05) but not from day 4 to day 8. Similarly, expression of the let-7 family members (a, b, c, d, f, and g) was also significantly increased at day 2 (p < 0.05 or p < 0.01), except for let-7e. Both let-7 and miRNA-103/107 are targets of H19 and play roles in insulin signaling. Insulin receptor substrate (IRS)-1, one of the key insulin signal transduction regulators, was significantly downregulated from day 2 to day 8 (p < 0.05). Gene expression of insulin receptor (IR) and IRS-2 were not altered by BBP exposure. The ratio of IRS1/IRS2 was significantly decreased from day 2 to day 8. On day 4, phospho-Akt protein expression was significantly decreased (p < 0.05). In conclusion, BBP exposure may lead to metabolic dysregulation by altering vital epigenetic regulators such as lncRNA H19 and its target microRNAs at an earlier stage, which further regulates insulin signaling.


Assuntos
Ácidos Ftálicos/farmacologia , RNA Longo não Codificante/metabolismo , Animais , Células Cultivadas , Camundongos , RNA Longo não Codificante/genética
5.
Chem Res Toxicol ; 34(11): 2251-2260, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34520170

RESUMO

Phthalates, a plasticizer group, are used extensively in many of the products we use every day. Public health concerns are growing as recent studies have implicated butyl benzyl phthalate (BBP) as an obesogen. However, BBP-induced epigenetic regulation during adipogenesis is still unknown. We investigated if BBP altered miR-34a-5p, a key miRNA involved in obesity, and regulated its downstream pathway. Differentiating 3T3-L1 cells were exposed to various doses of BBP without exogenous adipogenic stimuli, tested for adipogenesis markers (PPARγ and aP2), and stained for lipid accumulation with Oil Red O staining. We then measured the expression of miR-34a-5p and its target genes, Nampt and Sirt1, along with another significant epigenetic modulator, Sirt3. Furthermore, using antagomiR, we examined whether miR-34a-5p knockdown decreased adipogenesis. BBP exposure resulted in augmented expression levels of miR-34a-5p with an associated increase in adipogenesis. BBP significantly decreased the Nampt, Sirt1, and Sirt3 gene expression levels. However, a decrease in the protein expression was observed only for Nampt, indicating that miR-34a-5p under BBP exposure may regulate Sirt1/Sirt3 only at the transcriptional level. Interestingly, in the presence of BBP, knockdown of miR-34a-5p decreased adipogenesis in the differentiating 3T3-L1 cells. Furthermore, miR-34a-5p knockdown increased the Nampt protein expression levels as well as NAD+ levels, indicating that miR-34a-5p regulates Nampt during BBP exposure. Additionally, the NAD+-dependent sirtuin activity decreased in BBP-treated cells and increased in miR-34a-5p knockdown cells with BBP treatment. BBP exposure demonstrated the involvement of epigenetic regulation by altering the expression patterns of miR-34a-5p and its target Nampt, which may perturb the energy homeostasis of the differentiating adipocytes by altering NAD+ levels and sirtuin activity, resulting in increased adipogenesis.


Assuntos
Adipogenia/efeitos dos fármacos , MicroRNAs/metabolismo , Ácidos Ftálicos/farmacologia , Células 3T3-L1 , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Camundongos , Transdução de Sinais/efeitos dos fármacos
6.
Int J Mol Sci ; 22(4)2021 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-33673073

RESUMO

Western diets contribute to metabolic diseases. However, the effects of various diets and epigenetic mechanisms are mostly unknown. Here, six week-old C57BL/6J male and female mice were fed with a low-fat diet (LFD), high-fat diet (HFD), and high-fat high-fructose diet (HFD-HF) for 20 weeks. We determined that HFD-HF or HFD mice experienced significant metabolic dysregulation compared to the LFD. HFD-HF and HFD-fed male mice showed significantly increased body weight, liver size, and fasting glucose levels with downregulated PPARγ, SCD1, and FAS protein expression. In contrast, female mice were less affected by HFD and HFD-HF. As miR-27b contains a seed sequence in PPARγ, it was discovered that these changes are accompanied by male-specific upregulation of miR-27b-5p, which is even more pronounced in the HFD-HF group (p < 0.01 vs. LFD) compared to the HFD group (p < 0.05 vs. LFD). Other miR-27 subtypes were increased but not significantly. HFD-HF showed insignificant changes in fibrosis markers when compared to LFD. Interestingly, fat ballooning in hepatocytes was increased in HFD-fed mice compared to HFD-HF fed mice, however, the HFD-HF liver showed an increase in the number of small cells. Here, we concluded that chronic Western diet-composition administered for 20 weeks may surpass the non-alcoholic fatty liver (NAFL) stage but may be at an intermediate stage between fatty liver and fibrosis via miR-27b-5p-induced PPARγ downregulation.


Assuntos
Dieta Ocidental/efeitos adversos , Regulação da Expressão Gênica , MicroRNAs/biossíntese , Hepatopatia Gordurosa não Alcoólica/metabolismo , PPAR gama/metabolismo , Transdução de Sinais , Animais , Masculino , Camundongos , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/patologia
7.
Int J Mol Sci ; 22(12)2021 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-34207035

RESUMO

Breast cancer is the most commonly occurring cancer in women of Western countries and is the leading cause of cancer-related mortality. The breast tumor microenvironment contains immune cells, fibroblasts, adipocytes, mesenchymal stem cells, and extracellular matrix. Among these cells, macrophages or tumor-associated macrophages (TAMs) are the major components of the breast cancer microenvironment. TAMs facilitate metastasis of the breast tumor and are responsible for poor clinical outcomes. High TAM density was also found liable for the poor prognosis of breast cancer. These observations make altering TAM function a potential therapeutic target to treat breast cancer. The present review summarizes the origin of TAMs, mechanisms of macrophage recruitment and polarization in the tumor, and the contributions of TAMs in tumor progression. We have also discussed our current knowledge about TAM-targeted therapies and the roles of miRNAs and exosomes in re-educating TAM function.


Assuntos
Neoplasias da Mama/etiologia , Neoplasias da Mama/metabolismo , Microambiente Tumoral , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/metabolismo , Animais , Biomarcadores Tumorais , Neoplasias da Mama/mortalidade , Neoplasias da Mama/terapia , Comunicação Celular , Progressão da Doença , Suscetibilidade a Doenças , Exossomos/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Imunomodulação , Ativação de Macrófagos/imunologia , MicroRNAs/genética , Metástase Neoplásica , Estadiamento de Neoplasias , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Carga Tumoral , Macrófagos Associados a Tumor/patologia
8.
Chem Res Toxicol ; 33(2): 461-469, 2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-31875672

RESUMO

Mono-(2-ethylhexyl)phthalate (MEHP) is a major bioactive metabolite which occurs from the widely used industrial plasticizer di(2-ethylhexyl)phthalate, which has been found to be toxic to several human physiological systems, including the cardiometabolic system. Recently, RNA methylation has been shown to be involved in cardio-metabolic regulation. Despite the importance of m6A mRNA methylation in physiological processes, studies of RNA methylation associated with endocrine disrupting chemicals are lacking. Here, we investigated the effects of MEHP in a cholesterol efflux pathway and the roles of m6A methylation using murine macrophage Raw 264.7 cells. MEHP exposure significantly reduced (P < 0.01 for 50 µM) m6A mRNA methylation with decreases in both mRNA (P < 0.01 for 5 and 50 µM) and protein (P < 0.05 for 0.5 µM; P < 0.01 for 5 µM; and P < 0.001 for 50 µM) expression levels of METTL14, a component of the methyltransferase complex. However, m5C RNA methylation remained unchanged. MEHP significantly reduced the expression of Scavenger Receptor B type 1 (SR-B1) (P < 0.01 for 5 µM and P < 0.05 for 50 µM). Additionally, we demonstrated that silencing METTL14 with MEHP decreased SR-B1 gene expression compared to the MEHP treatment (P < 0.01) or silencing METTL14 alone (P < 0.05). Furthermore, MEHP significantly promoted the m6A modification in SR-B1 (P < 0.001) and activated miRNAs which are predicted to regulate METTL14, such as miR16-1-3p (P < 0.05 for 50 µM MEHP), miR101a-3p (P < 0.05 for 5 and 50 µM MEHP), miR362-3-5p (P < 0.05 for 50 µM MEHP), miR501-5p (P < 0.01 for 50 µM MEHP), miR532-3p (P < 0.05 for 50 µM MEHP), and miR542-3p (P < 0.05 for 50 µM MEHP). Together, these results reveal for the first time that MEHP can modulate RNA methylation to regulate SR-B1 in a cholesterol efflux pathway.


Assuntos
Adenosina/análogos & derivados , Colesterol/metabolismo , Dietilexilftalato/análogos & derivados , MicroRNAs/genética , RNA/genética , RNA/metabolismo , Adenosina/metabolismo , Animais , Células Cultivadas , Dietilexilftalato/metabolismo , Metilação , Camundongos , Células RAW 264.7 , RNA/química
9.
Mol Cell Biochem ; 463(1-2): 211-223, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31686316

RESUMO

Atherosclerosis is associated with deregulated cholesterol metabolism and formation of macrophage foam cells. CCAAT/enhancer-binding protein beta (C/EBPß) is a transcription factor, and its inhibition has recently been shown to prevent atherosclerosis development and foam cell formation. However, whether C/EBPß regulates inflammation, endoplasmic reticulum (ER) stress, and apoptosis, in macrophage foam cells and its underlying molecular mechanism remains unknown. Here, we investigated the effect of C/EBPß knockdown on proteins and genes implicated in inflammation, ER stress, apoptosis, and autophagy in macrophage foam cells. RAW264.7 macrophage cells were transfected with control and C/EBPß-siRNA and then treated with nLDL and oxLDL. Key proteins and genes involved in inflammation, ER stress, apoptosis, and autophagy were analyzed by western blot and qPCR. We found that short interfering RNA (siRNA)-mediated knockdown of C/EBPß attenuated atherogenic lipid-mediated induction of proteins and genes implicated in inflammation (P-NFkB-p65, NFkB-p65, and TNFα), ER stress (ATF4 and ATF6), and apoptosis (CHOP, caspase 1, 3, and 12). Interestingly, C/EBPß knockdown upregulated the expression of autophagy proteins (LC3A/B-II, ATG5) and genes (LC3B, ATG5) but decreased the mammalian target of rapamycin (mTOR) protein phosphorylation and mTORC1 gene expression in oxLDL-loaded RAW264.7 macrophage cells. More importantly, treatment with rapamycin (inhibitor of mTOR) increased expression of proteins implicated in autophagy and cholesterol efflux in oxLDL-loaded RAW 264.7 macrophage cells. The present results suggest that C/EBPß inactivation regulates macrophage foam cell formation in atherogenesis by reducing inflammation, ER stress, and apoptosis and by promoting autophagy and inactivating mTOR.


Assuntos
Apoptose , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Estresse do Retículo Endoplasmático , Células Espumosas/metabolismo , Regulação da Expressão Gênica , Lipoproteínas LDL/metabolismo , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Células Espumosas/patologia , Técnicas de Silenciamento de Genes , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Lipoproteínas LDL/genética , Camundongos , Células RAW 264.7
10.
Am J Physiol Endocrinol Metab ; 317(2): E261-E268, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31084500

RESUMO

Elucidating the mechanism underlying the transmission of metabolic disease to subsequent generations requires robust preclinical mouse breeding strategies. Western diets rich in fat and carbohydrates are contributing factors in the rise of diabetes and obesity rates worldwide. Therefore, determining the impact of Western diets consumed by parents on offspring and future generations is critical for understanding the perpetuation of these diseases. Specifically, epigenetic regulation and transgenerational inheritance of metabolic disease is an emerging field of study requiring robust murine models. However, a major challenge to transgenerational studies is offspring mortality, exacerbated by maternal stress during pregnancy. Here, we describe a challenge experienced in our metabolic research in Western diet-fed female mice leading to the loss of litters via pup mortality and cannibalism by the mother. Furthermore, our study evaluates various breeding schemes with pregnancy efficiency and refined husbandry techniques to overcome pup mortality and infanticide, to characterize dams' and pups' metabolic characteristics, and to determine the impact on physiology of dams under detailed breeding schemes.


Assuntos
Pesquisa Biomédica/tendências , Cruzamento/métodos , Viabilidade Fetal/fisiologia , Tamanho da Ninhada de Vivíparos/fisiologia , Doenças Metabólicas , Efeitos Tardios da Exposição Pré-Natal , Estresse Fisiológico/fisiologia , Criação de Animais Domésticos/métodos , Criação de Animais Domésticos/tendências , Animais , Pesquisa Biomédica/métodos , Dieta Ocidental , Metabolismo Energético/fisiologia , Epigênese Genética/fisiologia , Feminino , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Doenças Metabólicas/genética , Doenças Metabólicas/mortalidade , Doenças Metabólicas/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/genética , Obesidade/metabolismo , Gravidez , Complicações na Gravidez/genética , Complicações na Gravidez/metabolismo , Complicações na Gravidez/mortalidade , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/mortalidade
11.
Chem Res Toxicol ; 32(10): 2016-2027, 2019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31508952

RESUMO

Organic compounds have been linked to adverse pregnancy complications. Perfluorooctanesulfonic acid (PFOS), a man-made fluorosurfactant and global pollutant, has been shown to induce oxidative stress in various cell types. Oxidative stress plays a key role in leading several placental diseases including preeclampsia (PE), gestational diabetes, spontaneous abortion, preterm labor, and intrauterine growth restriction. Recently, epigenetic regulation such as histone modifications, DNA methylation, and microRNAs (miRNAs), are shown to be associated with oxidative stress as well as pregnancy complications such as PE. However, whether PFOS exerts its detrimental effects in the placenta through epigenetics remains to be unveiled. Therefore, we aimed to investigate the effect of PFOS-induced reactive oxygen species (ROS) generation in first trimester human trophoblast cell line (HTR-8/SVneo) and whether epigenetic regulation is involved in this process. When treated with a range of PFOS doses at 24 and 48 h, even at 10 µM, it significantly increased the ROS production and decreased gene and protein expression, respectively, of the DNA methyltransferases DNMT1 (p < 0.001; p < 0.05), DNMT3A (p < 0.001; p < 0.05), and DNMT3B (p < 0.01; p < 0.01) and the sirtuins, for example, SIRT1 (p < 0.001; p < 0.001) and SIRT3 (p < 0.001; p < 0.05), while reducing global DNA methylation (p < 0.01) and increasing protein lysine acetylation (p < 0.001) as compared to vehicle controls. Interestingly, PFOS (10 µM) significantly increased miR29-b (p < 0.01), which has been previously reported to be associated with PE. The observed epigenetic effects were shown to be dependent on the expression of miR-29b, as knockdown of miR-29b significantly alters the gene and protein expression of DNMT1, DNMT3A, DNMT3B, SIRT1, and SIRT3 and ROS production as well as global DNA methylation and protein acetylation. This study provides for the first time a novel insight into PFOS-induced ROS generation via regulation of sets of the interactive epigenetic circuit in the placenta, which may lead to pregnancy complications.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Epigênese Genética/efeitos dos fármacos , Fluorocarbonos/toxicidade , Primeiro Trimestre da Gravidez/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos , Ácidos Alcanossulfônicos/química , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Epigênese Genética/genética , Feminino , Fluorocarbonos/química , Humanos , Gravidez , Primeiro Trimestre da Gravidez/genética , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo
12.
Chem Res Toxicol ; 32(9): 1863-1870, 2019 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-31423773

RESUMO

Human exposure to phthalates has received special attention due to their possible adverse human health effects. Diisononyl phthalate (DINP) is a plasticizer still widely used in many products, despite being considered an endocrine disruptor. In this study, we evaluated DINP's cytotoxicity, its effect on the levels of reactive oxygen species (ROS), and its effect on sirtuin expression in HepG2 cells. Results showed that 1 µg/mL DINP significantly downregulated Sirt1, Sirt2, Sirt3, and Sirt5 gene expression (p < 0.05), while other sirtuins remained unaffected. Furthermore, protein levels of Sirt1 and Sirt3 were significantly downregulated by 1 µg/mL DINP. On the other hand, 100 µg/mL DINP doubled the levels of lysine acetylation proteins (increased 2-fold) as well as reactive oxygen species (ROS) compared with the controls. In conclusion, our study suggests, for the first time, that DINP regulates the potential epigenetic disruptor sirtuin family and leads to induction of ROS via sirtuins.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Ácidos Ftálicos/toxicidade , Plastificantes/toxicidade , Sirtuínas/metabolismo , Acetilação/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Células Hep G2 , Humanos , Espécies Reativas de Oxigênio/metabolismo
13.
Chem Res Toxicol ; 32(5): 935-942, 2019 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-31016965

RESUMO

Artificial environmental endocrine disrupting chemicals (EDCs) exert public health concerns. Exposure to EDCs may induce various disorders in the cardiometabolic system. However, the underlying mechanisms remain largely unknown. Over the past decade, an abundance of evidence has emerged demonstrating a close link between cardiometabolic disorders and inflammation. The aim of the present study was to evaluate the immunological effects on macrophages from six EDCs via sirtuin (SIRT) regulation using the murine macrophage RAW 264.7 cell. We studied first the effects of these EDCs, including a series of doses of benzyl butyl phthalate (BBP), bisphenol A (BPA), diethylhexyl phthalate (DEHP), mono-(2-ethylhexyl)phthalate (MEHP), perfluorooctanoate (PFOA), or perfluorooctanesulfonate (PFOS), on SIRT1-7 transcriptional level. Among these EDCs, MEHP significantly decreased all sirtuin genes' expression in a dose-dependent manner. Under MEHP treatment, SIRT activity and protein expression were significantly decreased, while the protein expression of acetylated NF-κB was significantly increased along with significant increases in IL-1ß transcription. These results indicate that MEHP may induce the inflammatory response via SIRT-mediated acetylation of NF-κB. Additionally, the enhanced IL-1ß secretion in the presence of 50 µM MEHP ( P < 0.01) also supports inflammasome activation (significant ASC and NLRP3 protein augmentation). Both events may be regulated by MEHP induced reactive oxygen species ( P < 0.01). In conclusion, our study suggests for the first time that EDCs differentially modulate sirtuins' gene expression levels in macrophages and that a specific phthalate MEHP can lead to an increased inflammatory response by impairing vital epigenetic regulators and inflammasome activation.


Assuntos
Dietilexilftalato/análogos & derivados , Disruptores Endócrinos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamassomos/metabolismo , Inflamação/etiologia , Sirtuínas/metabolismo , Acetilação/efeitos dos fármacos , Ácidos Alcanossulfônicos/farmacologia , Animais , Compostos Benzidrílicos/farmacologia , Caprilatos/farmacologia , Dietilexilftalato/farmacologia , Epigênese Genética/efeitos dos fármacos , Fluorocarbonos/farmacologia , Inflamação/genética , Interleucina-1beta/metabolismo , Camundongos , NF-kappa B/química , NF-kappa B/metabolismo , Fenóis/farmacologia , Ácidos Ftálicos/farmacologia , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Sirtuínas/genética
14.
Analyst ; 144(13): 4033-4044, 2019 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-31143920

RESUMO

Epigenetic biomarkers are powerful tools for early disease detection and are particularly useful for elusive conditions like preeclampsia. Predicting preeclampsia at an early stage is one of the most important goals of maternal-fetal medicine. To this end, recent studies have identified microRNAs-such as microRNA-17-as early biomarkers for preeclampsia. Yet clinical applications are lagging, owing in part to the sensing challenges presented by the biomarkers' small size and complex environment. Surface enhanced Raman spectroscopy (SERS) is an emergent optical technique that is recognized for its potential to overcome these challenges. In this study, DNA functionalized nanoparticles were designed as probes to capture and quantify miRNA-17 in solution. SERS was used to determine the presence and concentration of miRNA-17 based on the formation of plasmonic nanoparticle aggregates. The miRNA-17 assay was tested at concentrations of 1 pM to 1 nM in both PBS and a representative complex biological sample. In both situations the assay was unaffected by non-complementary microRNA samples. These results demonstrate SERS's specificity and sensitivity for a new biomarker (miRNA-17) that may ultimately be used in a detection platform for early diagnosis of preeclampsia.


Assuntos
Sondas de DNA/química , DNA/química , Nanopartículas Metálicas/química , MicroRNAs/sangue , Animais , Biomarcadores/sangue , Bovinos , DNA/genética , Sondas de DNA/genética , Feminino , Limite de Detecção , MicroRNAs/genética , Hibridização de Ácido Nucleico , Pré-Eclâmpsia/diagnóstico , Gravidez , Prata/química , Análise Espectral Raman/métodos
15.
Fish Physiol Biochem ; 45(3): 907-920, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30536137

RESUMO

Nitric oxide (NO) is an important signalling molecule that plays diverse physiological functions in several vertebrates including that of adaptation to various stressful stimuli. The air-breathing magur catfish (Clarias magur) is known to tolerate a very high external ammonia (HEA) stress in its natural habitats. We report here the possible induction of inducible nitric oxide (inos) gene and more generation of NO in magur catfish exposed to HEA. Exposure to HEA (25 mM NH4Cl) for 14 days led to the higher accumulation of NO in different tissues of magur catfish and also more efflux of NO from the perfused liver of NH4Cl-treated fish as a consequence of high build of toxic ammonia in body tissues. More synthesis and accumulation of NO in body tissues was associated with the induction of iNOS activity, which otherwise was not detectable in control fish. The stimulation of iNOS activity in HEA exposed fish was mainly due to induction of inos gene as evidenced by more expression of inos mRNA and also more abundance of iNOS protein in different tissues of magur catfish. Immunocytochemical analysis indicated the zonal specific expression of iNOS protein in different tissues of magur catfish. The augmentation of iNOS in the fish under HEA could be an adaptive strategy of the fish to defend against the ammonia stress through the generation of NO. Therefore, the present finding identifies the potential role of iNOS to enhance the adaptive capacity and survivability of catfish under various adverse environmental and pathological conditions that it faces in its natural habitats.


Assuntos
Adaptação Fisiológica/fisiologia , Amônia/toxicidade , Peixes-Gato/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Fisiológico/fisiologia , Animais , Proteínas de Peixes/metabolismo , Óxido Nítrico Sintase Tipo II/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Chem Res Toxicol ; 29(3): 430-5, 2016 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-26871967

RESUMO

Phthalates, an endocrine disruptor group, cause oxidative stress (OS) in the placenta. However, no studies have reported OS-related miRNAs induced by phthalates. In the present study, we demonstrate that mono-(2-ethylhexyl) phthalate (MEHP) induces OS responsive miR-17-5p, miR-155-5p, and miR-126-3p in HTR8/SVneo in a dose- and time-dependent manner. Furthermore, MEHP altered the expression of phosphoinositide-3-kinase regulatory subunit 1α, phosphatase and tensin homolog, CDKN2A interacting protein, superoxide dismutase 2, and 3ß-hydroxysterol-D24 reductase, which are involved in OS and predicted to be regulated by these miRNAs. Our results suggest that placental exposure to MEHP may result in aberrant miRNA expression leading to pregnancy complications.


Assuntos
Dietilexilftalato/análogos & derivados , MicroRNAs/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Primeiro Trimestre da Gravidez/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Dietilexilftalato/química , Dietilexilftalato/farmacologia , Relação Dose-Resposta a Droga , Feminino , Humanos , MicroRNAs/genética , Estrutura Molecular , Gravidez , Primeiro Trimestre da Gravidez/genética , Primeiro Trimestre da Gravidez/metabolismo , Espécies Reativas de Oxigênio/análise , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade , Fatores de Tempo
17.
Life (Basel) ; 14(6)2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38929712

RESUMO

Metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly emerging as the most prevalent chronic liver disease, closely linked to the escalating rates of diabesity. The Western diet's abundance of fat and fructose significantly contributes to MASLD, disrupting hepatic glucose metabolism. We previously demonstrated that a high-fat and high-fructose diet (HFHFD) led to increased body and liver weight compared to the low-fat diet (LFD) group, accompanied by glucose intolerance and liver abnormalities, indicating an intermediate state between fatty liver and liver fibrosis in the HFHFD group. Sirtuins are crucial epigenetic regulators associated with energy homeostasis and play a pivotal role in these hepatic dysregulations. Our investigation revealed that HFHFD significantly decreased Sirt1 and Sirt7 gene and protein expression levels, while other sirtuins remained unchanged. Additionally, glucose 6-phosphatase (G6Pase) gene expression was reduced in the HFHFD group, suggesting a potential pathway contributing to fibrosis progression. Chromatin immunoprecipitation analysis demonstrated a significant increase in histone H3 lysine 18 acetylation within the G6Pase promoter in HFHFD livers, potentially inhibiting G6Pase transcription. In summary, HFHFD may inhibit liver gluconeogenesis, potentially promoting liver fibrosis by regulating Sirt7 expression. This study offers an epigenetic perspective on the detrimental impact of fructose on MASLD progression.

18.
Curr Res Toxicol ; 7: 100188, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39175913

RESUMO

The exposure to the ubiquitous phthalate metabolite mono-(2-ethylhexyl) phthalate (MEHP) is connected to dysregulated trophoblast function and placenta health; however, the underlying mechanisms preluding this scenario remain to be elucidated. In this study, we explored the hypoxemic effects of MEHP on a human placental first-trimester trophoblast cell line (HTR-8/Svneo). MEHP-treated trophoblast cells displayed significantly increased levels of oxidative stress and hypoxia-inducible factor-1 alpha (HIF-1α) attributed by the induction of hypoxia. Further, HIF-1α exhibited higher DNA binding activity and upregulated gene expression of its downstream target vascular endothelial growth factor A (VEGFA). The hypoxia-induced microRNA miR-210-3p was also significantly increased upon MEHP treatment followed by disrupted mitochondrial ATP generation and membrane potential. This was identified to possibly be facilitated by lowered mitochondrial DNA copy number and inhibited expression of electron transport chain subunits, such as mitochondrial complex-IV. These results suggest potential adverse effects of MEHP exposure in a trophoblast cell line mediated by HIF-1α and the epigenetic modulator miR-210-3p. Chronic placental hypoxia and oxidative stress have long been implicated in the pathogenesis of pregnancy complications such as preeclampsia. As we've revealed genetic and epigenetic factors underscoring a potential mechanism induced by MEHP, this brings to light another significant implication of phthalate exposure on maternal and fetal health.

19.
Biochim Biophys Acta Mol Cell Res ; 1871(4): 119708, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38508420

RESUMO

Phthalates' pervasive presence in everyday life poses concern as they have been revealed to induce perturbing health defects. Utilized as a plasticizer, phthalates are riddled throughout many common consumer products including personal care products, food packaging, home furnishings, and medical supplies. Phthalates permeate into the environment by leaching out of these products which can subsequently be taken up by the human body. It is previously established that a connection exists between phthalate exposure and cardiovascular disease (CVD) development; however, the specific mitochondrial link in this scenario has not yet been described. Prior studies have indicated that one possible mechanism for how phthalates exert their effects is through mitochondrial dysfunction. By disturbing mitochondrial structure, function, and signaling, phthalates can contribute to the development of the foremost cause of death worldwide, CVD. This review will examine the potential link among phthalates and their effects on the mitochondria, permissive of CVD development.


Assuntos
Doenças Cardiovasculares , Ácidos Ftálicos , Humanos , Doenças Cardiovasculares/induzido quimicamente , Ácidos Ftálicos/toxicidade
20.
Biomedicines ; 12(7)2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-39062015

RESUMO

BACKGROUND: MicroRNAs (miRNAs) play a regulatory role in various human cancers. The roles of hsa-miR-15a-5p, hsa-miR-99b-5p, and hsa-miR-181a-5p have not been fully explored in the angiogenesis of renal cell carcinoma (RCC). AIMS: The present study aimed to evaluate the expression of these miRNAs in tumorous and adjacent healthy tissues of RCC. METHODS: Paired tumorous and adjacent normal kidney tissues from 20 patients were studied. The expression levels of hsa-miR-15b-5p, hsa-miR-99b-5p, and hsa-miR-181a-5p were quantified by TaqMan miRNA Assays. Putative targets were analyzed by qRT-PCR. RESULTS: Significant downregulation of all three miRNAs investigated was observed in tumorous samples compared to adjacent normal kidney tissues. Spearman analysis showed a negative correlation between the expression levels of miRNAs and the pathological grades of the patients. Increased expression of vascular endothelial growth factor-A (VEGF-A) and hypoxia-inducible factor-1α (HIF-1α), a tissue inhibitor of metalloproteinases-1 (TIMP-1), was observed in tumorous samples compared to adjacent normal tissues. Depletion of tissue inhibitors of metalloproteinase-2 (TIMP-2) and metalloproteinase-2 (MMP-2) was detected compared to normal adjacent tissues. The examined miRNAs might function as contributing factors to renal carcinogenesis. However, more prospective studies are warranted to evaluate the potential role of miRNAs in RCC angiogenesis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA