Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Cell Mol Gastroenterol Hepatol ; 12(5): 1669-1682, 2021.
Article in English | MEDLINE | ID: mdl-34245919

ABSTRACT

BACKGROUND & AIMS: Epigenetic regulation of gene expression plays a critical role in the development of liver cancer; however, the molecular mechanisms of epigenetic-driven liver cancers are not well understood. The aims of this study were to examine molecular mechanisms that cause the dedifferentiation of hepatocytes into cancer cells in aggressive hepatoblastoma and test if the inhibition of these mechanisms inhibits tumor growth. METHODS: We have analyzed CCAAT/Enhancer Binding Protein alpha (C/EBPα), Transcription factor Sp5, and histone deacetylase (HDAC)1 pathways from a large biobank of fresh hepatoblastoma (HBL) samples using high-pressure liquid chromatography-based examination of protein-protein complexes and have examined chromatin remodeling on the promoters of markers of hepatocytes and p21. The HDAC1 activity was inhibited in patient-derived xenograft models of HBL and in cultured hepatoblastoma cells and expression of HDAC1-dependent markers of hepatocytes was examined. RESULTS: Analyses of a biobank showed that a significant portion of HBL patients have increased levels of an oncogenic de-phosphorylated-S190-C/EBPα, Sp5, and HDAC1 compared with amounts of these proteins in adjacent regions. We found that the oncogenic de-phosphorylated-S190-C/EBPα is created in aggressive HBL by protein phosphatase 2A, which is increased within the nucleus and dephosphorylates C/EBPα at Ser190. C/EBPα-HDAC1 and Sp5-HDAC1 complexes are abundant in hepatocytes, which dedifferentiate into cancer cells. Studies in HBL cells have shown that C/EBPα-HDAC1 and Sp5-HDAC1 complexes reduce markers of hepatocytes and p21 via repression of their promoters. Pharmacologic inhibition of C/EBPα-HDAC1 and Sp5-HDAC1 complexes by Suberoylanilide hydroxamic acid (SAHA) and small interfering RNA-mediated inhibition of HDAC1 increase expression of hepatocyte markers, p21, and inhibit proliferation of cancer cells. CONCLUSIONS: HDAC1-mediated repression of markers of hepatocytes is an essential step for the development of HBL, providing background for generation of therapies for aggressive HBL by targeting HDAC1 activities.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Hepatocytes/metabolism , Histone Deacetylase 1/metabolism , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , p21-Activated Kinases/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Hepatocytes/pathology , Histone Deacetylase 1/genetics , Humans , Liver Neoplasms/pathology , Models, Biological , Multiprotein Complexes/metabolism , Promoter Regions, Genetic , Protein Binding , Signal Transduction , p21-Activated Kinases/genetics
2.
Article in English | MEDLINE | ID: mdl-34004356

ABSTRACT

The nutrient sensors peroxisome proliferator-activated receptor γ (PPARγ) and mechanistic target of rapamycin complex 1 (mTORC1) closely interact in the regulation of adipocyte lipid storage. The precise mechanisms underlying this interaction and whether this extends to other metabolic processes and the endocrine function of adipocytes are still unknown. We investigated herein the involvement of mTORC1 as a mediator of the actions of the PPARγ ligand rosiglitazone in subcutaneous inguinal white adipose tissue (iWAT) mass, endocrine function, lipidome, transcriptome and branched-chain amino acid (BCAA) metabolism. Mice bearing regulatory associated protein of mTOR (Raptor) deletion and therefore mTORC1 deficiency exclusively in adipocytes and littermate controls were fed a high-fat diet supplemented or not with the PPARγ agonist rosiglitazone (30 mg/kg/day) for 8 weeks and evaluated for iWAT mass, lipidome, transcriptome (Rnaseq), respiration and BCAA metabolism. Adipocyte mTORC1 deficiency not only impaired iWAT adiponectin transcription, synthesis and secretion, PEPCK mRNA levels, triacylglycerol synthesis and BCAA oxidation and mRNA levels of related proteins but also completely blocked the upregulation in these processes induced by pharmacological PPARγ activation with rosiglitazone. Mechanistically, adipocyte mTORC1 deficiency impairs PPARγ transcriptional activity by reducing PPARγ protein content, as well as by downregulating C/EBPα, a co-partner and facilitator of PPARγ. In conclusion, mTORC1 and PPARγ are essential partners involved in the regulation of subcutaneous adipose tissue adiponectin production and secretion and BCAA oxidative metabolism.


Subject(s)
Adiponectin/metabolism , Amino Acids, Branched-Chain/metabolism , Glycerol/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , PPAR gamma/metabolism , Subcutaneous Fat/metabolism , Up-Regulation , Animals , Mice , Oxidation-Reduction
3.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;54(2): e9869, 2021. tab, graf
Article in English | LILACS, Coleciona SUS | ID: biblio-1142585

ABSTRACT

Severe blockage in myeloid differentiation is the hallmark of acute myeloid leukemia (AML). Trdmt1 plays an important role in hematopoiesis. However, little is known about the function of Trdmt1 in AML cell differentiation. In the present study, Trdmt1 was up-regulated and miR-181a was down-regulated significantly during human leukemia HL-60 cell differentiation after TAT-CT3 fusion protein treatment. Accordingly, miR-181a overexpression in HL-60 cells inhibited granulocytic maturation. In addition, our "rescue" assay demonstrated that Trdmt1 3′-untranslated region promoted myeloid differentiation of HL-60 cells by sequestering miR-181a and up-regulating C/EBPα (a critical factor for normal myelopoiesis) via its competing endogenous RNA (ceRNA) activity on miR-181a. These findings revealed an unrecognized role of Trdmt1 as a potential ceRNA for therapeutic targets in AML.


Subject(s)
Humans , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Cell Differentiation , HL-60 Cells
4.
Reprod Biol ; 20(2): 247-253, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32089504

ABSTRACT

Hypothyroidism affects the content of triacylglycerol (TAG), total cholesterol (TC), oxidized lipids, glycogen, and infiltration of immune cells into the ovary and uterus. This study aimed to analyze the impact of hypothyroidism on the lipid content of different regions of the oviduct. Control (n = 6) and hypothyroid (n = 6; 10 mg/kg/day of methimazole in the drinking water for 30 days) adult rabbits were used. In the fimbriae/infundibulum (FIM/INF), ampulla, (AMP), isthmus (IST), and utero-tubal junction (UTJ), the TAG and TC concentrations, presence of oxidized lipid, relative expressions of perilipin A (PLIN A), peroxisome proliferator-activated receptor γ (PPARγ), CAAT/enhancer-binding protein α (C/EBPα), and farnesoid X receptor (FXRα) were analyzed. The content of glycogen and glycans, as well as the infiltration of lymphocytes, were also quantified. In the FIM/INF, hypothyroidism reduced the content of TC, expression of C/EBPα, and presence of glycans while increased the number of intraepithelial lymphocytes. In the AMP and IST-UTJ regions, hypothyroidism increased the content of TAG, oxidized lipids, expression of PPARγ, and glycogen content but decreased the expression of PLIN-A. The FXRα expression in secretory cells of IST-UTJ was higher in the hypothyroid rabbits compared to controls. Additionally, hypothyroidism reduced the C/EBPα expression and the number of intraepithelial lymphocytes in the AMP and IST-UTJ regions, respectively. We demonstrated that the effect of hypothyroidism depends on the oviductal region, possibly associated with different physiological functions specific to each region. These alterations may be related to infertility, tubal disturbances, and ectopic pregnancy observed in hypothyroid women.


Subject(s)
Fallopian Tubes/cytology , Glycogen/chemistry , Hypothyroidism/veterinary , Lipids/chemistry , Lymphocytes/physiology , Rabbits , Animals , Antithyroid Agents/toxicity , Female , Glycogen/metabolism , Hypothyroidism/chemically induced , Lipid Metabolism , Methimazole/toxicity
5.
Biomed Pharmacother ; 109: 1860-1866, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30551441

ABSTRACT

Previous studies have reported the anti-obesity effects of α, ß-Amyrin in high fat-fed mice. This study aimed to evaluate whether α, ß-Amyrin has an anti-adipogenic effect in 3T3-L1 murine adipocytes and to explore the possible underlying mechanisms. 3T3-L1 pre-adipocytes were differentiated in a medium containing insulin, dexamethasone, and 1-methyl-3-isobutylxanthine. Cytotoxicity of α, ß-Amyrin was assessed by MTT assay. Lipid content in adipocytes was determined by Oil-Red O staining. In addition, the protein expression levels of peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer binding proteins alpha (C/EBPα), beta (C/EBPß), and delta (C/EBPδ) and glucose transporter 4 (GLUT4) were determined by qRT-PCR and western blot analysis. Oil-Red O staining revealed markedly reduced fat accumulation by α, ß-Amyrin (6.25-50 µg/mL) without affecting cell viability. Furthermore, our results indicate that α, ß-Amyrin can significantly suppress the adipocyte differentiation by downregulating the expression levels of adipogenesis-related key transcription factors such as PPARγ and C/EBPα, but not C/EBPß or C/EPBδ. In addition, the protein expression of membrane GLUT4 in 3T3- L1 adipocytes treated with α, ß-Amyrin was significantly higher than in control cells, indicating that α, ß-Amyrin augments glucose uptake. These findings suggest that α, ß-Amyrin exerts an anti-adipogenic effect principally via modulation of lipid and carbohydrate metabolism in 3T3-L1cells. The present in vitro findings, taken together with our earlier observation of the anti-obesity effect in vivo, suggest that α, ß-Amyrin can be developed as a new therapeutic agent for treatment and prevention of obesity.


Subject(s)
Adipocytes/drug effects , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/drug effects , Down-Regulation/drug effects , Oleanolic Acid/analogs & derivatives , PPAR gamma/metabolism , Pentacyclic Triterpenes/pharmacology , 3T3-L1 Cells , Adipogenesis/drug effects , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Cell Survival/drug effects , Fatty Acid-Binding Proteins/metabolism , Glucose Transporter Type 4/metabolism , Mice , Obesity/drug therapy , Obesity/metabolism , Oleanolic Acid/pharmacology , Plant Extracts/pharmacology
6.
Biochim Biophys Acta Mol Cell Res ; 1866(2): 225-239, 2019 02.
Article in English | MEDLINE | ID: mdl-30389374

ABSTRACT

GM2-gangliosidosis, a subgroup of lysosomal storage disorders, is caused by deficiency of hexosaminidase activity, and comprises the closely related Tay-Sachs and Sandhoff diseases. The enzyme deficiency prevents normal metabolization of ganglioside GM2, usually resulting in progressive neurodegenerative disease. The molecular mechanisms whereby GM2 accumulation in neurons triggers neurodegeneration remain unclear. In vitro experiments, using microsomes from Sandhoff mouse model brain, showed that increase of GM2 content negatively modulates sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) (Pelled et al., 2003). Furthermore, Ca2+ depletion in endoplasmic reticulum (ER) triggers Unfolded Protein Response (UPR), which tends to restore homeostasis in the ER; however, if cellular damage persists, an apoptotic response is initiated. We found that ER GM2 accumulation in cultured neurons induces luminal Ca2+ depletion, which in turn activates PERK (protein kinase RNA [PKR]-like ER kinase), one of three UPR sensors. PERK signaling displayed biphasic activation; i.e., early upregulation of cytoprotective calcineurin (CN) and, under prolonged ER stress, enhanced expression of pro-apoptotic transcription factor C/EBP homologous protein (CHOP). Moreover, GM2 accumulation in neuronal cells induced neurite atrophy and apoptosis. Both processes were effectively modulated by treatment with the selective PERK inhibitor GSK2606414, by CN knockdown, and by CHOP knockdown. Overall, our findings demonstrate the essential role of PERK signaling pathway contributing to neurodegeneration in a model of GM2-gangliosidosis.


Subject(s)
Gangliosidoses, GM2/metabolism , Neurites/physiology , eIF-2 Kinase/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Atrophy/metabolism , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/physiology , G(M2) Ganglioside/metabolism , G(M2) Ganglioside/physiology , Gangliosidoses, GM2/genetics , Indoles/pharmacology , Mice , Neurites/metabolism , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Signal Transduction/genetics , Transcription Factor CHOP/metabolism , Unfolded Protein Response/physiology , eIF-2 Kinase/physiology
7.
Mol Cell Biochem ; 448(1-2): 299-309, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29468504

ABSTRACT

We investigated for the first time the expression of melanoma cell adhesion molecule (MCAM) and its involvement in the differentiation of 3T3-L1 fibroblasts to adipocytes. We found that MCAM mRNA increased subsequent to the activation of the master regulator of adipogenesis, PPARγ, and this increase was maintained in the mature adipocytes. On the other hand, MCAM knockdown impaired differentiation and induction of PPARγ as well as expression of genes activated by PPARγ. However, events that precede and are necessary for early PPARγ activation, such as C/EBPß induction, ß-catenin downregulation, and ERK activation, were not affected in the MCAM knockdown cells. In keeping with this, the increase in PPARγ mRNA that precedes MCAM induction was not altered in the knockdown cells. In conclusion, our findings suggest that MCAM is a gene upregulated and involved in maintaining PPARγ induction in the late but not in the early stages of 3T3-L1 fibroblasts adipogenesis.


Subject(s)
Adipocytes/metabolism , Adipogenesis , Cell Differentiation , Fibroblasts/metabolism , Gene Expression Regulation , PPAR gamma/biosynthesis , 3T3-L1 Cells , Adipocytes/cytology , Animals , CD146 Antigen/genetics , CD146 Antigen/metabolism , Fibroblasts/cytology , Gene Knockdown Techniques , Mice , PPAR gamma/genetics
8.
Br J Nutr ; 118(10): 788-803, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29110748

ABSTRACT

Perinatal maternal high-fat (HF) diet programmes offspring obesity. Obesity is associated with overactivation of the endocannabinoid system (ECS) in adult subjects, but the role of the ECS in the developmental origins of obesity is mostly unknown. The ECS consists of endocannabinoids, cannabinoid receptors (cannabinoid type-1 receptor (CB1) and cannabinoid type-2 receptor (CB2)) and metabolising enzymes. We hypothesised that perinatal maternal HF diet would alter the ECS in a sex-dependent manner in white and brown adipose tissue of rat offspring at weaning in parallel to obesity development. Female rats received standard diet (9 % energy content from fat) or HF diet (29 % energy content from fat) before mating, during pregnancy and lactation. At weaning, male and female offspring were killed for tissue harvest. Maternal HF diet induced early obesity, white adipocyte hypertrophy and increased lipid accumulation in brown adipose tissue associated with sex-specific changes of the ECS's components in weanling rats. In male pups, maternal HF diet decreased CB1 and CB2 protein in subcutaneous adipose tissue. In female pups, maternal HF diet increased visceral and decreased subcutaneous CB1. In brown adipose tissue, maternal HF diet increased CB1 regardless of pup sex. In addition, maternal HF diet differentially changed oestrogen receptor across the adipose depots in male and female pups. The ECS and oestrogen signalling play an important role in lipogenesis, adipogenesis and thermogenesis, and we observed early changes in their targets in adipose depots of the offspring. The present findings provide insights into the involvement of the ECS in the developmental origins of metabolic disease induced by inadequate maternal nutrition in early life.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Endocannabinoids/metabolism , Maternal Nutritional Physiological Phenomena , Obesity/etiology , Receptors, Cannabinoid/metabolism , Weaning , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adiposity , Animal Nutritional Physiological Phenomena , Animals , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Female , Lactation , Lipid Metabolism , Male , Obesity/metabolism , Pregnancy , Prenatal Nutritional Physiological Phenomena , Rats, Wistar , Receptors, Estrogen/metabolism , Sex Factors , Thermogenesis
9.
J Cell Biochem ; 117(8): 1797-805, 2016 08.
Article in English | MEDLINE | ID: mdl-26729411

ABSTRACT

Proper regulation of gene expression is essential for normal development, cellular growth, and differentiation. Differential expression profiles of mRNA coding for vertebrate Ric-8B during embryo and adult stages have been observed. In addition, Ric-8B is expressed in few cerebral nuclei subareas. These facts point to a dynamic control of RIC8B gene expression. In order to understand the transcriptional regulation of this gene, we searched for cis-elements in the sequence of the human RIC8B promoter region, identifying binding sites for the basic/leucine zipper (bZip) CREB transcription factor family (CRE sites) and C/EBP transcription factor family (C/EBP sites). CRE sites were found clustered near the transcription start site, while the C/EBP sites were found clustered at around 300 bp upstream the CRE sites. Here, we demonstrate the ability of CREB1 and C/EBPß to bind their respective elements identified in the RIC8B promoter. Comparative protein-DNA interaction analyses revealed only the proximal elements as high affinity sites for CREB1 and only the distal elements as high affinity sites for C/EBPß. Chromatin immunoprecipitation analyses, carried out using a human neuroblastoma cell line, confirmed the preferential association of CREB to the proximal region of the RIC8B promoter. By performing luciferase reporter assays, we found the CRE sites as the most relevant elements for its transcriptional activity. Taken together, these data show the existence of functional CREB and C/EBP binding sites in the human RIC8B gene promoter, a particular distribution of these sites and demonstrate a relevant role of CREB in stimulating transcriptional activity of this gene. J. Cell. Biochem. 117: 1797-1805, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Guanine Nucleotide Exchange Factors/biosynthesis , Response Elements , Transcription, Genetic/physiology , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , Guanine Nucleotide Exchange Factors/genetics , Humans
10.
Arq. bras. endocrinol. metab ; Arq. bras. endocrinol. metab;53(5): 582-594, jul. 2009. ilus
Article in Portuguese | LILACS | ID: lil-525420

ABSTRACT

A obesidade é um dos principais problemas de saúde pública. Indivíduos obesos são mais suscetíveis a desenvolver doenças cardiovasculares e diabetes melito tipo 2. A obesidade resulta do aumento no tamanho e no número de adipócitos. O balanço entre adipogênese e adiposidade determina o grau de obesidade do indivíduo. Adipócitos maduros secretam adipocinas, tais como TNFα, IL-6, leptina e adiponectina, e lipocina, o ácido palmitoleico ω-7. A produção de adipocinas é maior na obesidade, o que contribui para o estabelecimento de resistência periférica à insulina. O conhecimento dos eventos moleculares que regulam a diferenciação dos pré-adipócitos e de células-tronco mesenquimais em adipócitos (adipogênese) é importante para o entendimento da gênese da obesidade. A ativação do fator de transcrição PPARγ é essencial na adipogênese. Certos ácidos graxos são ligantes de PPARγ e podem, assim, controlar a adipogênese. Além disso, alguns ácidos graxos atuam como moléculas sinalizadoras em adipócitos, regulando sua diferenciação ou morte. Dessa forma, a composição lipídica da dieta e os agonistas de PPARγ podem regular o balanço entre adipogênese e morte de adipócitos e, portanto, a obesidade.


Obesity is one of the major Public Health problems. Obese individuals are more susceptible to develop cardiovascular diseases and type 2 diabetes mellitus. The obesity results from the increase in size and number of the adipocytes. The balance between adipogenesis and adiposity determines the degree of obesity. Mature adipocytes secrete adipokines, such as TNFα, IL-6, leptine and adiponectin, and lipokine, the palmitoleic acid ω-7. The production of adipokines is increased in obesity, contributing to the onset of peripheral insulin resistance. The knowledge about the molecular events that regulate the differentiation of pre-adipocytes and mesenchymal stem cells into adipocytes (adipogenesis) is important for the comprehension of the genesis of obesity. Activation of transcription factor PPARγ plays an essential role in the adipogenesis. Certain fatty acids are PPARγ ligands and can control adipogenesis. Moreover, some fatty acids act as signaling molecules regulating their differentiation into adipocytes or death. Accordingly, the lipid composition of the diet and PPARγ agonists can regulate the balance between adipogenesis and death of adipocytes and, therefore, the obesity.


Subject(s)
Animals , Humans , Adipogenesis/physiology , Adipose Tissue/metabolism , Cardiovascular Diseases , Fatty Acids/metabolism , Obesity/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Adipogenesis/drug effects , Adipose Tissue/drug effects , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Fatty Acids/therapeutic use , Linoleic Acids, Conjugated/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL