Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Arch Biochem Biophys ; 752: 109886, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38215960

RESUMEN

Recent studies have shown that some natural compounds from plants prevent obesity and related disorders, including the loss of skeletal muscle mass and strength. In this study, we investigated the effect of echinacoside (ECH), a caffeic acid glycoside from the phenylpropanoid class, on myogenesis and ATP-dependent thermogenesis in the skeletal muscle and its interaction with the dopaminergic receptors 1 and 5 (DRD1 and DRD5). We applied RT-PCR, immunoblot analysis, a staining method, and an assay kit to determine the effects of ECH on diverse target genes and proteins involved in skeletal muscle myogenesis and ATP-consuming futile processes. Our study demonstrated that ECH enhanced myogenic differentiation, glucose, and fatty acid uptake, as well as lipid catabolism, and induced ATP-dependent thermogenesis in vitro and in vivo. Moreover, ECH upregulated mitochondrial biogenesis proteins, mitochondrial oxidative phosphorylation (OXPHOS) complexes, and intracellular Ca2+ signaling as well as thermogenic proteins. These findings were further elucidated by mechanistic studies which showed that ECH mediates myogenesis via the DRD1/5 in C2C12 muscle cells. In addition, ECH stimulates α1-AR-mediated ATP-dependent thermogenesis via the DRD1/5/cAMP/SLN/SERCA1a pathway in C2C12 muscle cells. To the best of our knowledge, this is the first report that demonstrates the myogenic and thermogenic potential of ECH activity through the dopaminergic receptors. Understanding the novel functions of ECH in terms of its ability to prevent skeletal muscle loss and energy expenditure via ATP-consuming futile processes could help to develop potential alternative strategies to address muscle-related diseases, including combating obesity.


Asunto(s)
Músculo Esquelético , Obesidad , Humanos , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Glicósidos/farmacología , Adenosina Trifosfato/metabolismo , Desarrollo de Músculos/fisiología , Termogénesis/fisiología
2.
Arch Biochem Biophys ; 755: 109975, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38531438

RESUMEN

Capsaicin (CAP) is a natural bioactive compound in chili pepper that activates the transient receptor potential vanilloid subfamily 1 (TRPV1) and is known to stimulate uncoupling protein 1 (UCP1)-dependent thermogenesis. However, its effect on ATP-dependent thermogenesis remains unknown. In this study, we employed qRT-PCR, immunoblot, staining method, and assay kit to investigate the role of CAP on ATP-dependent thermogenesis and its modulatory roles on the TRPV1, ß3-adrenergic receptor (ß3-AR), and α1-AR using in vitro and in vivo models. The studies showed that CAP treatment in high-fat diet-induced obese mice resulted in lower body weight gain and elevated ATP-dependent thermogenic effectors' protein and gene expression through ATP-consuming calcium and creatine futile cycles. In both in vitro and in vivo experiments, CAP treatment elevated the protein and gene expressions of sarcoendoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2), ryanodine receptor 2 (RYR2), creatine kinase B (CKB), and creatine kinase mitochondrial 2 (CKMT2) mediated by the activation of ß3-AR, α1-AR, and TRPV1. Our study showed that CAP increased intracellular Ca2+ levels and the expression of voltage-dependent anion channel (VDAC) and mitochondrial calcium uniporter (MCU) which indicates that increased mitochondrial Ca2+ levels lead to increased expression of oxidative phosphorylation protein complexes as a result of ATP-futile cycle activation. A mechanistic study in 3T3-L1 adipocytes revealed that CAP induces UCP1- and ATP-dependent thermogenesis mediated by the ß3-AR/PKA/p38MAPK/ERK as well as calcium-dependent α1-AR/TRPV1/CaMKII/AMPK/SIRT1 pathway. Taken together, we identified CAP's novel functional and modulatory roles in UCP1- and ATP-dependent thermogenesis, which is important for developing therapeutic strategies for combating obesity and metabolic diseases.

3.
Pflugers Arch ; 475(6): 757-773, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37103560

RESUMEN

The activation of beige fat and muscle tissues is an interesting and encouraging target for therapeutic intervention in obesity owing to their remarkable lipolytic activity and energy-consuming futile cycles. This study examined the effect of dopamine receptor D4 (DRD4) on lipid metabolisms as well as UCP1- and ATP-dependent thermogenesis in Drd4-silenced 3T3-L1 adipocytes and C2C12 muscle cells. Silencing of Drd4, followed by quantitative real-time PCR, immunoblot analysis, immunofluorescence, and staining methods, were applied to evaluate the effects of DRD4 on diverse target genes and proteins of both cells. The findings showed that DRD4 was expressed in the adipose and muscle tissues of normal and obese mice. Furthermore, the knockdown of Drd4 upregulated the expression of brown adipocyte-specific genes and proteins while downregulating lipogenesis and the adipogenesis marker proteins. Drd4 silencing also upregulated the expression of key signaling molecules involved in ATP-dependent thermogenesis in both cells. This was further elucidated by mechanistic studies showing that a Drd4 knockdown mediates UCP1-dependent thermogenesis via the cAMP/PKA/p38MAPK pathway in 3T3-L1 adipocytes and UCP1-independent thermogenesis via the cAMP/SLN/SERCA2a pathway in C2C12 muscle cells. In addition, siDrd4 also mediates myogenesis via the cAMP/PKA/ERK1/2/Cyclin D3 pathway in C2C12 muscle cells. Silencing of Drd4 promotes ß3-AR-dependent browning in 3T3-L1 adipocytes and α1-AR/SERCA-based thermogenesis through an ATP-consuming futile process in C2C12 muscle cells. Understanding the novel functions of DRD4 on adipose and muscle tissues in terms of its ability to enhance energy expenditure and regulate whole-body energy metabolism will aid in developing novel obesity intervention techniques.


Asunto(s)
Adipocitos Marrones , Obesidad , Animales , Ratones , Células 3T3-L1 , Adenosina Trifosfato/metabolismo , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/metabolismo , Células Musculares/metabolismo , Obesidad/metabolismo , Receptores Dopaminérgicos/metabolismo , Receptores de Dopamina D4/metabolismo , Termogénesis , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
4.
Arch Biochem Biophys ; 739: 109581, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36948352

RESUMEN

The activation of brown fat and induction of beige adipocytes, so-called non-shivering thermogenesis, is emerging as a promising target for therapeutic intervention in obesity management. Our previous report demonstrated that ß-carotene (BC) induces beige adipocytes to increase UCP1-dependent thermogenic activity. However, the UCP1-independent thermogenic effect of BC on adipose tissues remains unexplored. In this study, we examined the effects of BC on UCP1-independent thermogenic activity with a focus on the ATP-consuming futile cycles in 3T3-L1 adipocytes. BC increased intracellular calcium levels and stimulated the expression of calcium cycling-related proteins, including sarcoendoplasmic reticulum Ca2+-ATPase (SERCA) 2b, ryanodine receptor 2 (RyR2), voltage-dependent anion channel (VDAC), mitochondrial calcium uniporter (MCU), and Ca2+/calmodulin-dependent protein kinase 2 (CaMK2) in 3T3-L1 white adipocytes. In addition, BC stimulated thermogenesis by activating the creatine metabolism-related thermogenic pathway. Moreover, BC activated ß-carotene oxygenase 1 (BCO1), which efficiently cleaved BC to retinal and consequently converted to its transcriptionally active form retinoic acid. These BC conversion products also exhibited thermogenic effects comparable to a similar level of BC. The mechanistic study revealed that retinal exhibited thermogenic activity independently of retinoic acid and retinoic acid-mediated thermogenesis was resulted partly from conversion of retinal. Moreover, BC activated α1-AR and UCP1-independent thermogenic effectors independently of UCP1 expression. In conclusion, the thermogenic response to BC and its conversion products in 3T3-L1 white adipocytes involves two interacting pathways, one mediated via ß3-adrenergic receptors (ß3-AR) and cyclic adenosine monophosphate (cAMP) and the other via α1-AR and increases in cytosolic Ca2+ levels activated by calcium regulatory proteins.


Asunto(s)
Adipocitos Blancos , beta Caroteno , Ratones , Animales , Adipocitos Blancos/metabolismo , beta Caroteno/farmacología , beta Caroteno/metabolismo , Calcio/metabolismo , Ciclo del Sustrato , Células 3T3-L1 , Tejido Adiposo Pardo/metabolismo , Adenosina Trifosfato/metabolismo , Termogénesis/fisiología , Tretinoina/metabolismo , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
5.
J Cell Biochem ; 123(5): 863-877, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35233844

RESUMEN

Obesity and related metabolic disorders are epidemic diseases. Promoting thermogenesis and a functional increase in the browning of white adipocytes may counteract obesity. On the other hand, the molecular mechanism that regulates brown and beige fat-mediated thermogenesis is unclear. This article reports a molecular network led by cytoplasmic FMR1-interacting protein 2 (CYFIP2) that negatively regulates adipocyte browning in white adipocytes. Although the function of CYFIP2 in Fragile X Syndrome (FXS) and autism have been reported, its physiological roles in adipocytes remain elusive. Therefore, this study examined the physiological consequences of its deprivation in cultured 3T3-L1 white adipocytes using loss-of-function studies. Combined real-time quantitative reverse-transcription polymerase chain reaction and immunoblot analysis showed that the loss of CYFIP2 induces fat browning, as evidenced by the gene and protein expression levels of the brown fat-associated markers. A deficiency of CYFIP2 promoted mitochondrial biogenesis and significantly enhanced the expression of the core set beige fat-specific genes (Cd137, Cidea, Cited1, Tbx1, and Tmem26) and proteins (PGC-1α, PRDM16, and UCP1). In addition, a CYFIP2 deficiency promoted lipid catabolism and suppressed adipogenesis, lipogenesis, and autophagy. A mechanistic study showed that the loss of CYFIP2 induces browning in white adipocytes, independently via the activation of mTORC1 and suppression of the GABA-BR signaling pathway. The present data revealed a previously unidentified mechanism of CYFIP2 in the browning of white adipocytes and emphasized the potential of CYFIP2 as a pharmacotherapeutic target for treating obesity and other metabolic disorders.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Adipocitos Marrones , Adipocitos Blancos , Diana Mecanicista del Complejo 1 de la Rapamicina , Células 3T3-L1 , Proteínas Adaptadoras Transductoras de Señales/genética , Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Animales , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Obesidad/metabolismo , Ácido gamma-Aminobutírico/metabolismo
6.
Arch Biochem Biophys ; 704: 108885, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33878327

RESUMEN

Induction of white fat browning (beiging) and activation of brown fat has been considered a promising strategy to treat obesity and associated metabolic complications. However, the molecular mechanisms regulating brown and beige fat-mediated thermogenesis remains unclear. Our study aimed to identify genes with a hitherto unknown mechanism in the metabolic functions of adipocytes and identified family with sequence similarity 107, member A (FAM107A) as a factor that interferes with fat browning in white adipocytes. We explored physiological roles of FAM107A in cultured 3T3-L1 white adipocytes and HIB1B brown adipocytes by using FAM107A-deficient adipocytes. Significant loss in FAM107A gene functionality induced fat browning was evidenced by evaluating the gene and protein expression level of brown fat-associated markers through real-time qRT-PCR and immunoblot analysis, respectively. Deficiency of FAM107A promoted mitochondrial biogenesis and significantly upregulated core fat-browning marker proteins (PGC-1α, PRDM16, and UCP1) and beige-specific genes (Cd137, Cited1, Tbx1, and Tmem26). Furthermore, FAM107A increased adipogenesis and negatively regulated lipid metabolism in 3T3-L1 adipocytes. In addition, in-silico analysis revealed a strong interaction between FAM107A and ß3-AR based on their energy binding score. Next, mechanistic study revealed that specific knockdown of FAM107A induces browning in white adipocytes via activation of ß3-AR, AMPK and p38 MAPK-dependent signaling pathways. Our data unveiled a previously unknown mechanism of FAM107A in the regulation of lipid metabolism and identified its significant role in metabolic homeostasis. This highlighted the potential of FAM107A as a pharmacotherapeutic target in treating obesity and related metabolic disorders.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Antígenos de Diferenciación/biosíntesis , Regulación de la Expresión Génica , Termogénesis , Proteínas Supresoras de Tumor/deficiencia , Células 3T3-L1 , Animales , Metabolismo de los Lípidos/genética , Sistema de Señalización de MAP Quinasas/genética , Ratones , Proteínas Supresoras de Tumor/metabolismo
7.
Mol Cell Biochem ; 476(5): 2085-2097, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33517521

RESUMEN

BMP10 plays an essential role in regulating cardiac growth, chamber maturation, and maintaining normal expressions of several key cardiogenic factors; however, other functional roles of BMP10 in muscle remain unexplored. This study therefore undertook to investigate the roles of BMP10 in muscle physiology, using mouse-derived C2C12 myoblasts. Bmp10 silencing prevented a number of biological processes such as myogenic differentiation, glucose uptake, and lipid catabolism, whereas exogenous induction of BMP10 in C2C12 cells significantly stimulated the expression of proteins and genes involved in these processes, as well as mitochondrial biogenesis and thermogenesis, resulting in reduced lipid accumulation. A mechanistic study revealed that BMP10 stimulates myogenesis mainly via the Smad 1/5/8 signaling pathway. In conclusion, our data unveiled a previously unknown mechanism in the regulation of lipid metabolisms by BMP10 in muscle cells and identified its significant roles in systemic metabolic homeostasis, shedding light on BMP10 as a pharmacotherapeutic target to treat metabolic disorders.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Desarrollo de Músculos , Mioblastos/metabolismo , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Línea Celular , Ratones , Proteína Smad1/genética , Proteína Smad5/genética , Proteína Smad8/genética
8.
Cell Biochem Funct ; 39(4): 496-510, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33527439

RESUMEN

Bone morphogenetic protein-11 (BMP11), also known as growth differentiation factor-11 (GDF11), is implicated in skeletal development and joint morphogenesis in mammals. However, its functions in adipogenesis and energy homeostasis are mostly unknown. The present study investigates crucial roles of BMP11 in cultured 3T3-L1 white and HIB1B brown adipocytes, using Bmp11 gene depletion and pharmacological inhibition of BMP11. The silencing of Bmp11 markedly decreases the expression levels of brown-fat signature proteins and beige-specific genes in white adipocytes and significantly down-regulates the expression levels of brown fat-specific genes in brown adipocytes. The deficiency of Bmp11 reduces the expressions of lipolytic protein markers in white and brown adipocytes. Moreover, BMP11 induces browning of 3T3-L1 adipocytes via coordination of multiple signalling pathways, including mTORC1-COX2 and p38MAPK-PGC-1α as non-canonical pathways, as well as Smad1/5/8 as a canonical pathway. We believe this study is the first to provide evidence of the potential roles of BMP11 for improvement of lipid catabolism in both cultured white and brown adipocytes, as well as the effect on browning of white adipocytes. Taken together, these results demonstrate the therapeutic potential for the treatment of obesity.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Factores de Diferenciación de Crecimiento/metabolismo , Termogénesis , Animales , Proteínas Morfogenéticas Óseas/deficiencia , Proteínas Morfogenéticas Óseas/genética , Células Cultivadas , Factores de Diferenciación de Crecimiento/deficiencia , Factores de Diferenciación de Crecimiento/genética , Ratones , Mitocondrias/metabolismo
9.
Phytother Res ; 35(11): 6281-6294, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34523169

RESUMEN

Microbial fermentation of grape-skin extracts is found to synthesize anthocyanin oligomers (AO), which are more active than the monomeric anthocyanins that are effective for some metabolic diseases such as diabetes and obesity. This study investigated the functional role of AO in 3T3-L1 white adipocyte metabolism, with a focus on inducing browning. To achieve this, we determined the expressions of core genes and protein markers responsible for browning and lipid metabolism in response to AO treatment of 3T3-L1 white adipocytes. AO exposure significantly increases the expressions of beige-specific genes (Cidea, Cited1, Ppargc1α, Prdm16, Tbx1, Tmem26, and Ucp1) and brown-fat signature proteins (UCP1, PRDM16, and PGC-1α), and suppresses the expressions of lipogenic marker proteins while enhancing the protein levels of lipolysis in white adipocytes. The mechanistic study revealed stimulation of white fat browning via activation of the ß3-AR/PKA/p38 axis and ERK/CREB signaling pathway subsequent to AO treatment. In conclusion, our current findings indicate the beneficial effects of AO for the treatment of obesity with interesting properties such as regulating the browning of adipocytes and increasing thermogenic activity. Although further research based on animal models or clinical trials remains, AO treatment can bring more insights into the treatment of obesity and metabolic syndrome.


Asunto(s)
Adipocitos Blancos , Antocianinas , Células 3T3-L1 , Adipocitos Marrones , Animales , Antocianinas/farmacología , Sistema de Señalización de MAP Quinasas , Ratones , Receptores Adrenérgicos , Transducción de Señal , Termogénesis
10.
Phytother Res ; 35(2): 1113-1124, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33015893

RESUMEN

Trigonelline, a major alkaloid component of fenugreek, has been demonstrated to have several biological activities, including antidiabetic and anticancer effects. This study aimed to examine the possible application of trigonelline as an anti-obesity compound based on an investigation of its enhancement of lipid catabolism and induction of browning in white adipocytes. Trigonelline induces browning of 3T3-L1 white adipocytes by enhancing the expressions of brown-fat signature proteins and genes as well as beige-specific genes, including Cd137, Cited1, Tbx1, and Tmem26. Trigonelline also improves lipid metabolism in white adipocytes by decreasing adipogenesis and lipogenesis as well as promotes lipolysis and fatty acid oxidation. Moreover, trigonelline increases the expression of Cox4, Nrf1, and Tfam genes that are responsible for mitochondrial biogenesis. Mechanistic studies revealed that the browning effect of trigonelline in 3T3-L1 white adipocytes is mediated by activating ß3-AR and inhibiting PDE4, thereby stimulating the p38 MAPK/ATF-2 signaling pathway. Considering its high bioavailability in humans and the results of this study, trigonelline may have potential as an anti-obesity compound.


Asunto(s)
Células 3T3-L1/metabolismo , Adipocitos Marrones/efectos de los fármacos , Adipocitos Blancos/efectos de los fármacos , Alcaloides/uso terapéutico , Obesidad/tratamiento farmacológico , Alcaloides/farmacología , Animales , Humanos , Ratones
11.
Pflugers Arch ; 472(5): 583-596, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32358780

RESUMEN

The nonsteroidal anti-inflammatory drug (NSAID) ketoprofen is commonly used as a pain reliever, but its role in mediating the energy metabolism in lipids is unclear. This paper reports for the first time the critical role of ketoprofen in ameliorating white fat browning and alleviating diet-induced obesity. The effects of ketoprofen were evaluated using C57BL/6 mice fed a high fat diet and the expression levels of the target genes and proteins in the lipid metabolisms were determined by quantitative real-time PCR, immunoblot analysis, histopathology study, immunofluorescence, and molecular docking techniques. Ketoprofen induced browning in cultured 3T3-L1 white adipocytes and inguinal white adipose tissue by increasing the expression of core fat browning marker proteins as well as beige-specific genes through COX-2 activation. Ketoprofen also led to the robust activation of brown adipocytes and enhanced brown fat adipogenesis. In addition, ketoprofen elevated lipolysis, thereby increasing mitochondrial biogenesis resulting in higher fat oxidation. Furthermore, the molecular docking and mechanistic study demonstrated the recruitment of beige fat by ketoprofen via mTORC1-p38-mediated activation of the COX-2 pathway. Overall, these results indicate the unique role of ketoprofen in body weight reduction by enhancing thermogenesis, suggesting its therapeutic potential in the treatment of obesity.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Antiinflamatorios no Esteroideos/uso terapéutico , Ciclooxigenasa 2/metabolismo , Cetoprofeno/uso terapéutico , Obesidad/tratamiento farmacológico , Biogénesis de Organelos , Células 3T3-L1 , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/farmacología , Dieta Alta en Grasa/efectos adversos , Cetoprofeno/farmacología , Lipólisis , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/metabolismo , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Eur J Nutr ; 59(8): 3503-3516, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31965293

RESUMEN

PURPOSE: Modern science has given much attention to the treatment of obesity by activating brown adipose tissue (BAT) and browning of white adipose tissue (WAT). Recent studies have identified theobromine, a derivative of cocoa, as a potent natural component actively browning white fat cells. Here, we aimed to deduce the anti-obesity effect of theobromine involving phosphodiesterase (PDE) dependent-regulatory pathway in obese animal models. METHODS: For examining activity of theobromine, C57BL/6 mice were fed with high fat diet and treated with theobromine to determine the expression levels of protein markers by immunoblot analysis and gene targets by quantitative real-time PCR. Other methods used include histopathological studies, immunofluorescence and molecular docking approaches. RESULTS: Theobromine alleviated diet-induced obesity in mice by browning of iWAT and activating BAT. Further, theobromine actively interacted with PDE4D and inhibited its activity in adipose tissues and cells potentiating energy expenditure. Moreover, the regulatory action of theobromine via inhibition of PDE4D was mediated by ß3-AR signaling pathway. CONCLUSION: Altogether, the current results signifies critical role of theobromine in reducing obesity by regulation of lipid metabolism through inhibition of PDE4, indicating its potential as a major therapeutic medicinal compound.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Teobromina , Tejido Adiposo Pardo , Tejido Adiposo Blanco , Animales , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Dieta Alta en Grasa/efectos adversos , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Obesidad/tratamiento farmacológico
14.
IUBMB Life ; 70(6): 563-573, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29638041

RESUMEN

Induction of the brown adipocyte-like phenotype in white adipocytes (browning) is considered as a novel strategy to fight obesity due to the ability of brown adipocytes to increase energy expenditure. Here, we report that L-rhamnose induced browning by elevating expression levels of beige-specific marker genes, including Cd137, Cited1, Tbx1, Prdm16, Tmem26, and Ucp1, in 3T3-L1 adipocytes. Moreover, L-rhamnose markedly elevated expression levels of proteins involved in thermogenesis both in 3T3-L1 white and HIB1B brown adipocytes. L-rhamnose treatment in 3T3-L1 adipocytes also significantly elevated protein levels of p-HSL, p-AMPK, ACOX, and CPT1 as well as reduced levels of ACC, FAS, C/EBPα, and PPARγ, suggesting its possible role in enhancement of lipolysis and lipid catabolism as well as reduced adipogenesis and lipogenesis, respectively. The quick technique of efficient molecular docking provided insight into the strong binding of L-rhamnose to the fat-digesting glycine residue of ß3 -adrenergic receptor (AR), indicating strong involvement of L-rhamnose in fat metabolism. Further examination of the molecular mechanism of L-rhamnose revealed that it induced browning of 3T3-L1 adipocytes via coordination of multiple signaling pathways through ß3 -AR, SIRT1, PKA, and p-38. To the best of our knowledge, this is the first study to demonstrate that L-rhamnose plays multiple modulatory roles in the induction of white fat browning, activation of brown adipocytes, as well as promotion of lipid metabolism, thereby demonstrating its therapeutic potential for treatment of obesity. © 2018 IUBMB Life, 70(6):563-573, 2018.


Asunto(s)
Adipocitos Marrones/fisiología , Adipocitos Blancos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Ramnosa/farmacología , Células 3T3-L1 , Adipocitos Marrones/citología , Adipocitos Marrones/efectos de los fármacos , Adipocitos Blancos/citología , Adipocitos Blancos/efectos de los fármacos , Animales , Ratones
15.
IUBMB Life ; 69(7): 510-521, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28444942

RESUMEN

Our previous study demonstrated that thiodigalactoside (TDG) ameliorates obesity by targeted inhibition of galectin-1 (GAL1). Here, for the first time, we report the unexpected role of GAL1 and ATG5 inhibition by TDG in lipid metabolism. Core thermogenic marker proteins and genes were highly induced in white adipose tissue (WAT) of rats fed a high fat diet (HFD) and TDG, resulting in the significant development of brown fat-like adipocytes in inguinal WAT. TDG treatment reduced weight gain and fat mass as well as activated brown adipose tissue (BAT) in HFD-fed rats. TDG also reduced protein levels of LC3-II and increased protein levels of P62, suggesting its possible role in suppression of autophagy. Combined inhibition of GAL1 and ATG5 by TDG treatment protected rats against both HFD-induced adipogenesis as well as lipogenesis, as evidenced by suppression of CCAAT/enhancer-binding protein alpha, peroxisome proliferator-activated receptor gamma and fatty acid synthase. In conclusion, the present findings suggest that TDG plays a role in browning and lipid catabolism by combined inhibition of GAL1 and ATG5 and thus may have potential therapeutic implications in the regulation of energy homeostasis via its action in WAT. © 2017 IUBMB Life, 69(7):510-521, 2017.


Asunto(s)
Adipocitos Blancos/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/metabolismo , Galectina 1/metabolismo , Obesidad/tratamiento farmacológico , Tiogalactósidos/farmacología , Adipocitos Marrones/efectos de los fármacos , Animales , Dieta Alta en Grasa/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Obesidad/etiología , Obesidad/patología , Ratas Sprague-Dawley
16.
Eur J Nutr ; 56(7): 2329-2341, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27431894

RESUMEN

PURPOSE: Appearance of brown-like adipocytes within white adipose tissue depots (browning) is associated with improved metabolic phenotypes, and thus a wide variety of dietary agents that contribute to browning of white adipocytes are being studied. The aim of this study was to assess the browning effect of thymol, a dietary monoterpene phenolic compound, in 3T3-L1 white adipocytes. METHODS: Thymol-induced fat browning was investigated by determining expression levels of brown fat-specific genes and proteins by real-time RT-PCR and immunoblot analysis, respectively. Moreover, the molecular mechanism underlying the fat-browning effect of thymol was investigated by determining expression levels of key players responsible for browning in the presence of kinase inhibitors. RESULTS: Thymol promoted mitochondrial biogenesis and enhanced expression of a core set of brown fat-specific markers as well as increased protein levels of PPARγ, PPARδ, pAMPK, pACC, HSL, PLIN, CPT1, ACO, PGC-1α, and UCP1, suggesting its possible role in browning of white adipocytes, augmentation of lipolysis, fat oxidation, and thermogenesis, and reduction of lipogenesis. Increased expression of UCP1 and other brown fat-specific markers by thymol was tightly coordinated with activation of ß3-AR as well as AMPK, PKA, and p38 MAPK. CONCLUSION: Our findings suggest that 3T3-L1 is a potential cell model for screening browning agents. Thymol plays multiple modulatory roles in the form of inducing the brown-like phenotype as well as enhancing lipid metabolism. Thus, thymol may be explored as a potentially promising food additive for prevention of obesity.


Asunto(s)
Adipocitos/efectos de los fármacos , Monoterpenos/farmacología , Timol/farmacología , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos Marrones/efectos de los fármacos , Animales , Ligasas de Carbono-Carbono/genética , Ligasas de Carbono-Carbono/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Marcadores Genéticos , Lipólisis/efectos de los fármacos , Ratones , PPAR delta/genética , PPAR delta/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fenotipo , Esterol Esterasa/genética , Esterol Esterasa/metabolismo , Termogénesis/efectos de los fármacos , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
17.
Mol Cell Biochem ; 416(1-2): 131-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27067870

RESUMEN

Recruitment of the brown-like phenotype in white adipocytes (browning) and activation of existing brown adipocytes are currently being investigated as a means to combat obesity. Thus, a wide variety of dietary agents that contribute to browning of white adipocytes have been identified. The present study was designed to investigate the effects of cannabidiol (CBD), a major nonpsychotropic phytocannabinoid of Cannabis sativa, on induction of browning in 3T3-L1 adipocytes. CBD enhanced expression of a core set of brown fat-specific marker genes (Ucp1, Cited1, Tmem26, Prdm16, Cidea, Tbx1, Fgf21, and Pgc-1α) and proteins (UCP1, PRDM16, and PGC-1α). Increased expression of UCP1 and other brown fat-specific markers contributed to the browning of 3T3-L1 adipocytes possibly via activation of PPARγ and PI3K. In addition, CBD increased protein expression levels of CPT1, ACSL, SIRT1, and PLIN while down-regulating JNK2, SREBP1, and LPL. These data suggest possible roles for CBD in browning of white adipocytes, augmentation of lipolysis, thermogenesis, and reduction of lipogenesis. In conclusion, the current data suggest that CBD plays dual modulatory roles in the form of inducing the brown-like phenotype as well as promoting lipid metabolism. Thus, CBD may be explored as a potentially promising therapeutic agent for the prevention of obesity.


Asunto(s)
Adipocitos/metabolismo , Antígenos de Diferenciación/biosíntesis , Cannabidiol/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células 3T3-L1 , Adipocitos/citología , Animales , Lipogénesis/efectos de los fármacos , Lipólisis/efectos de los fármacos , Ratones , Termogénesis/efectos de los fármacos
18.
Mol Cell Biochem ; 415(1-2): 51-66, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26915100

RESUMEN

We previously reported that curcumin induces browning of primary white adipocytes via enhanced expression of brown adipocyte-specific genes. In this study, we attempted to identify target proteins responsible for this fat-browning effect by analyzing proteomic changes in cultured white adipocytes in response to curcumin treatment. To elucidate the role of curcumin in fat-browning, we conducted comparative proteomic analysis of primary adipocytes between control and curcumin-treated cells using two-dimensional electrophoresis combined with MALDI-TOF-MS. We also investigated fatty acid metabolic targets, mitochondrial biogenesis, and fat-browning-associated proteins using combined proteomic and network analyses. Proteomic analysis revealed that 58 protein spots from a total of 325 matched spots showed differential expression between control and curcumin-treated adipocytes. Using network analysis, most of the identified proteins were proven to be involved in various metabolic and cellular processes based on the PANTHER classification system. One of the most striking findings is that hormone-sensitive lipase (HSL) was highly correlated with main browning markers based on the STRING database. HSL and two browning markers (UCP1, PGC-1α) were co-immunoprecipitated with these markers, suggesting that HSL possibly plays a role in fat-browning of white adipocytes. Our results suggest that curcumin increased HSL levels and other browning-specific markers, suggesting its possible role in augmentation of lipolysis and suppression of lipogenesis by trans-differentiation from white adipocytes into brown adipocytes (beige).


Asunto(s)
Adipocitos/efectos de los fármacos , Biomarcadores/metabolismo , Curcumina/farmacología , Proteómica , Adipocitos/citología , Adipocitos/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Electroforesis en Gel Bidimensional , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
19.
J Cell Mol Med ; 19(5): 996-1009, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25726699

RESUMEN

Regulation of DJ1 is associated with a number of human diseases. To determine the involvement of DJ1 in progression of diabetes in a gender-dependent manner, we investigated its tissue-specific expression in streptozotocin (STZ)-induced diabetic male and female rats in this study. In animal experiments, females showed greater susceptibility towards developing diabetes because of lower insulin secretion and higher blood glucose levels as compared to male diabetic rats upon exposure to STZ. Immunoblotting confirmed sexually dimorphic regulation of DJ1 in various metabolic tissues such as the liver, pancreas and skeletal muscle. Immunofluorescence analysis revealed the location as well as reinforced the gender-dependent expression of DJ1 in hepatic tissue. Co-immunoprecipitation assay identified several interacting proteins with DJ1 whose functions were shown to be involved in various metabolic pathways viz. antioxidative and stress defence system, protein and methionine metabolism, nitrogen metabolism, urea metabolism, etc. Using GeneMANIA, a predictive web interface for gene functions, we showed for the first time that DJ1 may regulate T1DM via the JNK1 pathway, suggesting DJ1 interacts with other proteins from various metabolic pathways. We anticipate that the current data will provide insights into the aetiology of T1DM.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Metabolismo Energético , Proteínas Asociadas a Microtúbulos/metabolismo , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/genética , Femenino , Perfilación de la Expresión Génica , Immunoblotting , Hígado/metabolismo , Masculino , Microscopía Fluorescente , Proteínas Asociadas a Microtúbulos/genética , Músculo Esquelético/metabolismo , Especificidad de Órganos , Páncreas/metabolismo , Unión Proteica , Proteína Desglicasa DJ-1 , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores Sexuales
20.
J Neurochem ; 132(6): 687-702, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25359615

RESUMEN

Mitochondrial dysfunction is implicated in age-related degenerative disorders such as Alzheimer's disease (AD). Maintenance of mitochondrial dynamics is essential for regulating mitochondrial function. Aß oligomers (AßOs), the typical cause of AD, lead to mitochondrial dysfunction and neuronal loss. AßOs have been shown to induce mitochondrial fragmentation, and their inhibition suppresses mitochondrial dysfunction and neuronal cell death. Oxidative stress is one of the earliest hallmarks of AD. Cyclin-dependent kinase 5 (Cdk5) may cause oxidative stress by disrupting the antioxidant system, including Prx2. Cdk5 is also regarded as a modulator of mitochondrial fission; however, a precise mechanistic link between Cdk5 and mitochondrial dynamics is lacking. We estimated mitochondrial morphology and alterations in mitochondrial morphology-related proteins in Neuro-2a (N2a) cells stably expressing the Swedish mutation of amyloid precursor protein (APP), which is known to increase AßO production. We demonstrated that mitochondrial fragmentation by AßOs accompanies reduced mitofusin 1 and 2 (Mfn1/2) levels. Interestingly, the Cdk5 pathway, including phosphorylation of the Prx2-related oxidative stress, has been shown to regulate Mfn1 and Mfn2 levels. Furthermore, Mfn2, but not Mfn1, over-expression significantly inhibits the AßO-mediated cell death pathway. Therefore, these results indicate that AßO-mediated oxidative stress triggers mitochondrial fragmentation via decreased Mfn2 expression by activating Cdk5-induced Prx2 phosphorylation. Mitochondrial fragmentation induced by amyloid-beta oligomer (AßOs) which is generated from the Swedish mutation of amyloid precursor protein (APP) accompanies reduced Mfn1/2 levels. Interestingly, the Cdk5 pathway, including phosphorylation of the Prx2-related oxidative stress, has been shown to regulate Mfn1/2. Furthermore, Mfn2 over-expression significantly inhibits the AßO-mediated neuronal cells death pathway, but not Mfn1 over-expression. Therefore, these results indicate that AßO-mediated oxidative stress triggers mitochondrial fragmentation via decreased Mfn2 expression by activating Cdk5-induced Prx2 phosphorylation. ATP, adenosine triphosphate; Bax, Bcl-2-associated X protein; Bcl-2, B-cell lymphoma 2; Cdk5, Cyclin-dependent kinase; Cyt C, cytochrome C; Mfn2, mitofusin 2; Prx2, peroxiredoxin 2; ROS, reactive oxygen species.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Quinasa 5 Dependiente de la Ciclina/metabolismo , GTP Fosfohidrolasas/biosíntesis , Mitocondrias/metabolismo , Neuronas/metabolismo , Estrés Oxidativo/fisiología , Péptidos beta-Amiloides/metabolismo , Animales , Línea Celular Tumoral , GTP Fosfohidrolasas/deficiencia , Ratones , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA