Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Transl Med ; 22(1): 748, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39118142

RESUMEN

BACKGROUND: Sjögren's Syndrome (SS) is a rare chronic autoimmune disorder primarily affecting adult females, characterized by chronic inflammation and salivary and lacrimal gland dysfunction. It is often associated with systemic lupus erythematosus, rheumatoid arthritis and kidney disease, which can lead to increased mortality. Early diagnosis is critical, but traditional methods for diagnosing SS, mainly through histopathological evaluation of salivary gland tissue, have limitations. METHODS: The study used 100 labial gland biopsy, creating whole-slide images (WSIs) for analysis. The proposed model, named Cell-tissue-graph-based pathological image analysis model (CTG-PAM) and based on graph theory, characterizes single-cell feature, cell-cell feature, and cell-tissue feature. Building upon these features, CTG-PAM achieves cellular-level classification, enabling lymphocyte recognition. Furthermore, it leverages connected component analysis techniques in the cell graph structure to perform SS diagnosis based on lymphocyte counts. FINDINGS: CTG-PAM outperforms traditional deep learning methods in diagnosing SS. Its area under the receiver operating characteristic curve (AUC) is 1.0 for the internal validation dataset and 0.8035 for the external test dataset. This indicates high accuracy. The sensitivity of CTG-PAM for the external dataset is 98.21%, while the accuracy is 93.75%. In comparison, the sensitivity and accuracy for traditional deep learning methods (ResNet-50) are lower. The study also shows that CTG-PAM's diagnostic accuracy is closer to skilled pathologists compared to beginners. INTERPRETATION: Our findings indicate that CTG-PAM is a reliable method for diagnosing SS. Additionally, CTG-PAM shows promise in enhancing the prognosis of SS patients and holds significant potential for the differential diagnosis of both non-neoplastic and neoplastic diseases. The AI model potentially extends its application to diagnosing immune cells in tumor microenvironments.


Asunto(s)
Síndrome de Sjögren , Síndrome de Sjögren/diagnóstico , Síndrome de Sjögren/patología , Humanos , Femenino , Estudios de Cohortes , Curva ROC , Procesamiento de Imagen Asistido por Computador/métodos , Persona de Mediana Edad , Aprendizaje Profundo , Área Bajo la Curva , Adulto , Automatización
2.
Sci Adv ; 10(26): eadn5229, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38924414

RESUMEN

There is a regional preference around lymph nodes (LNs) for adipose beiging. Here, we show that local LN removal within inguinal white adipose tissue (iWAT) greatly impairs cold-induced beiging, and this impairment can be restored by injecting M2 macrophages or macrophage-derived C-C motif chemokine (CCL22) into iWAT. CCL22 injection into iWAT effectively promotes iWAT beiging, while blocking CCL22 with antibodies can prevent it. Mechanistically, the CCL22 receptor, C-C motif chemokine receptor 4 (CCR4), within eosinophils and its downstream focal adhesion kinase/p65/interleukin-4 signaling are essential for CCL22-mediated beige adipocyte formation. Moreover, CCL22 levels are inversely correlated with body weight and fat mass in mice and humans. Acute elevation of CCL22 levels effectively prevents diet-induced body weight and fat gain by enhancing adipose beiging. Together, our data identify the CCL22-CCR4 axis as an essential mediator for LN-controlled adaptive thermogenesis and highlight its potential to combat obesity and its associated complications.


Asunto(s)
Tejido Adiposo Blanco , Quimiocina CCL22 , Metabolismo Energético , Ganglios Linfáticos , Macrófagos , Termogénesis , Animales , Femenino , Humanos , Masculino , Ratones , Adipocitos Beige/metabolismo , Tejido Adiposo Blanco/metabolismo , Quimiocina CCL22/metabolismo , Eosinófilos/metabolismo , Ganglios Linfáticos/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Obesidad/metabolismo , Receptores CCR4/metabolismo , Transducción de Señal
3.
Food Funct ; 15(9): 5000-5011, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38618651

RESUMEN

The anti-obesity effect of conjugated linoleic acid (CLA) has been well elucidated, but whether CLA affects fat deposition by regulating intestinal dietary fat absorption remains largely unknown. Thus, this study aimed to investigate the effects of CLA on intestinal fatty acid uptake and chylomicron formation and explore the possible underlying mechanisms. We found that CLA supplementation reduced the intestinal fat absorption in HFD (high fat diet)-fed mice accompanied by the decreased serum TG level, increased fecal lipids and decreased intestinal expression of ApoB48 and MTTP. Correspondingly, c9, t11-CLA, but not t10, c12-CLA induced the reduction of fatty acid uptake and TG content in PA (palmitic acid)-treated MODE-K cells. In the mechanism of fatty acid uptake, c9, t11-CLA inhibited the binding of CD36 with palmitoyltransferase DHHC7, thus leading to the decreases of CD36 palmitoylation level and localization on the cell membrane of the PA-treated MODE-K cells. In the mechanism of chylomicron formation, c9, t11-CLA inhibited the formation of the CD36/FYN/LYN complex and the activation of the ERK pathway in the PA-treated MODE-K cells. In in vivo verification, CLA supplementation reduced the DHHC7-mediated total and cell membrane CD36 palmitoylation and suppressed the formation of the CD36/FYN/LYN complex and the activation of the ERK pathway in the jejunum of HFD-fed mice. Altogether, these data showed that CLA reduced intestinal fatty acid uptake and chylomicron formation in HFD-fed mice associated with the inhibition of DHHC7-mediated CD36 palmitoylation and the downstream ERK pathway.


Asunto(s)
Quilomicrones , Dieta Alta en Grasa , Sistema de Señalización de MAP Quinasas , Animales , Masculino , Ratones , Aciltransferasas/metabolismo , Aciltransferasas/genética , Antígenos CD36/metabolismo , Antígenos CD36/genética , Quilomicrones/metabolismo , Dieta Alta en Grasa/efectos adversos , Ácidos Grasos/metabolismo , Absorción Intestinal/efectos de los fármacos , Ácidos Linoleicos Conjugados/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Endogámicos C57BL
4.
Int J Biol Macromol ; 264(Pt 2): 130782, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38471613

RESUMEN

Vascular endothelial growth factor B (VEGFB) has been well demonstrated to play a crucial role in regulating vascular function by binding to the VEGF receptors (VEGFRs). However, the specific role of VEGFB and VEGFRs in pubertal mammary gland development remains unclear. In this study, we observed that blocking the VEGF receptors with Axitinib suppressed the pubertal mammary gland development. Meanwhile, the proliferation of mammary epithelial cells (HC11) was repressed by blocking the VEGF receptors with Axitinib. Additionally, knockdown of VEGFR1 rather than VEGFR2 and NRP1 elicited the inhibition of HC11 proliferation, suggesting the essential role of VEGFR1 during this process. Furthermore, Axitinib or VEGFR1 knockdown led to the inhibition of the PI3K/Akt pathway. However, the inhibition of HC11 proliferation induced by Axitinib and or VEGFR1 knockdown was eliminated by the Akt activator SC79, indicating the involvement of the PI3K/Akt pathway. Finally, the knockdown of VEGFB and VEGFR1 suppressed the pubertal development of mice mammary gland with the inhibition of the PI3K/Akt pathway. In summary, the results showed that knockdown of the VEGFB/VEGFR1 signaling suppresses pubertal mammary gland development of mice via the inhibition of the PI3K/Akt pathway, which provides a new target for the regulation of pubertal mammary gland development.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Factor B de Crecimiento Endotelial Vascular , Animales , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Axitinib/farmacología , Receptores de Factores de Crecimiento Endotelial Vascular , Proliferación Celular
5.
Entropy (Basel) ; 26(2)2024 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-38392410

RESUMEN

The two-dimensional sample entropy marks a significant advance in evaluating the regularity and predictability of images in the information domain. Unlike the direct computation of sample entropy, which incurs a time complexity of O(N2) for the series with N length, the Monte Carlo-based algorithm for computing one-dimensional sample entropy (MCSampEn) markedly reduces computational costs by minimizing the dependence on N. This paper extends MCSampEn to two dimensions, referred to as MCSampEn2D. This new approach substantially accelerates the estimation of two-dimensional sample entropy, outperforming the direct method by more than a thousand fold. Despite these advancements, MCSampEn2D encounters challenges with significant errors and slow convergence rates. To counter these issues, we have incorporated an upper confidence bound (UCB) strategy in MCSampEn2D. This strategy involves assigning varied upper confidence bounds in each Monte Carlo experiment iteration to enhance the algorithm's speed and accuracy. Our evaluation of this enhanced approach, dubbed UCBMCSampEn2D, involved the use of medical and natural image data sets. The experiments demonstrate that UCBMCSampEn2D achieves a 40% reduction in computational time compared to MCSampEn2D. Furthermore, the errors with UCBMCSampEn2D are only 30% of those observed in MCSampEn2D, highlighting its improved accuracy and efficiency.

6.
Rev Sci Instrum ; 94(4)2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38081251

RESUMEN

A cold collision between atoms and molecules (<1 K) is one of the hot research fields in atomic and molecular physics. At low temperatures, the number of partial waves participating in the collision process decreases dramatically, and quantum phenomena start to emerge. The reaction is often dominated by quantum tunneling, and pronounced resonances can exist on collision cross sections. Here, we report on an apparatus designed for studying cold collisions between metastable noble gas atoms and alkali atoms. Our apparatus features a combined Magneto-Optical-Trap (MOT) and velocity map imaging (VMI) system. The center of a Rb MOT is overlapped with the VMI system. Cold Kr* atoms are launched toward the Rb atoms to induce Kr* + Rb reactions. The collision energy between the two species can be varied from 100 mK to 20 K. With this setup, we are planning to explore the quantum phenomena in Kr* + Rb cold collisions, including the shape resonance and stereodynamics in the reaction.

7.
Mol Metab ; 73: 101747, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37279828

RESUMEN

OBJECTIVE: Brown adipose tissue (BAT) plays a crucial role in regulating non-shivering thermogenesis under cold exposure. Proline hydroxylases (PHDs) were found to be involved in adipocyte differentiation and lipid deposition. However, the effects of PHDs on regulatory mechanisms of BAT thermogenesis are not fully understood. METHODS: We detected the expression of PHDs in different adipose tissues by using immunoblotting and real-time PCR. Further, immunoblotting, real-time PCR, and immunostaining were performed to determine the correlation between proline hydroxylase 2 (PHD2) and UCP1 expression. Inhibitor of PHDs and PHD2-sgRNA viruses were used to construct the PHD2-deficiency model in vivo and in vitro to investigate the impacts of PHD2 on BAT thermogenesis. Afterward, the interaction between UCP1 and PHD2 and the hydroxylation modification level of UCP1 were verified by Co-IP assays and immunoblotting. Finally, the effect of specific proline hydroxylation on the expression/activity of UCP1 was further confirmed by site-directed mutation of UCP1 and mass spectrometry analysis. RESULTS: PHD2, but not PHD1 and PHD3, was highly enriched in BAT, colocalized, and positively correlated with UCP1. Inhibition or knockdown of PHD2 significantly suppressed BAT thermogenesis under cold exposure and aggravated obesity of mice fed HFD. Mechanistically, mitochondrial PHD2 bound to UCP1 and regulated the hydroxylation level of UCP1, which was enhanced by thermogenic activation and attenuated by PHD2 knockdown. Furthermore, PHD2-dependent hydroxylation of UCP1 promoted the expression and stability of UCP1 protein. Mutation of the specific prolines (Pro-33, 133, and 232) in UCP1 significantly mitigated the PHD2-elevated UCP1 hydroxylation level and reversed the PHD2-increased UCP1 stability. CONCLUSIONS: This study suggested an important role for PHD2 in BAT thermogenesis regulation by enhancing the hydroxylation of UCP1.


Asunto(s)
Obesidad , Prolil Hidroxilasas , Animales , Ratones , Tejido Adiposo Pardo/metabolismo , Hidroxilación , Obesidad/metabolismo , Prolina/metabolismo , Prolil Hidroxilasas/metabolismo , Termogénesis/fisiología
8.
Nat Commun ; 14(1): 2731, 2023 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-37169793

RESUMEN

A potential therapeutic target to curb obesity and diabetes is thermogenic beige adipocytes. However, beige adipocytes quickly transition into white adipocytes upon removing stimuli. Here, we define the critical role of cyclin dependent kinase inhibitor 2A (Cdkn2a) as a molecular pedal for the beige-to-white transition. Beige adipocytes lacking Cdkn2a exhibit prolonged lifespan, and male mice confer long-term metabolic protection from diet-induced obesity, along with enhanced energy expenditure and improved glucose tolerance. Mechanistically, Cdkn2a promotes the expression and activity of beclin 1 (BECN1) by directly binding to its mRNA and its negative regulator BCL2 like 1 (BCL2L1), activating autophagy and accelerating the beige-to-white transition. Reactivating autophagy by pharmacological or genetic methods abolishes beige adipocyte maintenance induced by Cdkn2a ablation. Furthermore, hyperactive BECN1 alone accelerates the beige-to-white transition in mice and human. Notably, both Cdkn2a and Becn1 exhibit striking positive correlations with adiposity. Hence, blocking Cdkn2a-mediated BECN1 activity holds therapeutic potential to sustain beige adipocytes in treating obesity and related metabolic diseases.


Asunto(s)
Adipocitos Beige , Tejido Adiposo Beige , Obesidad , Animales , Humanos , Masculino , Ratones , Adipocitos Beige/metabolismo , Adipocitos Blancos/metabolismo , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Blanco/metabolismo , Adiposidad/genética , Adiposidad/fisiología , Obesidad/genética , Obesidad/metabolismo , Termogénesis
9.
Cell Death Dis ; 14(1): 29, 2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36642732

RESUMEN

Obesity is strongly associated with metabolic diseases, which have become a global health problem. Exploring the underlying mechanism of adipogenesis is crucial for the treatment of excess white fat. Oncogene YBX1 is a multifunctional DNA- and RNA-binding protein that regulates brown adipogenesis. However, the role of YBX1 in white adipogenesis and adipose tissue expansion remains unknown. Here, we showed that YBX1 deficiency inhibited murine and porcine adipocyte differentiation. YBX1 positively regulated adipogenesis through promoting ULK1- and ULK2-mediated autophagy. Mechanistically, we identified YBX1 serves as a 5-methylcytosine (m5C)-binding protein directly targeting m5C-containing Ulk1 mRNA by using RNA immunoprecipitation. RNA decay assay further proved that YBX1 upregulated ULK1 expression though stabilizing its mRNA. Meanwhile, YBX1 promoted Ulk2 transcription and expression as a transcription factor, thereby enhancing autophagy and adipogenesis. Importantly, YBX1 overexpression in white fat enhanced ULK1/ULK2-mediated autophagy and promoted adipose tissue expansion in mice. Collectively, these findings unveil the post-transcriptional and transcriptional mechanism and functional importance of YBX1 in autophagy and adipogenesis regulation, providing an attractive molecular target for therapies of obesity and metabolic diseases.


Asunto(s)
Adipogénesis , Autofagia , Regulación de la Expresión Génica , Factores de Transcripción , Animales , Ratones , Adipogénesis/genética , Autofagia/genética , Obesidad/genética , ARN Mensajero , Porcinos , Factores de Transcripción/genética
10.
Cell Mol Life Sci ; 79(9): 481, 2022 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-35962235

RESUMEN

Although 5-methylcytosine (m5C) has been identified as a novel and abundant mRNA modification and associated with energy metabolism, its regulation function in adipose tissue and skeletal muscle is still limited. This study aimed at investigating the effect of mRNA m5C on adipogenesis and myogenesis using Jinhua pigs (J), Yorkshire pigs (Y) and their hybrids Yorkshire-Jinhua pigs (YJ). We found that Y grow faster than J and YJ, while fatness-related characteristics observed in Y were lower than those of J and YJ. Besides, total mRNA m5C levels and expression rates of NSUN2 were higher both in backfat layer (BL) and longissimus dorsi muscle (LDM) of Y compared to J and YJ, suggesting that higher mRNA m5C levels positively correlate with lower fat and higher muscle mass. RNA bisulfite sequencing profiling of m5C revealed tissue-specific and dynamic features in pigs. Functionally, hyper-methylated m5C-containing genes were enriched in pathways linked to impaired adipogenesis and enhanced myogenesis. In in vitro, m5C inhibited lipid accumulation and promoted myogenic differentiation. Furthermore, YBX2 and SMO were identified as m5C targets. Mechanistically, YBX2 and SMO mRNAs with m5C modification were recognized and exported into the cytoplasm from the nucleus by ALYREF, thus leading to increased YBX2 and SMO protein expression and thereby inhibiting adipogenesis and promoting myogenesis, respectively. Our work uncovered the critical role of mRNA m5C in regulating adipogenesis and myogenesis via ALYREF-m5C-YBX2 and ALYREF-m5C-SMO manners, providing a potential therapeutic target in the prevention and treatment of obesity, skeletal muscle dysfunction and metabolic disorder diseases.


Asunto(s)
Adipogénesis , Proteínas de Unión al ARN , Adipogénesis/genética , Animales , Desarrollo de Músculos/genética , Transporte de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Porcinos
11.
FASEB J ; 36(3): e22219, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35195911

RESUMEN

Promoting the thermogenic function of brown adipose tissue (BAT) is a promising strategy to combat obesity and metabolic disorders. While much is known about the transcriptional regulation of BAT activation, however, the underlying mechanism of post-transcriptional control by RNA binding proteins remains largely unknown. Here, we found that RNA binding protein Y-box binding protein 1 (YBX1) expression was abundant in BAT and induced by cold exposure and a ß-adrenergic agonist in mice. Loss-of-function experiments showed that YBX1 deficiency inhibited mouse primary brown adipocyte differentiation and thermogenic function. Further study showed that YBX1 positively regulates thermogenesis through enhancing mitophagy. Mechanistically, RNA immunoprecipitation identified that YBX1 directly targeted the transcripts of PTEN-induced kinase 1 (Pink1) and parkin RBR E3 ubiquitin-protein ligase (Prkn), two key regulators of mitophagy. RNA decay assay proved that loss of YBX1 decreased mRNA stability of Pink1 and Prkn, leading to reduced protein expression, thereby alleviating mitophagy and inhibiting thermogenic program. Importantly, in vivo experiments demonstrated that YBX1 overexpression in BAT promoted thermogenesis and mitophagy in mice. Collectively, our results reveal novel insight into the molecular mechanism of YBX1 in post-transcriptional regulation of PINK1/PRKN-mediated mitophagy and highlight the critical role of YBX1 in brown adipogenesis and thermogenesis.


Asunto(s)
Adipogénesis , Mitofagia , Termogénesis , Factores de Transcripción/metabolismo , Adipocitos Marrones/citología , Adipocitos Marrones/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas/metabolismo , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/metabolismo
12.
BMC Biol ; 20(1): 39, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35135551

RESUMEN

BACKGROUND: Obesity leads to a decline in the exercise capacity of skeletal muscle, thereby reducing mobility and promoting obesity-associated health risks. Dietary intervention has been shown to be an important measure to regulate skeletal muscle function, and previous studies have demonstrated the beneficial effects of docosahexaenoic acid (DHA; 22:6 ω-3) on skeletal muscle function. At the molecular level, DHA and its metabolites were shown to be extensively involved in regulating epigenetic modifications, including DNA methylation, histone modifications, and small non-coding microRNAs. However, whether and how epigenetic modification of mRNA such as N6-methyladenosine (m6A) mediates DHA regulation of skeletal muscle function remains unknown. Here, we analyze the regulatory effect of DHA on skeletal muscle function and explore the involvement of m6A mRNA modifications in mediating such regulation. RESULTS: DHA supplement prevented HFD-induced decline in exercise capacity and conversion of muscle fiber types from slow to fast in mice. DHA-treated myoblasts display increased mitochondrial biogenesis, while slow muscle fiber formation was promoted through DHA-induced expression of PGC1α. Further analysis of the associated molecular mechanism revealed that DHA enhanced expression of the fat mass and obesity-associated gene (FTO), leading to reduced m6A levels of DNA damage-induced transcript 4 (Ddit4). Ddit4 mRNA with lower m6A marks could not be recognized and bound by the cytoplasmic m6A reader YTH domain family 2 (YTHDF2), thereby blocking the decay of Ddit4 mRNA. Accumulated Ddit4 mRNA levels accelerated its protein translation, and the consequential increased DDIT4 protein abundance promoted the expression of PGC1α, which finally elevated mitochondria biogenesis and slow muscle fiber formation. CONCLUSIONS: DHA promotes mitochondrial biogenesis and skeletal muscle fiber remodeling via FTO/m6A/DDIT4/PGC1α signaling, protecting against obesity-induced decline in skeletal muscle function.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Ácidos Docosahexaenoicos , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Animales , Dieta , Ácidos Docosahexaenoicos/metabolismo , Ácidos Docosahexaenoicos/farmacología , Ratones , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Obesidad , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/farmacología , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo
13.
Gene ; 818: 146224, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35085712

RESUMEN

OBJECTIVE: Adipose dysfunction affects the secretion of adipokines and mediates the hepatic physiological changes. Fat mass and obesity associated protein (FTO) plays a crucial part in fat deposition but the crosstalk between FTO-mediated secretion of adipokines and hepatic steatosis is not clear. METHODS: Firstly, adipose-selective FTO knockout (FTOAKO) and control (FTOflox/flox) mice were induced by high fat diet (HFD). Then qRT-PCR assay was performed to analyze the expressions of hepatic lipid metabolism genes and adipocytokines gene of inguinal white adipose tissue (iWAT) and epididymal white adipose tissue (eWAT). Afterwards, 3T3-L1 cells were knocked out IL-6 and co-cultured with AML12 cells (3T3-L1 siIL-6/AML12) and the expressions of hepatic lipid lipolysis genes were measured. Finally, we detected the hepatic lipid metabolism genes expressions in AML12 cells with the medium from 3T3-L1 cells or IL-6 treatment. RESULTS: FTOAKO effectively alleviated HFD-induced hepatic steatosis in mice and improved the transcription level of genes involved in hepatic lipolysis. Further investigation demonstrated that FTO knockout increased level of IL-6 in adipose tissues and 3T3-L1 cells. Compared to 3T3-L1/AML12, our results showed lipolysis-related genes expressions were dramatically inhibited in 3T3-L1 siIL-6/AML12. Finally, both depletion of FTO in adipocytes and IL-6 supplement led to increased lipolysis genes expressions in AML12 cells. CONCLUSIONS: FTO knockout in adipose tissue alleviated hepatic steatosis via targeting adipocyte-derived IL-6.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/deficiencia , Hígado Graso/metabolismo , Hígado Graso/patología , Interleucina-6/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Animales , Dieta Alta en Grasa , Regulación de la Expresión Génica , Lipólisis/genética , Hígado/metabolismo , Hígado/patología , Ratones Noqueados
14.
EMBO Rep ; 22(11): e52348, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34569703

RESUMEN

Obesity mainly results from a chronic energy imbalance. Promoting browning of white adipocytes is a promising strategy to enhance energy expenditure and combat obesity. N6-methyladenosine (m6A), the most abundant mRNA modification in eukaryotes, plays an important role in regulating adipogenesis. However, whether m6A regulates white adipocyte browning was unknown. Here, we report that adipose tissue-specific deletion of Fto, an m6A demethylase, predisposes mice to prevent high-fat diet (HFD)-induced obesity by enhancing energy expenditure. Additionally, deletion of FTO in vitro promotes thermogenesis and white-to-beige adipocyte transition. Mechanistically, FTO deficiency increases the m6A level of Hif1a mRNA, which is recognized by m6A-binding protein YTHDC2, facilitating mRNA translation and increasing HIF1A protein abundance. HIF1A activates the transcription of thermogenic genes, including Ppaggc1a, Prdm16, and Pparg, thereby promoting Ucp1 expression and the browning process. Collectively, these results unveil an epigenetic mechanism by which m6A-facilitated HIF1A expression controls browning of white adipocytes and thermogenesis, providing a potential target to counteract obesity and metabolic disease.


Asunto(s)
Tejido Adiposo Beige , Tejido Adiposo Blanco , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Adenosina/análogos & derivados , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético , Metilación , Ratones , Ratones Endogámicos C57BL , Termogénesis
15.
RNA Biol ; 18(sup2): 711-721, 2021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34570675

RESUMEN

5-Methylcytosine (m5C) is a type of RNA modification that exists in tRNAs and rRNAs and was recently found in mRNA. Although mRNA m5C modification has been reported to regulate diverse biological process, its function in adipogenesis remains unknown. Here, we demonstrated that knockdown of NOL1/NOP2/Sun domain family member 2 (NSUN2), a m5C methyltransferase, increased lipid accumulation of 3T3-L1 preadipocytes through accelerating cell cycle progression during mitotic clonal expansion (MCE) at the early stage of adipogenesis. Mechanistically, we proved that NSUN2 directly targeted cyclin-dependent kinase inhibitor 1A (CDKN1A) mRNA, a key inhibitory regulator of cell cycle progression, and upregulated its protein expression in an m5C-dependent manner. Further study identified that CDKN1A was the target of Aly/REF export factor (ALYREF), a reader of m5C modified mRNA. Upon NSUN2 deficiency, the recognition of CDKN1A mRNA by ALYREF was suppressed, resulting in the decrease of CDKN1A mRNA shuttling from nucleus to cytoplasm. Thereby, the translation of CDKN1A was reduced, leading to the acceleration of cell cycle and the promotion of adipogenesis. Together, these findings unveiled an important function and mechanism of the m5C modification on adipogenesis by controlling cell cycle progression, providing a potential therapeutic target to prevent obesity.


Asunto(s)
5-Metilcitosina , Adipogénesis/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Biosíntesis de Proteínas , ARN Mensajero/genética , Células 3T3-L1 , 5-Metilcitosina/metabolismo , Animales , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación de la Expresión Génica , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Biosíntesis de Proteínas/genética , Transporte de ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo
16.
EMBO Rep ; 22(5): e52146, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33880847

RESUMEN

Obesity has become a major health problem that has rapidly prevailed over the past several decades worldwide. Curcumin, a natural polyphenolic compound present in turmeric, has been shown to have a protective effect on against obesity and metabolic diseases. However, its underlying mechanism remains largely unknown. Here, we show that the administration of curcumin significantly prevents HFD-induced obesity and decreases the fat mass of the subcutaneous inguinal WAT (iWAT) and visceral epididymal WAT (eWAT) in mice. Mechanistically, curcumin inhibits adipogenesis by reducing the expression of AlkB homolog 5 (ALKHB5), an m6 A demethylase, which leads to higher m6 A-modified TNF receptor-associated factor 4 (TRAF4) mRNA. TRAF4 mRNA with higher m6 A level is recognized and bound by YTHDF1, leading to enhanced translation of TRAF4. TRAF4, acting as an E3 RING ubiquitin ligase, promotes degradation of adipocyte differentiation regulator PPARγ by a ubiquitin-proteasome pathway thereby inhibiting adipogenesis. Thus, m6 A-dependent TRAF4 expression upregulation by ALKBH5 and YTHDF1 contributes to curcumin-induced obesity prevention. Our findings provide mechanistic insights into how m6 A is involved in the anti-obesity effect of curcumin.


Asunto(s)
Curcumina , Factor 4 Asociado a Receptor de TNF , Células 3T3-L1 , Adipogénesis , Animales , Curcumina/farmacología , Dieta Alta en Grasa , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/genética , Factor 4 Asociado a Receptor de TNF/genética , Factor 4 Asociado a Receptor de TNF/metabolismo , Ubiquitinación
17.
Obes Rev ; 22(2): e13124, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32935469

RESUMEN

Obesity, defined as excessive fat accumulation, is strongly associated with metabolic diseases and cancer, and its prevalence is rising worldwide. Thus, understanding the molecular mechanism of adipogenesis is of fundamental significance. Epigenetic modifications play important roles in regulating adipogenesis. N6 -methyladenosine (m6 A), the most prevalent and abundant mRNA modification in eukaryotic cells, modulates multiple aspects of RNA metabolism, including mRNA stability, translation, splicing and export. Recent studies indicate that m6 A methylation plays important roles in modulating gene expression and signal pathways in various physiologic processes and diseases. Notably, the significant function and regulatory mechanisms of m6 A in adipogenesis are now emerging. In this review, we summarize recent studies that elucidate the vital roles of m6 A modifications in regulating adipogenesis and adipose tissue expansion. Furthermore, we highlight the nutritional regulation of m6 A methylation and adipogenesis, which may prove a novel therapeutic strategy to fight against obesity.


Asunto(s)
Adenosina/análogos & derivados , Adipogénesis , Tejido Adiposo/crecimiento & desarrollo , Epigénesis Genética , Procesamiento Postranscripcional del ARN , Adenosina/química , Adipogénesis/genética , Humanos , Metilación
18.
Autophagy ; 16(7): 1221-1235, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31451060

RESUMEN

N: 6-methyladenosine (m6A), the most abundant internal modification on mRNAs in eukaryotes, play roles in adipogenesis. However, the underlying mechanism remains largely unclear. Here, we show that m6A plays a critical role in regulating macroautophagy/autophagy and adipogenesis through targeting Atg5 and Atg7. Mechanistically, knockdown of FTO, a well-known m6A demethylase, decreased the expression of ATG5 and ATG7, leading to attenuation of autophagosome formation, thereby inhibiting autophagy and adipogenesis. We proved that FTO directly targeted Atg5 and Atg7 transcripts and mediated their expression in an m6A-dependent manner. Further study identified that Atg5 and Atg7 were the targets of YTHDF2 (YTH N6-methyladenosine RNA binding protein 2). Upon FTO silencing, Atg5 and Atg7 transcripts with higher m6A levels were captured by YTHDF2, which resulted in mRNA degradation and reduction of protein expression, thus alleviating autophagy and adipogenesis. Furthermore, we generated an adipose-selective fto knockout mouse and find that FTO deficiency decreased white fat mass and impairs ATG5- and ATG7-dependent autophagy in vivo. Together, these findings unveil the functional importance of the m6A methylation machinery in autophagy and adipogenesis regulation, which expands our understanding of such interplay that is essential for development of therapeutic strategies in the prevention and treatment of obesity. ABBREVIATIONS: 3-MA: 3-methyladenine; ACTB: actin, beta; ATG: autophagy-related; Baf A1: bafilomycin A1; CEBPA: CCAAT/enhancer binding protein (C/EBP), alpha; CEBPB: CCAAT/enhancer binding protein (C/EBP), beta; FABP4: fatty acid binding protein 4, adipocyte; FTO: fat mass and obesity associated; HFD: high-fat diet; LC-MS/MS: liquid chromatography-tandem mass spectrometry; MAP1LC3B/LC3: microtubule-associated protein 1 light chain 3 beta; m6A: N6-methyladenosine; MEFs: mouse embryo fibroblasts; MeRIP-qPCR: methylated RNA immunoprecipitation-qPCR; PPARG: peroxisome proliferator activated receptor gamma; RIP: RNA-immunoprecipitation; SAT: subcutaneous adipose tissue; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; ULK1: unc-51 like kinase 1; VAT: visceral adipose tissue; WAT: white adipose tissue; YTHDF: YTH N6-methyladenosine RNA binding protein.


Asunto(s)
Adenosina/análogos & derivados , Adipogénesis , Proteína 5 Relacionada con la Autofagia/metabolismo , Proteína 7 Relacionada con la Autofagia/metabolismo , Autofagia , Células 3T3-L1 , Adenosina/metabolismo , Adipocitos/metabolismo , Adipocitos/ultraestructura , Adiposidad , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Técnicas de Silenciamiento del Gen , Metilación , Ratones , Ratones Noqueados , Modelos Biológicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo
19.
RNA Biol ; 16(12): 1785-1793, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31434544

RESUMEN

Obesity is becoming a global problem. Research into the detailed mechanism of adipocyte development is crucial for the treatment of excess fat. Zinc finger protein 217 plays roles in adipogenesis. However, the underlying mechanism remains unclear. Here, we demonstrated that ZFP217 knockdown prevented the mitotic clonal expansion process and caused adipogenesis inhibition. Depletion of ZFP217 increased the expression of the m6A methyltransferase METTL3, which upregulated the m6A level of cyclin D1 mRNA. METTL3 knockdown rescued the siZFP217-inhibited MCE and promoted CCND1 expression. YTH domain family 2 recognized and degraded the methylated CCND1 mRNA, leading to the downregulation of CCND1. Consequently, cell-cycle progression was blocked, and adipogenesis was inhibited. YTHDF2 knockdown relieved siZFP217-inhibited adipocyte differentiation. These findings reveal that ZFP217 knockdown-induced adipogenesis inhibition was caused by CCND1, which was mediated by METTL3 and YTHDF2 in an m6A-dependent manner. We have provided novel insight into the underlying molecular mechanisms by which m6A methylation is involved in the ZFP217 regulation of adipogenesis.


Asunto(s)
Adenosina/análogos & derivados , Adipocitos/metabolismo , Adipogénesis/genética , Metiltransferasas/genética , Transactivadores/genética , Células 3T3-L1 , Adenosina/metabolismo , Adipocitos/citología , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular , Células Clonales , Ciclina D1/genética , Ciclina D1/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Metiltransferasas/metabolismo , Ratones , Mitosis , PPAR gamma/genética , PPAR gamma/metabolismo , Plásmidos/química , Plásmidos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Transactivadores/antagonistas & inhibidores , Transactivadores/metabolismo , Transfección
20.
Biochim Biophys Acta Gene Regul Mech ; 1862(8): 796-806, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31295563

RESUMEN

N6-methyladenosine (m6A), the most abundant internal mRNA modification in eukaryotes, plays a vital role in regulating adipogenesis. However, its underlying mechanism remains largely unknown. Here, we reveal that deletion of m6A demethylase FTO in porcine and mouse preadipocytes inhibits adipogenesis through JAK2-STAT3-C/EBPß signaling. Mechanistically, FTO deficiency suppresses JAK2 expression and STAT3 phosphorylation, leading to attenuated transcription of C/EBPß, which is essential for the early stage of adipocyte differentiation. Using dual-luciferase assay, we validate that knockdown of FTO reduces expression of JAK2 in an m6A-dependent manner. Furthermore, we find that m6A "reader" protein YTHDF2 directly targets m6A-modified transcripts of JAK2 and accelerates mRNA decay, which results in decreased JAK2 expression and inactivated JAK2-STAT3-C/EBPß signaling, thereby inhibiting adipogenesis. Collectively, our results provide a novel insight into the molecular mechanism of m6A methylation in post-transcriptional regulation of JAK2-STAT3-C/EBPß signaling axis and highlight the crucial role of m6A modification and its modulators in adipogenesis.


Asunto(s)
Adenosina/análogos & derivados , Adipogénesis , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Janus Quinasa 2/genética , Factor de Transcripción STAT3/metabolismo , Células 3T3-L1 , Adenosina/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Animales , Diferenciación Celular , Eliminación de Gen , Regulación de la Expresión Génica , Ratones , Fosforilación , Estabilidad del ARN , Proteínas de Unión al ARN , Transducción de Señal , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA