Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Adv Nurs ; 2024 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-39092879

RESUMO

AIM: To critically evaluate missed care measurement approaches and their application in long-term aged care (LTAC) settings. DESIGN: Systematic review using Tawfik's guideline. DATA SOURCES: PubMed, Scopus, Web of Science, CINAHL and ProQuest were searched. Supplemental searching was from reference lists of retrieved records, first authors' ORCID homepages and Google advanced search for grey literature. Search limitations were English language, published between 1 January 2001 and 31 December 2022. REVIEW METHOD: COVIDENCE was utilized for screening, data extraction and quality appraisal. JBI Critical Appraisal Tools and COSMIN Risk of Bias Tool were used for quality appraisal. Data were summarized and synthesized using narrative analysis. RESULTS: Twenty-four publications across 11 regions were included, with two principal methods of missed care measurement: modified standard scales and tailored specific approaches. They were applied inconsistently and generated diverse measurement outcomes. There were challenges even with the most commonly used tool, the BERNCA-NH, including absence of high-quality verification through comparative analysis against an established 'gold standard', reliance on self-administration, incomplete assessment of constructs and inadequate exploration of psychometric properties. CONCLUSION: Globally, there are deficiencies in the effectiveness and comprehensiveness of the instruments measuring missed care in LTAC settings. Further research on theoretical and practical perspectives is required. IMPLICATIONS: Findings highlighted a critical need to establish a standardized, validated approach to measure missed care in LTAC settings. This review calls for collaborative efforts by researchers, clinical staff and policymakers to develop and implement evidence-based practices as a way of safeguarding the well-being of older clients living in LTAC settings. IMPACT: Measurements of missed care in LTAC settings rely on adapting acute care tools. There is a critical gap in measuring missed care in LTAC settings. Developing a new tool could improve care quality and safety in LTAC settings globally. REPORTING METHOD: Adhered to PRISMA guideline. PATIENT OR PUBLIC CONTRIBUTION: No patient or public contribution.

2.
EMBO Rep ; 22(11): e52348, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34569703

RESUMO

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.


Assuntos
Tecido Adiposo Bege , Tecido Adiposo Branco , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Adenosina/análogos & derivados , Tecido Adiposo Bege/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Termogênese
3.
EMBO Rep ; 22(5): e52146, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33880847

RESUMO

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.


Assuntos
Curcumina , Fator 4 Associado a Receptor de TNF , Células 3T3-L1 , Adipogenia , Animais , Curcumina/farmacologia , Dieta Hiperlipídica , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/genética , Fator 4 Associado a Receptor de TNF/genética , Fator 4 Associado a Receptor de TNF/metabolismo , Ubiquitinação
4.
Cell Mol Life Sci ; 79(9): 481, 2022 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-35962235

RESUMO

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.


Assuntos
Adipogenia , Proteínas de Ligação a RNA , Adipogenia/genética , Animais , Desenvolvimento Muscular/genética , Transporte de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Suínos
5.
BMC Biol ; 20(1): 39, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35135551

RESUMO

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.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato , Ácidos Docosa-Hexaenoicos , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Animais , Dieta , Ácidos Docosa-Hexaenoicos/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Obesidade , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/farmacologia , RNA Mensageiro/metabolismo , Fatores de Transcrição/metabolismo
6.
J Cell Physiol ; 236(10): 7242-7255, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33843059

RESUMO

Family with sequence similarity 134 member B (FAM134B)/RETREG1/JK1 is a novel gene with recently reported roles in various diseases. Understanding the function and mechanism of action of FAM134B is necessary to develop disease therapies. Notably, emerging data are clarifying the molecular mechanisms of FAM134B function in organelle membrane morphogenesis and the regulation of signaling pathways, such as the Wnt and AKT signaling pathways. In addition, transcription factors, RNA N6 -methyladenosine-mediated epigenetic regulation, microRNA, and small molecules are involved in the regulation of FAM134B expression. This review comprehensively considers recent studies on the role of FAM134B and its potential mechanisms in neurodegenerative diseases, obesity, viral diseases, cancer, and other diseases. The functions of FAM134B in maintaining cell homeostasis by regulating Golgi morphology, endoplasmic reticulum autophagy, and mitophagy are also highlighted, which may be the underlying mechanism of FAM134B gene mutation-induced diseases. Moreover, the molecular mechanisms of the FAM134B function during numerous biological processes are discussed. This review provides novel insights into the functions and mechanisms of FAM134B in various diseases, which will inform the development of effective drugs to treat diseases.


Assuntos
Membranas Intracelulares/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias/metabolismo , Doenças Neurodegenerativas/metabolismo , Obesidade/metabolismo , Organelas/metabolismo , Viroses/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Homeostase , Humanos , Membranas Intracelulares/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Morfogênese , Mutação , Neoplasias/genética , Neoplasias/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Obesidade/genética , Obesidade/patologia , Organelas/genética , Organelas/patologia , Transdução de Sinais , Viroses/genética , Viroses/patologia
7.
FASEB J ; 34(4): 4852-4869, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32048347

RESUMO

Cellular long-chain fatty acids' (LCFAs) uptake is a crucial physiological process that regulates cellular energy homeostasis. AMPK has been shown to modulate LCFAs uptake in several kinds of cells, but whether it exerts an impact on intestinal LCFAs uptake is not quite clear. In the current study, we found that AMPK reinforced LCFAs uptake in intestinal epithelial cells (IECs). Moreover, intestinal epithelium-specific AMPK deletion impaired intestinal LCFAs absorption and protected mice from high-fat diet-induced obesity. Mechanistically, we discovered that AMPK deletion reduced the CD36 protein level by upregulating Parkin-mediated polyubiquitination of CD36 in IECs. Furthermore, our results revealed that AMPK affected PARK2 (gene name of Parkin) mRNA stability in a YTHDF2-dependent manner through FTO-dependent demethylation of N6 -methyladenosine (m6 A). Besides, AMPK promoted the translocation of CD36 to the plasma membrane in IECs, but the inhibition of AKT signaling suppressed this effect, which also halted the accelerated fatty acid uptake induced by AMPK. These results suggest that AMPK facilitates the intestinal LCFAs uptake by upregulating CD36 protein abundance and promoting its membrane translocation simultaneously. Such findings shed light on the role of AMPK in the regulation of intestinal LCFAs uptake.


Assuntos
Antígenos CD36/metabolismo , Ácidos Graxos/metabolismo , Absorção Intestinal , Mucosa Intestinal/metabolismo , Proteínas Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Antígenos CD36/genética , Células CACO-2 , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Estabilidade de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
8.
RNA Biol ; 18(sup2): 711-721, 2021 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-34570675

RESUMO

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.


Assuntos
5-Metilcitosina , Adipogenia/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Biossíntese de Proteínas , RNA Mensageiro/genética , Células 3T3-L1 , 5-Metilcitosina/metabolismo , Animais , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação da Expressão Gênica , Metilação , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Biossíntese de Proteínas/genética , Transporte de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo
9.
J Gerontol Nurs ; 47(4): 45-52, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34038250

RESUMO

The current study explored compliance with hand hygiene and related influencing factors among nursing assistants (NAs) in nursing homes. A descriptive observational research design was used. Seven nursing homes in Chongqing, China, including hospital-affiliated, public, and private, were selected. A hand hygiene observation tool was used to assess NAs' (N = 237) hand hygiene practice (N = 2,370 opportunities). NAs' overall compliance rate was 3.6%: 6.8%, 3.1%, and 1.9% at hospital-affiliated, public, and private nursing homes, respectively. Compliance rate between two opportunities, after contact with residents and after contact with residents' surroundings, differed significantly (p = 0.002 and 0.038, respectively). The highest and lowest compliance rates occurred after bodily fluid exposure (8.3%; odds ratio [OR] = 0.37, 95% confidence interval [CI] [0.218, 0.627], p < 0.001) and before resident contact (1.2%; OR = 3.142, 95% CI [1.265, 7.805], p = 0.014), respectively. Working experience and educational background were the two major influencing factors for hand hygiene. It is urgent to improve NAs' hand hygiene accordingly. [Journal of Gerontological Nursing, 47(4), 45-52.].


Assuntos
Enfermagem Geriátrica , Higiene das Mãos , Assistentes de Enfermagem , Idoso , China , Fidelidade a Diretrizes , Humanos , Casas de Saúde
10.
FASEB J ; 33(6): 7529-7544, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30865855

RESUMO

Bone marrow stem cells (BMSCs) are multipotent stem cells that can regenerate mesenchymal tissues, such as adipose tissue, bone, and muscle. Recent studies have shown that N6-methyladenosine (m6A) methylation, one of the most prevalent epigenetic modifications, is involved in the development process. However, whether it plays roles in BMSC differentiation is still elusive. Here, we found that the deletion of m6A "writer" protein methyltransferase-like (METTL)3 in porcine BMSCs (pBMSCs) could promote adipogenesis and janus kinase (JAK)1 protein expression via an m6A-dependent way. Knockdown of METTL3 decreased mRNA m6A levels of JAK1, leading to enhanced YTH m6A RNA binding protein 2 (YTHDF2)-dependent JAK1 mRNA stability. We further demonstrated that JAK1 activated signal transducer and activator of transcription (STAT) 5 through regulation of its phosphorylation to bind to the promoter of CCAAT/enhancer binding protein (C/EBP) ß, which could ultimately lead to a modulated adipogenic process. Collectively, our results reveal an orchestrated network linking the m6A methylation and JAK1/STAT5/C/EBPß pathway in pBMSCs adipogenic differentiation. Our findings provide novel insights into the underlying molecular mechanisms of m6A modification in the regulation of BMSCs differentiating into adipocytes, which may pave a way to develop more effective therapeutic strategies in stem cell regenerative medicine and the treatment of obesity.-Yao, Y., Bi, Z., Wu, R., Zhao, Y., Liu, Y., Liu, Q., Wang, Y., Wang, X. METTL3 inhibits BMSC adipogenic differentiation by targeting the JAK1/STAT5/C/EBPß pathway via an m6A-YTHDF2-dependent manner.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Diferenciação Celular/fisiologia , Janus Quinase 1/metabolismo , Células-Tronco Mesenquimais/química , Metiltransferases/fisiologia , Fator de Transcrição STAT5/metabolismo , Adipogenia/fisiologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Humanos , Metiltransferases/genética , Suínos , Transcrição Gênica
11.
FASEB J ; 33(2): 2971-2981, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30339471

RESUMO

Intramuscular fat is considered a potential factor that is associated with meat quality in animal production and insulin resistance in humans. N6-methyladenosine (m6A) modification of mRNA plays an important role in regulating adipogenesis. However, the effects of m6A on the adipogenesis of intramuscular preadipocytes and associated mechanisms remain unknown. Here, we performed m6A sequencing to compare m6A methylome of the longissimus dorsi muscles (LDMs) between Landrace pigs (lean-type breed) and Jinhua pigs (obese-type breed with higher levels of intramuscular fat). Transcriptome-wide m6A profiling of porcine LDMs was highly conserved with humans and mice. Furthermore, we identified a unique methylated gene in Jinhua pigs named mitochondrial carrier homology 2 ( MTCH2). The m6A levels of MTCH2 mRNA were reduced by introducing a synonymous mutation, and adipogenesis test results showed that the MTCH2 mutant was inferior with regard to adipogenesis compared with the MTCH2 wild-type. We then found that MTCH2 protein expression was positively associated with m6A levels, and an YTH domain family protein 1-RNA immunoprecipitation-quantitative PCR assay indicated that MTCH2 mRNA was a target of the YTH domain family protein 1. This study provides comprehensive m6A profiles of LDM transcriptomes in pigs and suggests an essential role for m6A modification of MTCH2 in intramuscular fat regulation.-Jiang, Q., Sun, B., Liu, Q., Cai, M., Wu, R., Wang, F., Yao, Y., Wang, Y., Wang, X. MTCH2 promotes adipogenesis in intramuscular preadipocytes via an m6A-YTHDF1-dependent mechanism.


Assuntos
Adenosina/análogos & derivados , Adipócitos/citologia , Adipogenia , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Músculo Esquelético/citologia , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Adenosina/química , Adipócitos/metabolismo , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Metilação , Proteínas de Transporte da Membrana Mitocondrial/genética , Desenvolvimento Muscular , Músculo Esquelético/metabolismo , Estabilidade de RNA , RNA Mensageiro/química , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Suínos
12.
Int J Mol Sci ; 21(7)2020 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-32230940

RESUMO

The present study aimed to elucidate how Atlantic salmon adipocytes pre-enriched with palmitic (16:0, PA), oleic (18:1n-9, OA), or eicosapentaenoic (20:5n-3, EPA) acid respond to a fasting condition mimicked by nutrient deprivation and glucagon. All experimental groups were supplemented with radiolabeled PA to trace secreted lipids and distribution of radioactivity in different lipid classes. There was a higher content of intracellular lipid droplets in adipocytes pre-enriched with OA than in adipocytes pre-enriched with PA or EPA. In the EPA group, the radiolabeled PA was mainly esterified in phospholipids and triacylglycerols, whereas in the OA and PA groups, the radioactivity was mainly recovered in phospholipids and cholesterol-ester. By subjecting the experimental groups to nutrient-deprived media supplemented with glucagon, lipolysis occurred in all groups, although to a lower extent in the OA group. The lipids were mainly secreted as esterified lipids in triacylglycerols and phospholipids, indicating mobilization in lipoproteins. A significant proportion was secreted as free fatty acids and glycerol. Leptin secretion was reduced in all experimental groups in response to fasting, while the mitochondria area responded to changes in the energy supply and demand by increasing after 3 h of fasting. Overall, different lipid classes in adipocytes influenced their mobilization during fasting.


Assuntos
Adipócitos/metabolismo , Metabolismo dos Lipídeos/fisiologia , Salmo salar/metabolismo , Animais , Jejum , Ácidos Graxos/metabolismo , Óleos de Peixe/metabolismo , Glucagon/metabolismo , Glicerol/metabolismo , Gotículas Lipídicas , Lipídeos , Lipólise , Mitocôndrias/metabolismo , Fosfolipídeos/metabolismo , Salmo salar/genética , Triglicerídeos/metabolismo
13.
Int J Mol Sci ; 21(11)2020 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-32521827

RESUMO

Adipocytes play a central role in overall energy homeostasis and are important contributors to the immune system. Fatty acids (FAs) act as signaling molecules capable to modulate adipocyte metabolism and functions. To identify the effects of two commonly used FAs in Atlantic salmon diets, primary adipocytes were cultured in the presence of oleic (OA) or docosahexaenoic (DHA) acid. DHA decreased adipocyte lipid droplet number and area compared to OA. The increase in lipid load in OA treated adipocytes was paralleled by an increase in iNOS activity and mitochondrial SOD2-GFP activity, which was probably directed to counteract increase in oxidative stress. Under lipopolysaccharide (LPS)-induced inflammation, DHA had a greater anti-inflammatory effect than OA, as evidenced by the higher SOD2 activity and the transcriptional regulation of antioxidant enzymes and pro- and anti-inflammatory markers. In addition, DHA maintained a healthy mitochondrial structure under induced inflammation while OA led to elongated mitochondria with a thin thread like structures in adipocytes exposed to LPS. Overall, DHA possess anti-inflammatory properties and protects Atlantic salmon against oxidative stress and limits lipid deposition. Furthermore, DHA plays a key role in protecting mitochondria shape and function.


Assuntos
Adipócitos/imunologia , Adipócitos/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Imunidade/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Salmo salar/metabolismo , Adipócitos/efeitos dos fármacos , Animais , Antioxidantes/metabolismo , Biomarcadores , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipopolissacarídeos/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos
14.
J Cell Physiol ; 234(6): 7948-7956, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30644095

RESUMO

N6 -methyladenosine (m6 A), as the most abundant RNA epigenetic modifications, has been shown to play critical roles in various biological functions. Research about enzymes that can catalyze and remove m6 A have revealed its comprehensive roles in messenger RNA (mRNA) metabolism and other physiological processes. The "readers" including YTH domain-containing proteins, hnRNPC, hnRNPG, hnRNPA2B1, IGF2BP1, IGF2BP2, and IGF2BP3, which can affect the fates of mRNA in an m6 A-dependent manner. In this review, we focus on recent advances in the research of the m6 A modifications, especially about the latest functions of its writers, erasers, readers in RNA metabolism, cancer, and lipid metabolism. In the end, we provide insights into the underlying molecular mechanisms of m6 A modifications.


Assuntos
Adenosina/genética , Epigênese Genética , Neoplasias/genética , RNA Mensageiro/genética , Adenosina/análogos & derivados , Humanos , Metabolismo dos Lipídeos/genética , Metilação , Metiltransferases/genética , Processamento Pós-Transcricional do RNA/genética
15.
Biol Chem ; 400(4): 523-532, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30291780

RESUMO

Family with sequence similarity 134, Member B (FAM134B), is a cis-Golgi transmembrane protein that is known to be necessary for the long-term survival of nociceptive and autonomic ganglion neurons. Recent work has shown that FAM134B plays a pivotal role in autophagy-mediated turnover of endoplasmic reticulum (ER) membranes, tumor inhibition and lipid homeostasis. In this study, we provide mechanistic links between FAM134B and ARF-related protein 1 (ARFRP1) and further show that FAM134B resides in the Golgi apparatus. Here, we found that FAM134B increased lipid accumulation in adipocytes. Transport vehicle number and ADP-ribosylation factor (ARF) family gene expression were also increased by FAM134B overexpression, suggesting that vesicular transport activity enhanced lipid accumulation. ARF-related protein 1 (ARFRP1) is a GTPase that promotes protein trafficking. We show that FAM134B regulates the expression of ARFRP1, and the knockdown of ARFRP1 abolishes enhancement on lipid accumulation caused by FAM134B. In addition, FAM134B upregulates the PAT family protein (PAT), which associates with the lipid droplets (LDs) surface and promotes lipolysis by recruiting adipocyte triglyceride lipase (ATGL). These findings indicate that FAM134B promotes lipid accumulation and adipogenic differentiation by increasing vesicle transport activity in the Golgi apparatus and inhibiting the lipolysis of LDs.


Assuntos
Adipócitos/metabolismo , Adipogenia , Proteínas de Membrana/metabolismo , Células 3T3-L1 , Animais , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Membrana/genética , Camundongos , Suínos
16.
IUBMB Life ; 71(5): 580-586, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30506811

RESUMO

N6 -methyladenosine (m6 A) mRNA modification plays an important role in adipogenesis, but its role on single gene remains unexplored. Family with Sequence Similarity 134, Member B (FAM134B) is a cis-Golgi transmembrane protein that known to be necessary for the long-term survival of nociceptive and autonomic ganglion neurons. Recent work has shown that FAM134B plays a pivotal role in lipid homeostasis and was identified as its significant m6 A level difference between Chinese local Jinhua pigs and Landrace through RNA-sequence. Here, we construct the non-m6 A FAM134B coding sequence (CDS) plasmid (FAM134B-MUT) and found one important m6 A site on its CDS. Expression of FAM134B-MUT was more effective in promoting porcine preadipocytes adipogenic differentiation and lipid deposition than wild-type FAM134B (FAM134B-WT) both in early and ultimate differentiation stage. FAM134B-MUT functions better in promoting fat deposition by upregulating peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein (C/EBPα) level. The m6 A reader protein YTH m6 A RNA binding protein 2 (YTHDF2) interacts with FAM134B mRNA and down regulated its protein level. These results demonstrate that FAM134B was the target of YTHDF2, which may recognize and binds the m6 A site of FAM134B to reduce its mRNA lifetime and reduce its protein abundance. © 2018 IUBMB Life, 71(5):580-586, 2019.


Assuntos
Adenosina/análogos & derivados , Adipócitos/citologia , Adipogenia , Diferenciação Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Ligação a RNA/metabolismo , Adenosina/deficiência , Adipócitos/metabolismo , Animais , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Proteínas de Ligação a RNA/genética , Suínos
17.
Protein Expr Purif ; 161: 17-27, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30926463

RESUMO

A new alginate lyase gene of algl17 was cloned from an alginate-degrading marine bacterium Microbulbifer sp. ALW1. The gene contained 2220 bp and encoded a 739-amino acid protein classified into the PL-17 family. The recombinant alginate lyase AlgL17 was overexpressed and purified from Escherichia coli BL21 (DE3) with a molecular mass of 84.9 kDa. This enzyme showed activities towards sodium alginate, polyM and alginate oligosaccharide, but very low activity towards polyG. These results indicate that AlgL17 is a polyM-specific oligoalginate lyase. When sodium alginate was used as a substrate, the optimum reaction temperature and pH for the enzyme were 35 °C and pH 8.0, respectively. Recombinant AlgL17 was stable at 25 °C, but not stable at 30 °C and 35 °C. It showed good stability over a pH range of 5.0-8.0. The enzyme activity was increased to 1.7 times by adding NaCl to a final concentration of 0.7 M. The ability of the recombinant AlgL17 producing 4-deoxy-L-erythro-5-hexoseulose uronic acid (DEH) from sodium alginate and polyM block indicates that AlgL17 is an exo-type alginate lyase.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Clonagem Molecular , Gammaproteobacteria/enzimologia , Polissacarídeo-Liases/química , Polissacarídeo-Liases/genética , Alginatos/metabolismo , Proteínas de Bactérias/metabolismo , Estabilidade Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Gammaproteobacteria/genética , Concentração de Íons de Hidrogênio , Cinética , Peso Molecular , Polissacarídeo-Liases/metabolismo , Especificidade por Substrato , Temperatura
18.
RNA Biol ; 16(12): 1785-1793, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31434544

RESUMO

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.


Assuntos
Adenosina/análogos & derivados , Adipócitos/metabolismo , Adipogenia/genética , Metiltransferases/genética , Transativadores/genética , Células 3T3-L1 , Adenosina/metabolismo , Adipócitos/citologia , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diferenciação Celular , Células Clonais , Ciclina D1/genética , Ciclina D1/metabolismo , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Regulação da Expressão Gênica , Metiltransferases/metabolismo , Camundongos , Mitose , PPAR gama/genética , PPAR gama/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Transativadores/antagonistas & inibidores , Transativadores/metabolismo , Transfecção
19.
Int J Obes (Lond) ; 42(7): 1378-1388, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29795461

RESUMO

BACKGROUND/OBJECTIVE: N6-methyladenosine (m6A) modification of mRNA plays a role in regulating adipogenesis. However, its underlying mechanism remains largely unknown. Epigallocatechin gallate (EGCG), the most abundant catechin in green tea, plays a critical role in anti-obesity and anti-adipogenesis. METHODS: High-performance liquid chromatography coupled with triple-quadrupole tandem mass spectrometry (HPLC-QqQ-MS/MS) was performed to determine the m6A levels in 3T3-L1 preadipocytes. The effects of EGCG on the m6A levels in specific genes were determined by methylated RNA immunoprecipitation coupled with quantitative real-time PCR (meRIP-qPCR). Several adipogenesis makers and cell cycle genes were analyzed by quantitative real-time PCR (qPCR) and western blotting. Lipid accumulation was evaluated by oil red O staining. All measurements were performed at least for three times. RESULTS: Here we showed that EGCG inhibited adipogenesis by blocking the mitotic clonal expansion (MCE) at the early stage of adipocyte differentiation. Exposing 3T3-L1 cells to EGCG reduced the expression of fat mass and obesity-associated (FTO) protein, an m6A demethylase, which led to increased overall levels of RNA m6A methylation. Cyclin A2 (CCNA2) and cyclin dependent kinase 2 (CDK2) play vital roles in MCE. The m6A levels of CCNA2 and CDK2 mRNA were dramatically enhanced by EGCG. Interestingly, EGCG increased the expression of YTH N6-methyladenosine RNA binding protein 2 (YTHDF2), which recognized and decayed methylated mRNAs, resulting in decreased protein levels of CCNA2 and CDK2. As a result, MCE was blocked and adipogenesis was inhibited. FTO overexpression and YTHDF2 knockdown in 3T3-L1 cells significantly increased CCNA2 and CDK2 protein levels and ameliorated the EGCG-induced adipogenesis inhibition. Thus, m6A-dependent CCNA2 and CDK2 expressions mediated by FTO and YTHDF2 contributed to EGCG-induced adipogenesis inhibition. CONCLUSION: Our findings provide mechanistic insights into how m6A is involved in the EGCG regulation of adipogenesis and shed light on its anti-obesity effect.


Assuntos
Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Fármacos Antiobesidade/farmacologia , Catequina/análogos & derivados , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Células 3T3-L1/citologia , Adipócitos/citologia , Dioxigenase FTO Dependente de alfa-Cetoglutarato/deficiência , Animais , Catequina/farmacologia , Modelos Animais de Doenças , Camundongos , RNA Mensageiro/química , RNA Mensageiro/genética , Chá/química
20.
Int J Obes (Lond) ; 42(11): 1912-1924, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29487348

RESUMO

BACKGROUND/OBJECTIVE: N6-methyladenosine (m6A) modification of mRNA plays an important role in regulating adipogenesis. However, its underlying mechanism remains largely unknown. SUBJECTS/METHODS: Using Jinhua and Landrace pigs as fat and lean models, we presented a comprehensive transcriptome-wide m6A profiling in adipose tissues from these two pig breeds. Two differentially methylated genes were selected to explore the mechanisms of m6A-mediated regulation of gene function. RESULTS: The ratio of m6A/A in the layer of backfat (LB) was significantly higher in Landrace than that in Jinhua. Transcriptome-wide m6A profiling revealed that m6A modification on mRNA occurs in the conserved sequence motif of RRACH and that the pig transcriptome contains 0.53-0.91 peak per actively expressed transcript. The relative density of m6A peaks in the 3'UTR were higher than in 5'UTR. Genes with common m6A peaks from both Landrace (L-LB) and Jinhua (J-LB) were enriched in RNA splicing and cellular lipid metabolic process. The unique m6A peak genes (UMGs) from L-LB were mainly enriched in the extracellular matrix (ECM) and collagen catabolic process, whereas the UMGs from J-LB are mainly involved in RNA splicing, etc. Lipid metabolism processes were not significantly enriched in the UMGs from L-LB or J-LB. Uncoupling protein-2 (UCP2) and patatin-like phospholipase domain containing 2 (PNPLA2) were two of the UMGs in L-LB. Synonymous mutations (MUT) were conducted to reduce m6A level of UCP2 and PNPLA2 mRNAs. Adipogenesis test showed that UCP2-MUT further inhibited adipogenesis, while PNPLA2-MUT promoted lipid accumulation compared with UCP2-WT and PNPLA2-WT, respectively. Further study showed m6A negatively mediates UCP2 protein expression and positively mediates PNPLA2 protein expression. m6A modification affects the translation of PNPLA2 most likely through YTHDF1, whereas UCP2 is likely neither the target of YTHDF2 nor the target of YTHDF1. CONCLUSION: Our data demonstrated a conserved and yet dynamically regulated m6A methylome in pig transcriptomes and provided an important resource for studying the function of m6A epitranscriptomic modification in obesity development.


Assuntos
Adipócitos/metabolismo , Metabolismo dos Lipídeos/fisiologia , Obesidade/patologia , RNA Mensageiro/metabolismo , Magreza/patologia , Proteína Desacopladora 2/metabolismo , Animais , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Lipase/metabolismo , Análise de Sequência de RNA , Suínos , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA