Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(5)2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38473768

ABSTRACT

Intramuscular fat, also referred to as marbling fat, is the white fat deposited within skeletal muscle tissue. The content of intramuscular fat in the skeletal muscle, particularly the longissimus dorsi muscle, of cattle is a critical determinant of beef quality and value. In this review, we summarize the process of intramuscular fat development and growth, the factors that affect this process, and the molecular and epigenetic mechanisms that mediate this process in cattle. Compared to other species, cattle have a remarkable ability to accumulate intramuscular fat, partly attributed to the abundance of sources of fatty acids for synthesizing triglycerides. Compared to other adipose depots such as subcutaneous fat, intramuscular fat develops later and grows more slowly. The commitment and differentiation of adipose precursor cells into adipocytes as well as the maturation of adipocytes are crucial steps in intramuscular fat development and growth in cattle. Each of these steps is controlled by various factors, underscoring the complexity of the regulatory network governing adipogenesis in the skeletal muscle. These factors include genetics, epigenetics, nutrition (including maternal nutrition), rumen microbiome, vitamins, hormones, weaning age, slaughter age, slaughter weight, and stress. Many of these factors seem to affect intramuscular fat deposition through the transcriptional or epigenetic regulation of genes directly involved in the development and growth of intramuscular fat. A better understanding of the molecular and cellular mechanisms by which intramuscular fat develops and grows in cattle will help us develop more effective strategies to optimize intramuscular fat deposition in cattle, thereby maximizing the quality and value of beef meat.


Subject(s)
Adipocytes , Epigenesis, Genetic , Cattle , Animals , Cell Differentiation , Adipogenesis , Growth and Development
2.
Int J Surg ; 109(11): 3476-3489, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37578452

ABSTRACT

OBJECTIVE: To construct a novel tumor-node-morphology (TNMor) staging system derived from natural language processing (NLP) of pathology reports to predict outcomes of pancreatic ductal adenocarcinoma. METHOD: This retrospective study with 1657 participants was based on a large referral center and The Cancer Genome Atlas Program (TCGA) dataset. In the training cohort, NLP was used to extract and screen prognostic predictors from pathology reports to develop the TNMor system, which was further evaluated with the tumor-node-metastasis (TNM) system in the internal and external validation cohort, respectively. Main outcomes were evaluated by the log-rank test of Kaplan-Meier curves, the concordance index (C-index), and the area under the receiver operating curve (AUC). RESULTS: The precision, recall, and F1 scores of the NLP model were 88.83, 89.89, and 89.21%, respectively. In Kaplan-Meier analysis, survival differences between stages in the TNMor system were more significant than that in the TNM system. In addition, our system provided an improved C-index (internal validation, 0.58 vs. 0.54, P <0.001; external validation, 0.64 vs. 0.63, P <0.001), and higher AUCs for 1, 2, and 3-year survival (internal validation: 0.62 vs. 0.54, P <0.001; 0.64 vs. 0.60, P= 0.017; 0.69 vs. 0.62, P= 0.001; external validation: 0.69 vs. 0.65, P= 0.098; 0.68 vs. 0.64, P= 0.154; 0.64 vs. 0.55, P= 0.032, respectively). Finally, our system was particularly beneficial for precise stratification of patients receiving adjuvant therapy, with an improved C-index (0.61 vs. 0.57, P <0.001), and higher AUCs for 1-year, 2-year, and 3-year survival (0.64 vs. 0.57, P <0.001; 0.64 vs. 0.58, P <0.001; 0.67 vs. 0.61, P <0.001; respectively) compared with the TNM system. CONCLUSION: These findings suggest that the TNMor system performed better than the TNM system in predicting pancreatic ductal adenocarcinoma prognosis. It is a promising system to screen risk-adjusted strategies for precision medicine.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Prognosis , Retrospective Studies , Neoplasm Staging , Natural Language Processing , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/therapy , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms
3.
Front Endocrinol (Lausanne) ; 14: 1199589, 2023.
Article in English | MEDLINE | ID: mdl-37305046

ABSTRACT

The inhibitory effect of growth hormone (GH) on adipose tissue growth is well known, but the underlying mechanism is not fully understood. In this study, we determined the possibility that GH inhibits adipose tissue growth by inhibiting adipogenesis, the process of formation of adipocytes from stem cells, in the lit/lit mice. The lit/lit mice are GH deficient because of a spontaneous mutation to the GH releasing hormone receptor (ghrhr) gene, and they have more subcutaneous fat despite being smaller than the lit/+ mice at the same age. We found that cells of the stromal vascular fraction (SVF) of subcutaneous fat from the lit/lit mice had greater adipogenic potential than those from the lit/+ mice, as evidenced by forming greater numbers of lipid droplets-containing adipocytes and having greater expression of adipocyte marker genes during induced adipocyte differentiation in culture. However, addition of GH to the culture did not reverse the superior adipogenic potential of subcutaneous SVF from the lit/lit mice. Through florescence-activated cell sorting and quantification of mRNAs of preadipocyte markers, including CD34, CD29, Sca-1, CD24, Pref-1, and PPARγ, we found that subcutaneous SVF from the lit/lit mice contained more preadipocytes than that from the lit/+ mice. These results support the notion that GH inhibits adipose tissue growth in mice at least in part by inhibiting adipogenesis. Furthermore, these results suggest that GH inhibits adipogenesis in mice not by inhibiting the terminal differentiation of preadipocytes into adipocytes, rather by inhibiting the formation of preadipocytes from stem cells or the recruitment of stem cells to the fat depot.


Subject(s)
Human Growth Hormone , Subcutaneous Fat , Animals , Mice , Adipose Tissue , Growth Hormone , Adipocytes
4.
Front Physiol ; 13: 863860, 2022.
Article in English | MEDLINE | ID: mdl-35547590

ABSTRACT

Flavonoids, naturally-occurring compounds with multiple phenolic structures, are the most widely distributed phytochemicals in the plant kingdom, and are mainly found in vegetables, fruits, grains, roots, herbs, and tea and red wine products. Flavonoids have health-promoting effects and are indispensable compounds in nutritional and pharmaceutical (i.e., nutraceutical) applications. Among the demonstrated bioactive effects of flavonoids are anti-oxidant, anti-inflammatory, and anti-microbial in a range of research models. Through dietary formulation strategies, numerous flavonoids provide the ability to support bird health while improving the nutritional quality of poultry meat and eggs by changing the profile of fatty acids and reducing cholesterol content. A number of such compounds have been shown to inhibit adipogenesis, and promote lipolysis and apoptosis in adipose tissue cells, and thereby have the potential to affect fat accretion in poultry at various ages and stages of production. Antioxidant and anti-inflammatory properties contribute to animal health by preventing free radical damage in tissues and ameliorating inflammation in adipose tissue, which are concerns in broiler breeders and laying hens. In this review, we summarize the progress in understanding the effects of dietary flavonoids on lipid metabolism and fat deposition in poultry, and discuss the associated physiological mechanisms.

5.
Biomolecules ; 9(7)2019 07 11.
Article in English | MEDLINE | ID: mdl-31336727

ABSTRACT

tRNA-derived fragments (tRFs), a novel type of non-coding RNA derived from tRNAs, play an important part in governing gene expressions at a post-transcriptional level. To date, the regulatory mechanism of tRFs governing fat deposition and adipogenesis is completely unknown. In this study, high fat diet was employed to induce an obese rat model, and tRFs transcriptome sequencing was conducted to identify differentially expressed tRFs that response to obesity. We found out that tRFGluTTC, which promoted preadipocyte proliferation by increasing expressions of cell cycle regulatory factors, had the highest fold change in the 296 differentially expressed tRFs. Moreover, tRFGluTTC also suppressed preadipocyte differentiation by reducing triglyceride content and lipid accumulation, and by decreasing expressions of genes that related to fatty acid synthesis. According to results of luciferase activity analysis, tRFGluTTC directly targeted Kruppel-like factor (KLF) 9, KLF11, and KLF12, thus significantly suppressing mRNA expressions of these target genes. Moreover, tRFGluTTC suppressed adipogenesis, accompanying by suppressing expressions of adipogenic transcription factors (aP2, PPARγ, and C/EBPα). In conclusion, these results imply that tRFGluTTC may act as a novel epigenetic molecule regulating adipogenesis and could provide a new strategy for the intervention treatment of obesity.


Subject(s)
Adipogenesis/physiology , Diet, High-Fat/adverse effects , Obesity/metabolism , 3T3-L1 Cells , Adipogenesis/genetics , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Epigenesis, Genetic/genetics , Epigenesis, Genetic/physiology , Male , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcriptome/genetics
6.
J Mol Med (Berl) ; 96(7): 685-700, 2018 07.
Article in English | MEDLINE | ID: mdl-29876588

ABSTRACT

Increasing evidence indicates that muscular dysfunction or alterations in skeletal muscle fiber-type composition not only are involved in muscle metabolism and function but also can limit functional capacity. Therefore, understanding the mechanisms regulating key events during skeletal myogenesis is necessary. Betaine is a naturally occurring component of commonly eaten foods. Here, we showed that 10 mM betaine supplementation in vitro significantly repressed myoblast proliferation and enhanced myoblast differentiation. This effect can be mediated by regulation of miR-29b-3p. Further analysis showed that betaine supplementation in vitro regulated skeletal muscle fiber-type composition through the induction of NFATc1 and the negative regulation of MyoD expression. Furthermore, mice fed with 10 mM betaine in water for 133 days showed no impairment in overall health. Consistently, betaine supplementation increased muscle mass, promoted muscle formation, and modulated the ratio of fiber types in skeletal muscle in vivo. These findings shed light on the diverse biological functions of betaine and indicate that betaine supplementation may lead to new therapies for diseases such as muscular dystrophy or other diseases related to muscle dysfunction. KEY MESSAGES: Betaine supplementation inhibits proliferation and promotes differentiation of C2C12 myoblasts. Betaine supplementation regulates fast to slow muscle fiber-type conversion and is associated with NFATc1/MyoD. Betaine supplementation enhances skeletal myogenesis in vivo. Betaine supplementation does not impair health of mice.


Subject(s)
Betaine/pharmacology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , MyoD Protein/metabolism , NFATC Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , DNA Methylation , Dietary Supplements , Female , Immunohistochemistry , Mice , Models, Biological , Muscle Development/drug effects , Muscle Development/genetics , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism
7.
Biomed Pharmacother ; 101: 528-535, 2018 May.
Article in English | MEDLINE | ID: mdl-29505923

ABSTRACT

Myogenic differentiation, which occurs in the process of muscle development, is a highly ordered process. Increasing evidence indicates that microRNAs (miRNAs) are important regulators in myogenic processes. In this study, we found that miR-204-5p expression gradually decreased when myoblasts were induced to differentiate. Our results suggested that miR-204-5p blunted myoblast differentiation, which was accompanied with a decreased proportion of myosin heavy chain (MyHC)-positive cells in myoblasts with augmented expression of miR-204-5p. Furthermore, overexpression of miR-204-5p significantly decreased the MyHC composition of slow-twitch fibers in myoblasts. Luciferase activity assays confirmed that miR-204-5p directly targeted the 3'-untranslated region (3'-UTR) of myocyte enhancer factor 2C (MEF2C) and estrogen-related receptor gamma (ERRγ). Small interfering RNA (siRNA) technology successfully inhibited the expression of MEF2C and ERRγ. Interference with MEF2C or ERRγ inhibited myoblast differentiation and the formation of slow-twitch fibers. Meanwhile, co-transfection of either si-MEF2C or si-ERRγ with miR-204-5p mimics resulted in a more severe attenuation of myogenic differentiation. In summary, this study demonstrates that miR-204-5p inhibits myoblast differentiation by targeting MEF2C and ERRγ. Our findings suggest that miR-204-5p is a potential regulator that could influence myogenesis.


Subject(s)
Cell Differentiation/genetics , MicroRNAs/genetics , Myoblasts/physiology , Receptors, Estrogen/genetics , 3' Untranslated Regions/genetics , Animals , Cell Line , Cell Line, Tumor , HeLa Cells , Humans , MEF2 Transcription Factors/genetics , Mice , Muscle Development/genetics , RNA, Small Interfering/genetics
8.
Nutrients ; 10(2)2018 Jan 26.
Article in English | MEDLINE | ID: mdl-29373534

ABSTRACT

Obesity is a major driver of metabolic diseases such as nonalcoholic fatty liver disease, certain cancers, and insulin resistance. However, there are no effective drugs to treat obesity. Betaine is a nontoxic, chemically stable and naturally occurring molecule. This study shows that dietary betaine supplementation significantly inhibits the white fat production in a high-fat diet (HFD)-induced obese mice. This might be due to betaine preventing the formation of new white fat (WAT), and guiding the original WAT to burn through stimulated mitochondrial biogenesis and promoting browning of WAT. Furthermore, dietary betaine supplementation decreases intramyocellular lipid accumulation in HFD-induced obese mice. Further analysis shows that betaine supplementation reduced intramyocellular lipid accumulation might be associated with increasing polyunsaturated fatty acids (PUFA), fatty acid oxidation, and the inhibition of fatty acid synthesis in muscle. Notably, by performing insulin-tolerance tests (ITTs) and glucose-tolerance tests (GTTs), dietary betaine supplementation could be observed for improvement of obesity and non-obesity induced insulin resistance. Together, these findings could suggest that inhibiting WAT production, intramyocellular lipid accumulation and inflammation, betaine supplementation limits HFD-induced obesity and improves insulin resistance.


Subject(s)
Adiposity , Anti-Obesity Agents/therapeutic use , Betaine/therapeutic use , Dietary Supplements , Insulin Resistance , Lipid Metabolism , Obesity/diet therapy , 3T3-L1 Cells , Adipocytes, White/cytology , Adipocytes, White/metabolism , Adipocytes, White/pathology , Adipogenesis , Animals , Animals, Outbred Strains , Betaine/adverse effects , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/diet therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat/adverse effects , Female , Hyperglycemia/prevention & control , Hypoglycemic Agents/therapeutic use , Lipid Droplets/metabolism , Lipid Droplets/pathology , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Weight Gain
9.
Yi Chuan ; 39(9): 784-797, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28936977

ABSTRACT

Enhancers are key cis-acting gene regulatory elements in eukaryotes, which can effectively promote the expression of target genes. Emerging evidence showed that enhancers in activation state could be transcribed to enhancer RNAs (eRNAs), the processes of which are regulated by various signaling systems and actions of signal-dependent transcription factors. Compared with the other transcripts (e.g. lncRNA, mRNA), eRNAs have shorter sequences, lower stability, and higher tissue specificity. eRNAs play roles in the initiation or stabilization of enhancer-promoter looping, and promote the expression of target genes. Recent studies have further showed that eRNAs have crucial roles in biological processes, such as development and disease initiation and progression. However, functional studies of eRNAs are currently lacking, and the regulatory mechanisms of eRNAs are still uncertain. Herein, we focus on the features, research methods and functional properties of eRNAs, and discusse the possibility of using eRNAs as therapeutic targets. We hope this discussion might provide some insights for further research on eRNAs.


Subject(s)
Enhancer Elements, Genetic/genetics , RNA/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Gene Expression Regulation/genetics , Humans , Promoter Regions, Genetic/genetics
10.
Biochem Biophys Res Commun ; 492(1): 82-88, 2017 10 07.
Article in English | MEDLINE | ID: mdl-28803985

ABSTRACT

Body fat mass is closely associated to diseases related to obesity. MicroRNAs (miRNAs, miR) are important regulatory molecules that function as post-transcriptional gene regulators of adipocyte development. In the current study, we revealed that reduced expression of miR-199a-3p in adipose tissue resulting from high fat diet (HFD)-induced obesity in mice. Overexpression of miR-199a-3p promoted adipocyte proliferation by regulating the expression of regulating factors of the cell cycle. Furthermore, miR-199a-3p blunted lipid accumulation in 3T3-L1 adipocytes. This was accompanied by a marked decrease in the expression of adipocyte-specific genes involved in lipogenic transcription, fatty acid synthesis, and fatty acid transportation. Furthermore, the fatty acid oxidation process was enhanced. Luciferase activity assays confirmed that miR-199a-3p regulates adipocyte differentiation by directly targeting the 3'-untranslated region (3'-UTR) of stearoyl-CoA desaturase (SCD). Moreover, miR-199a-3p regulates fatty acid composition by decreasing the ratio of unsaturated fatty acids (UFAs) in adipocytes transfected with miR-199a-3p mimics. These results suggest that miR-199a-3p may promote adipocyte proliferation, while also repressing adipocyte differentiation by down-regulating SCD and changing fatty acid composition during adipogenesis.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Cell Differentiation/genetics , Fatty Acids/metabolism , MicroRNAs/genetics , Stearoyl-CoA Desaturase/deficiency , Stearoyl-CoA Desaturase/genetics , 3T3-L1 Cells , Animals , Diet, High-Fat , Male , Mice , Mice, Inbred Strains , Mice, Obese
11.
Asian-Australas J Anim Sci ; 30(7): 938-943, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28002929

ABSTRACT

OBJECTIVE: Qingyu pig, a Chinese indigenous pig breed, exhibits two types of coat colour phenotypes, including pure black and white with black spotting respectively. Melanocortin receptor 1 (MC1R) and agouti signaling protein (ASIP) are two widely reported pivotal genes that significantly affect the regulation of coat colour. The objectives of this study were to investigate whether the polymorphisms of these two genes are associated with coat colour and analyze the molecular mechanism of the coat colour separation in Qingyu pig. METHODS: We studied the phenotype segregation and used polymerase chain reaction amplification and Sanger sequencing to investigate the polymorphism of MC1R and ASIP in 121 Qingyu pigs, consisting of 115 black and 6 white with black spotted pigs. RESULTS: Coat colour of Qingyu pig is associated with the polymorphisms of MC1R but not ASIP. We only found 2 haplotypes, EQY and Eqy , based on the 13 observed mutations from MC1R gene. Among which, Eqy presented a recessive inheritance mode in black spotted Qingyu pigs. Further analysis revealed a g.462-463CC insertion that caused a frameshift mutation and a premature stop codon, thus changed the first transmembrane domain completely and lost the remaining six transmembrane domains. Altogether, our results strongly support that the variety of Qingyu pig's coat colour is related to MC1R. CONCLUSION: Our findings indicated that black coat colour in Qingyu pig was dominant to white with black spotted phenotype and MC1R gene polymorphism was associated with coat colour separation in Qingyu pig.

12.
Sci Rep ; 6: 32186, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27561200

ABSTRACT

The physiological, biochemical and functional differences between oxidative and glycolytic muscles play important roles in human metabolic health and in animal meat quality. To explore these differences, we determined the genome-wide landscape of DNA methylomes and their relationship with the mRNA and miRNA transcriptomes of the oxidative muscle psoas major (PMM) and the glycolytic muscle longissimus dorsi (LDM). We observed the hypo-methylation of sub-telomeric regions. A high mitochondrial content contributed to fast replicative senescence in PMM. The differentially methylated regions (DMRs) in promoters (478) and gene bodies (5,718) were mainly enriched in GTPase regulator activity and signaling cascade-mediated pathways. Integration analysis revealed that the methylation status within gene promoters (or gene bodies) and miRNA promoters was negatively correlated with mRNA and miRNA expression, respectively. Numerous genes were closely related to distinct phenotypic traits between LDM and PMM. For example, the hyper-methylation and down-regulation of HK-2 and PFKFB4 were related to decrease glycolytic potential in PMM. In addition, promoter hypo-methylation and the up-regulation of miR-378 silenced the expression of the target genes and promoted capillary biosynthesis in PMM. Together, these results improve understanding of muscle metabolism and development from genomic and epigenetic perspectives.


Subject(s)
DNA Methylation/physiology , Genome-Wide Association Study , MicroRNAs , Muscle, Skeletal/metabolism , RNA, Messenger , Transcriptome/physiology , Animals , Glycolysis/physiology , MicroRNAs/biosynthesis , MicroRNAs/genetics , Oxidation-Reduction , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Swine
13.
Biochem Biophys Res Commun ; 475(1): 140-8, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27179777

ABSTRACT

MicroRNAs (miRNAs, miR) play important roles in adipocyte development. Recent studies showed that the expression of several miRNAs is closely related with promoter methylation. However, it is not known whether miRNA mediates adipocytes differentiation by means of DNA methylation. Here, we showed that miR-145a-5p was poorly expressed in adipose tissue from mice fed a high fat diet (HFD). Overexpression or inhibition of miR-145a-5p was unfavorable or beneficial, respectively, for adipogenesis, and these effects were achieved by regulating adipocyte-specific genes involved in lipogenic transcription, fatty acid synthesis, and fatty acid transportation. Particularly, we first suggested that miR-145a-5p mimics or inhibitors promoted or repressed adipocytes proliferation by regulating p53 and p21, which act as cell cycle regulating factors. Surprisingly, the miR-145a-5p-repressed adipocyte differentiation was enhanced or rescued when cells treated with 5-Aza-dC were transfected with miR-145a-5p mimics or inhibitors, respectively. These data indicated that, as a new mean to positively regulate adipocyte proliferation, the process of miR-145a-5p-inhibited adipogenesis may be regulated by DNA methylation.


Subject(s)
Adipocytes/cytology , Adipogenesis , DNA Methylation , MicroRNAs/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Cell Proliferation , Gene Expression Regulation , Male , Mice , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...