High-fat diet

高脂肪饮食
  • 文章类型: Journal Article
    已知高脂肪饮食(HFD)喂养会导致肠道屏障破坏,从而引发严重的肠道炎症性疾病。吲哚-3-醛(IAld)已成为减轻炎症反应和维持肠道稳态的潜在候选者。然而,ILD在HFD相关肠道破坏中的作用尚不清楚.在这项研究中,将48只7周龄雄性C57BL/6J小鼠分为四组:正常饮食(NCD)组接受NCD;HFD组喂养HFD;HFDIAld200组在HFD中补充了200mg/kgIAld;HFDIAld600组在HFD中补充了600mg/kgIAld。结果表明,膳食补充ILD可以改善脂肪积累和代谢紊乱,与肠道通透性降低有关。这种减少潜在地导致HFD喂养的小鼠中全身性炎症减少和肠屏障功能增强。此外,我们发现ILD通过体内和离体激活芳香烃受体(AHRs)促进肠干细胞(ISC)的增殖。这些发现表明Ild通过促进AHR介导的ISC增殖来恢复HFD诱导的肠屏障破坏。
    High-fat diet (HFD) feeding is known to cause intestinal barrier disruption, thereby triggering severe intestinal inflammatory disease. Indole-3-aldehyde (IAld) has emerged as a potential candidate for mitigating inflammatory responses and maintaining intestinal homeostasis. However, the role of IAld in the HFD-related intestinal disruption remains unclear. In this study, 48 7 week-old male C57BL/6J mice were assigned to four groups: the normal chow diet (NCD) group received a NCD; the HFD group was fed an HFD; the HFD + IAld200 group was supplemented with 200 mg/kg IAld in the HFD; and the HFD + IAld600 group was supplemented with 600 mg/kg IAld in the HFD. The results showed that dietary IAld supplementation ameliorated fat accumulation and metabolic disorders, which are associated with reduced intestinal permeability. This reduction potentially led to decreased systemic inflammation and enhanced intestinal barrier function in HFD-fed mice. Furthermore, we found that IAld promoted intestinal stem cell (ISC) proliferation by activating aryl hydrocarbon receptors (AHRs) in vivo and ex vivo. These findings suggest that IAld restores the HFD-induced intestinal barrier disruption by promoting AHR-mediated ISC proliferation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    我们先前的研究表明,重金属铅(Pb)暴露会加剧高脂饮食(HFD)诱导的代谢损伤,并显着消耗肠道微生物群衍生的代谢产物短链脂肪酸(SCFA)水平。然而,目前尚不清楚SCFA是否是促进铅暴露后不良后果的关键代谢产物。在这项研究中,我们探索了外源补充乙酸盐的效果,丙酸盐,和丁酸盐对铅暴露的HFD小鼠代谢紊乱模型的影响。我们发现三种SCFA干预措施可以减轻糖脂代谢紊乱和肝损伤,丁酸盐在改善肥胖相关症状方面效果最佳。所有三个SCFA都促进了Muribaculaceae和Muibaculum的丰度,醋酸盐特别丰富的Christensenellaceae,Blautia,和Ruminococus,和丁酸盐特别富含副曲霉,Rikenella,Prevotellaceae_UCG-001和拟杆菌,促进了SCFA生产形成良性循环的积极推进。此外,丁酸抑制革兰氏阴性菌大肠杆菌-志贺氏菌。所有这些事件通过G蛋白偶联受体(GPR)/组蛋白脱乙酰酶3(HDAC3)和脂多糖(LPS)/toll样受体4(TLR4)/核因子κ-B(NF-κB)途径之间的串扰减轻了肠道Th17/Treg失衡和炎症反应,并最终改善了肠道屏障功能。SCFA进一步上调单羧酸转运蛋白1(MCT1)和GPR43/腺苷5'-单磷酸活化蛋白激酶(AMPK)途径以抑制肝脂质积累。总的来说,SCFA,尤其是丁酸,是一种有效的调节剂,通过靶向肠道微生态来改善暴露于重金属的肥胖个体的代谢紊乱。
    Our previous study showed that heavy metal lead (Pb) exposure exacerbates high-fat-diet (HFD)-induced metabolic damage and significantly depletes the gut microbiota-derived metabolite short-chain fatty acid (SCFA) levels. However, it remains unclear whether SCFA is a key metabolite involved in accelerating adverse consequences after Pb exposure. In this study, we explored the effects of exogenous supplementation of acetate, propionate, and butyrate on a metabolic disorder model in Pb-exposed HFD mice. We found that three SCFA interventions attenuated glycolipid metabolism disorders and liver damage, with butyrate performing the best effects in improving obesity-related symptoms. All three SCFA promoted the abundance of Muribaculaceae and Muribaculum, acetate specifically enriched Christensenellaceae, Blautia, and Ruminococcus, and butyrate specifically enriched Parasutterella, Rikenella, Prevotellaceae_UCG-001, and Bacteroides, which contributed to the positive promotion of SCFA production forming a virtuous cycle. Besides, butyrate inhibited Gram-negative bacteria Escherichia-Shigella. All of these events alleviated the intestinal Th17/Treg imbalance and inflammatory response through crosstalk between the G protein-coupled receptor (GPR)/histone deacetylase 3 (HDAC3) and lipopolysaccharide (LPS)/toll-like receptors 4 (TLR4)/nuclear factor κ-B (NF-κB) pathways and ultimately improved the intestinal barrier function. SCFA further upregulated the monocarboxylate transporter 1 (MCT1) and GPR43/adenosine 5\'-monophosphate-activated protein kinase (AMPK) pathways to inhibit hepatic lipid accumulation. Overall, SCFA, especially butyrate, is an effective modulator to improve metabolic disorders in obese individuals exposed to heavy metals by targeting gut microecology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    作为一种多因素疾病,肥胖已成为世界上主要的健康问题之一,而且还在迅速增加。魔芋补充剂,作为一种方便的饮食疗法,已被证明能够调节肠道微生物群和改善肥胖。然而,魔芋通过肠道微生物群改善肥胖的具体机制还有待研究。在这项研究中,高脂饮食(HFD)用于诱导小鼠肥胖模型,使用16SrDNA测序和非靶向代谢组学研究魔芋对HFD诱导的肥胖小鼠肠道菌群和肠道代谢产物的影响。结果表明,魔芋可以减轻体重,脂肪组织重量,和脂质水平的高脂饮食诱导肥胖小鼠通过改变肠道菌群结构和肠道代谢谱。关联分析显示,魔芋的补充诱导肠道菌群的变化,导致7-脱氢胆固醇和6-磷酸海藻糖的上调,以及在次级胆汁酸生物合成途径中下调甘氨胆酸和乌索胆酸,最终导致肥胖的改善。其中,g_不动杆菌(GreengeneID:911888)可通过合成ERG3促进7-脱氢胆固醇的合成。g_Allobaculum(GreengeneID:271516)和g_Allobaculum(GreengeneID:259370)可以通过合成glvA来促进海藻糖6-磷酸的分解。此外,糖胆酸和熊胆酸的下调可能受Lachnoshispiliaceae_NK4A136_组上调的影响。总之,魔芋通过调节肠道微生物群对肠道代谢产物产生影响,从而在减轻高脂肪饮食引起的肥胖中起关键作用。
    As a multi-factorial disease, obesity has become one of the major health problems in the world, and it is still increasing rapidly. Konjac supplementation, as a convenient dietary therapy, has been shown to be able to regulate gut microbiota and improve obesity. However, the specific mechanism by which konjac improves obesity through gut microbiota remains to be studied. In this study, a high-fat diet (HFD) was used to induce a mouse obesity model, and 16S rDNA sequencing and an untargeted metabolomics were used to investigate the impact of konjac on gut microbiota and gut metabolites in HFD-induced obese mice. The results show that konjac can reduce the body weight, adipose tissue weight, and lipid level of high-fat diet induced obese mice by changing the gut microbiota structure and gut metabolic profile. Association analysis revealed that konjac supplementation induced changes in gut microbiota, resulting in the up-regulation of 7-dehydrocholesterol and trehalose 6-phosphate, as well as the down-regulation of glycocholic acid and ursocholic acid within the Secondary bile acid biosynthesis pathway, ultimately leading to improvements in obesity. Among them, g_Acinetobacter (Greengene ID: 911888) can promote the synthesis of 7-dehydrocholesterol by synthesizing ERG3. g_Allobaculum (Greengene ID: 271516) and g_Allobaculum (Greengene ID: 259370) can promote the breakdown of trehalose 6-phosphate by synthesizing glvA. Additionally, the down-regulation of glycocholic acid and ursocholic acid may be influenced by the up-regulation of Lachnospiraceae_NK4A136_group. In conclusion, konjac exerts an influence on gut metabolites through the regulation of gut microbiota, thereby playing a pivotal role in alleviating obesity induced by a high-fat diet.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肥胖已成为威胁人类健康的最严重的慢性疾病之一。它的发生和发展与肠道菌群密切相关,因为肠道菌群的破坏会促进内毒素的产生并诱导炎症反应。本研究旨在研究各种精制茶籽油的理化性质变化及其对高脂饮食(HFD)引起的肠道微生物群紊乱的影响。在本研究中,将接受HFD的C57BL/6J小鼠随机分为三组:HFD,T-TSO,N-TSO给予T-TSO和N-TSO小鼠传统精制和优化的茶籽油12周。数据显示,通过在70°C脱胶获得的茶籽油,50°C脱酸,在90°C下脱色,并在180°C下进行除臭(在0.06MPa下持续1小时)有效地去除杂质,同时最大程度地减少了活性成分的损失。此外,优化的茶籽油减轻了由HFD引起的脂肪积累和炎症反应,与传统的精炼方法相比,减少了肝脏组织的损伤。更重要的是,N-TSO可以作为膳食补充剂,以增强肠道微生物群的多样性和丰度,增加有益菌的存在(norank_f__Muibaculaceae,乳酸菌,和拟杆菌),同时减少致病菌(Alistipes和Mucispirillum)。因此,在HFD诱导的肥胖C57BL/6J小鼠中,与T-TSO饮食相比,N-TSO能更好地改善肥胖,这在缓解HFD诱导的肠道微生物群紊乱方面是有希望的。
    Obesity has become one of the most serious chronic diseases threatening human health. Its onset and progression are closely related to the intestinal microbiota, as disruption of the intestinal flora promotes the production of endotoxins and induces an inflammatory response. This study aimed to investigate the variations in the physicochemical properties of various refined tea seed oils and their impact on intestinal microbiota disorders induced by a high-fat diet (HFD) through dietary intervention. In the present study, C57BL/6J mice on a HFD were randomly divided into three groups: HFD, T-TSO, and N-TSO. T-TSO and N-TSO mice were given traditionally refined and optimized tea seed oil for 12 weeks. The data revealed that tea seed oil obtained through degumming at 70 °C, deacidification at 50 °C, decolorization at 90 °C, and deodorization at 180 °C (at 0.06 MPa for 1 h) effectively removed impurities while minimizing the loss of active ingredients. Additionally, the optimized tea seed oil mitigated fat accumulation and inflammatory responses resulting from HFD, and reduced liver tissue damage in comparison to traditional refining methods. More importantly, N-TSO can serve as a dietary supplement to enhance the diversity and abundance of intestinal microbiota, increasing the presence of beneficial bacteria (norank_f__Muribaculaceae, Lactobacillus, and Bacteroides) while reducing pathogenic bacteria (Alistipes and Mucispirillum). Therefore, in HFD-induced obese C57BL/6J mice, N-TSO can better ameliorate obesity compared with a T-TSO diet, which is promising in alleviating HFD-induced intestinal microbiota disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    全氟烷基和多氟烷基物质(PFAS),广泛应用于消费品中,与心血管疾病(CVD)的风险增加有关。随着高脂饮食患病率的增加,心血管疾病的常见危险因素,摄入高脂饮食的PFAS暴露人群不可避免地会增长,并且可能有更高的CVD风险.然而,潜在的毒性作用和作用方式仍然难以捉摸。我们构建了口服全氟辛烷磺酸(PFOS)的小鼠模型,典型的PFAS,高脂肪饮食.全氟辛烷磺酸暴露导致小鼠心脏心肌病和结构异常。此外,观察到从需氧到厌氧过程的能量代谢重塑特征。有趣的是,心脏中很少检测到全氟辛烷磺酸,但在血清中显示出高水平,提示全氟辛烷磺酸引起的心脏毒性的间接作用途径。我们进一步证明,全氟辛烷磺酸引起的循环炎症促进了心肌细胞的代谢重塑和收缩功能障碍。其中,PFOS刺激NF-κB和caspase-1介导的循环促炎巨噬细胞释放IL-1β。这项研究提供了有关PFAS引起的心脏风险的有价值的数据,这些风险与暴露人群的高脂肪饮食消耗增加有关。强调全氟辛烷磺酸对心脏影响的间接途径的重要性,基于内部暴露的分布。
    Per- and polyfluoroalkyl substances (PFAS), widely utilized in consumer products, have been linked to an increased risk of cardiovascular disease (CVD). With the increasing prevalence of high-fat diet, a common risk factor for CVD, the PFAS exposed populations who consume a high-fat diet will inevitably grow and may have a higher CVD risk. However, the potential toxic effect and mode of action remain elusive. We constructed a mouse model orally exposed to perfluorooctane sulfonate (PFOS), a prototypical PFAS, and fed a high-fat diet. PFOS exposure induced cardiomyopathy and structural abnormalities in the mice heart. Moreover, a characteristic of energy metabolism remodeling from aerobic to anaerobic process was observed. Interestingly, PFOS was rarely detected in heart but showed high level in serum, suggesting an indirect route of action for PFOS-caused cardiac toxicity. We further demonstrated that PFOS-caused circulating inflammation promoted metabolic remodeling and contractile dysfunction in cardiomyocytes. Wherein, PFOS stimulated the release of IL-1β from circulating proinflammatory macrophages mediated by NF-κB and caspase-1. This study provides valuable data on PFAS-induced cardiac risks associated with exposed populations with increasing high-fat diet consumption, highlighting the significance of indirect pathways in PFOS\'s impact on the heart, based on the distribution of internal exposure.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    骨质疏松症是一种常见的糖尿病后果,对患者的健康和生活质量产生负面影响。然而,旨在调节血糖和骨代谢的临床药物之间存在相互干扰。因此,寻找有效控制血糖和安全保护糖尿病患者骨骼健康的新治疗靶点至关重要。在这项研究中,当使用蛋白磷酸酶5(PP5)敲除(KO)小鼠时,给予高脂肪饮食的小鼠对骨质疏松症和糖尿病具有抗性.骨重建的血清标志物显示PP5KO小鼠对糖尿病引起的骨形成减少和骨吸收增加具有抗性。缺乏PP5抵抗糖尿病小鼠成骨细胞分化的减少和破骨细胞分化的增强,根据骨髓间充质干细胞的体外成骨细胞分化和骨髓源性巨噬细胞的破骨细胞分化。随后的研究表明,PP5缺乏增加成骨细胞分化的关键调节因子的表达,runt相关转录因子2,并降低核因子-κB配体/骨保护素途径的受体激活剂的活性,破骨细胞分化的关键调节信号通路。总之,我们首次发现PP5缺乏可以保护糖尿病小鼠免受骨质疏松的影响.
    Osteoporosis is a common diabetic consequence that negatively affects patients\' health and quality of life. Nevertheless, there is mutual interference between clinical drugs intended to regulate blood glucose and bone metabolism. Therefore, it is crucial to look for new treatment targets that effectively control blood glucose and safely protect the bone health of patients with diabetes. In this study, mice given a high-fat diet were shown to be resistant to osteoporosis and diabetes when protein phosphatase 5 (PP5) knockout (KO) mice were used. Serum markers of bone remodeling show that PP5 KO mice are resistant to decreased bone formation and increased bone resorption brought on by diabetes. The absence of PP5 resists the reduction of osteoblast differentiation and the enhancement of osteoclast differentiation in diabetic mice, according to the in vitro osteoblast differentiation of bone mesenchymal stem cells and osteoclast differentiation of bone marrow-derived macrophages. Subsequent investigation revealed that PP5 deficiency increases the expression of the key regulator of osteoblast differentiation, runt-related transcription factor 2, and decreases the activity of the receptor activator of the nuclear factor-κB ligand/osteoprotegerin pathway, a crucial regulatory signaling pathway for osteoclast differentiation. In conclusion, we discovered that PP5 deficiency protects diabetic mice against osteoporosis for the first time.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:动脉粥样硬化(AS)是最常见的心血管疾病,负担非常高。高脂饮食(HFD)是一种流行的饮食行为,而低剂量辐射(LDR)是一种环境物理因素。有证据表明HFD可能会加剧动脉粥样硬化的发作。尚不完全清楚HFD和LDR的联合作用是否对动脉粥样硬化的发展具有潜力。
    方法:在本研究中,ApoE-/-小鼠作为动脉粥样硬化模型动物,研究HFD和LDR的联合作用(10×0.01Gy,或20×0.01Gy)对血管病变。多普勒超声成像,H&E染色,油红O染色,西方印迹,和免疫组织化学(IHC)用于评估前动脉粥样硬化作用。LC-MS用于检测非靶向脂质组学。
    结果:在累积剂量为0.2Gy的低剂量辐射的长期暴露显著增加了ApoE-/-小鼠的血管僵硬和主动脉损伤的发生。观察到HFD和LDR在动脉粥样硬化发展中的协同作用,这可能与脂质代谢的生态失调和炎症信号系统的刺激有关。此外,LDR而非HFD可以通过增加胞质线粒体DNA的产量以及cGAS蛋白的表达来激活cGAS-STING信号传导。cGAS-STING信号的激活引发IFN-α/-β的释放,它在动脉粥样硬化斑块的形成中起着炎症放大器的作用。
    结论:当前的研究为LDR导致动脉粥样硬化发展的风险和机制提供了新的见解,LDR和HFD的联合作用与cGAS-STING信号通路有关。
    BACKGROUND: Atherosclerosis (AS) is the most prevalent cardiovascular disease, with an exceptionally high burden. High-fat diet (HFD) is a popular diet behavior, whereas low-dose radiation (LDR) is an environmental physical factor. There is evidence to suggest that an HFD may exacerbate the onset of atherosclerosis. Whether the combination effect of HFD and LDR would have potential on atherosclerosis development remains incompletely unclear.
    METHODS: In this study, ApoE-/- mice were used as atherosclerosis model animals to investigate the combination effects of HFD and LDR (10 × 0.01Gy, or 20 × 0.01Gy) on vascular lesions. Doppler ultrasound imaging, H&E staining, oil red O staining, western blotting, and immunohistochemistry (IHC) were used to assess the pro-atherosclerotic effects. LC-MS was used to detect the non-targeted lipidomic.
    RESULTS: Long-term exposure of low-dose radiation at an accumulated dose of 0.2Gy significantly increased the occurrence of vascular stiffness and the aortic lesion in ApoE-/- mice. The synergistic effect of HFD and LDR was observed in the development of atherosclerosis, which might be linked to both the dysbiosis of lipid metabolism and the stimulation of the inflammatory signaling system. Moreover, LDR but not HFD can activate the cGAS-STING signaling through increasing the yield of cytosolic mitochondrial DNAs as well as the expression of cGAS protein. The activation of cGAS-STING signal triggers the release of IFN-α/-β, which functions as an inflammatory amplifier in the formation of atherosclerotic plaque.
    CONCLUSIONS: The current study offers fresh insights into the risks and mechanism that underlie the development of atherosclerosis by LDR, and there is a combination effect of LDR and HFD with the involvement of cGAS-STING signal pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    高脂饮食(HFD)有助于各种炎症和代谢疾病的发病机理。先前的研究证实,在HFD条件下,眶外泪腺(ELGs)可能受损,促炎巨噬细胞(Mps)明显浸润。然而,ELG中HFD和Mps极化之间的关系仍未被探索。我们首先通过RNA测序鉴定并验证了PPAR-γ在饲喂ND和HFD的鼠ELG中的差异表达。使用Schirmer试验测量泪液分泌。通过油红O染色和透射电子显微镜观察ELG内的脂质液滴沉积。通过定量RT-PCR测定Mps表型,免疫荧光,和流式细胞术分析。用棕榈酸(PA)建立了Mps的体外高脂培养体系,与收集的上清液用于与泪腺腺泡细胞共培养。通过ELISA测定基因表达,免疫荧光,免疫组织化学,定量RT-PCR,和蛋白质印迹分析。吡格列酮通过增加ELGs中的PPAR-γ水平减少HFD诱导的M1占优势的浸润,从而减轻脂质沉积并增强泪液分泌。体外试验表明,PPAR-γ激动剂在PA诱导的Mps中,将Mps从M1占优势的表型转变为M2占优势的表型,减少LGAC中的脂质合成并促进脂质分解代谢,从而减轻ELGs内的脂质代谢紊乱。相反,PPAR-γ拮抗剂诱导相反的作用。总之,泪腺对高脂肪和脂质代谢紊乱高度敏感。ELGs中PPAR-γ表达下调诱导Mps向主要M1表型极化,通过NF-κb/ERK/JNK/P38途径导致脂质代谢紊乱和炎症反应。
    A high-fat diet (HFD) contributes to the pathogenesis of various inflammatory and metabolic diseases. Previous research confirms that under HFD conditions, the extraorbital lacrimal glands (ELGs) can be impaired, with significant infiltration of pro-inflammatory macrophages (Mps). However, the relationship between HFD and Mps polarization in the ELGs remains unexplored. We first identified and validated the differential expression of PPAR-γ in murine ELGs fed ND and HFD through RNA sequencing. Tear secretion was measured using the Schirmer test. Lipid droplet deposition within the ELGs was observed through Oil Red O staining and transmission electron microscopy. Mps phenotypes were determined through quantitative RT-PCR, immunofluorescence, and flow cytometric analysis. An in vitro high-fat culture system for Mps was established using palmitic acid (PA), with supernatants collected for co-culture with lacrimal gland acinar cells. Gene expression was determined through ELISA, immunofluorescence, immunohistochemistry, quantitative RT-PCR, and Western blot analysis. Pioglitazone reduced M1-predominant infiltration induced by HFD by increasing PPAR-γ levels in ELGs, thereby alleviating lipid deposition and enhancing tear secretion. In vitro tests indicated that PPAR-γ agonist shifted Mps from M1-predominant to M2-predominant phenotype in PA-induced Mps, reducing lipid synthesis in LGACs and promoting lipid catabolism, thus alleviating lipid metabolic disorders within ELGs. Conversely, the PPAR-γ antagonist induced opposite effects. In summary, the lacrimal gland is highly sensitive to high-fat and lipid metabolic disorders. Downregulation of PPAR-γ expression in ELGs induces Mps polarization toward predominantly M1 phenotype, leading to lipid metabolic disorder and inflammatory responses via the NF-κb/ERK/JNK/P38 pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    非酒精性脂肪性肝病(NAFLD)现在被认为是全球最普遍的肝病。豌豆白蛋白(PA)已显示出对减少肥胖和改善葡萄糖代谢的积极影响。在这项研究中,采用高脂饮食(HFD)诱导的NAFLD小鼠模型,研究PA对NAFLD的影响,并探讨其潜在机制.研究结果表明,接受HFD的小鼠发生了明显的脂肪肝改变。PA干预显著降低血清TC达26.81%,TG下降43.55%,LDL-C下降57.79%。在饲喂HFD的小鼠中,它还将HDL-C水平提高了1.2倍,并将血清ALT降低了37.94%,AST降低了31.21%。这些变化有助于减少肝脂肪变性和脂质积累。此外,PA改善胰岛素抵抗并抑制肝脏氧化应激和炎症反应。机制研究表明,PA通过激活AMPKα和ACC的磷酸化减轻HFD诱导的NAFLD中的脂质积累,抑制SREBF1和FASN的表达以减少肝脏脂肪生成,增加ATGL的表达,PPARα,和PPARγ促进脂解和脂肪酸氧化。这些结果表明,PA可以作为减轻NAFLD的膳食补充剂,为NAFLD干预中PA的合理摄入提供理论基础。
    Non-alcoholic fatty liver disease (NAFLD) is now recognized as the most prevalent liver disease globally. Pea albumin (PA) has demonstrated positive impacts on reducing obesity and improving glucose metabolism. In this research, a mouse model of NAFLD induced by a high-fat diet (HFD) was employed to examine the impact of PA on NAFLD and explore its potential mechanisms. The findings revealed that mice subjected to a HFD developed pronounced fatty liver alterations. The intervention with PA significantly lowered serum TC by 26.81%, TG by 43.55%, and LDL-C by 57.79%. It also elevated HDL-C levels by 1.2 fold and reduced serum ALT by 37.94% and AST by 31.21% in mice fed a HFD. These changes contributed to the reduction in hepatic steatosis and lipid accumulation. Additionally, PA improved insulin resistance and inhibited hepatic oxidative stress and inflammatory responses. Mechanistic studies revealed that PA alleviated lipid accumulation in HFD-induced NAFLD by activating the phosphorylation of AMPKα and ACC, inhibiting the expression of SREBF1 and FASN to reduce hepatic lipogenesis, and increasing the expression of ATGL, PPARα, and PPARγ to promote lipolysis and fatty acid oxidation. These results indicate that PA could serve as a dietary supplement for alleviating NAFLD, offering a theoretical foundation for the rational intake of PA in NAFLD intervention.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    由于食用高脂肪饮食(HFD),出现了各种健康问题,特别是它们对线粒体动力学和亚序列认知功能的有害影响。特别是,线粒体裂变可以作为调节皮质炎症和神经焦亡的上游信号。我们的研究旨在验证神经炎症在HFD诱导的认知功能障碍的发病机理中的存在,并证明白藜芦醇(RSV)通过调节皮质线粒体裂变来减轻神经缺陷。将50只雄性SD大鼠随机分为五组:对照组(Cont,正常啮齿动物饮食26周);高脂饮食(HFD);饮食调整(HFDND);白藜芦醇干预(HFDR);联合干预(HFDNDR)26周。空间学习和记忆功能,脊柱密度,NLRP3炎性体相关蛋白,检测脑线粒体动力学和SIRT1/PGC-1α信号通路的mRNA和蛋白表达。此外,检测到暴露于棕榈酸(PA)或Drp1抑制剂(Mdivi-1)的PC12细胞中活性氧(ROS)的积累和线粒体膜电位(MMP)的改变,以反映线粒体功能。研究结果表明,长期治疗RSV可改善HFD引起的认知缺陷和神经元损伤,可能归因于SIRT1/PGC-1α轴的激活。我们进一步表明,在PA(200μM)处理的PC12细胞中NLRP3炎性体的激活可以被Mdivi-1抑制。更重要的是,Mdivi-1(10μM)通过逆转Drp1介导的异常线粒体裂变降低细胞内ROS水平并增强MMP。总结一下,这些结果清楚地表明,HFD抑制SIRT1/PGC-1α途径,这导致了线粒体动力学的失衡和NLRP3介导的焦亡的发生。RSV可能通过触发SIRT1/PGC-1α轴来减轻这种影响,防止异常线粒体裂变,从而抑制NLRP3炎症途径的激活。
    Various health issues have emerged due to consuming high-fat diets (HFD), particularly the detrimental impact they have on mitochondrial dynamics and subsequet cognition functions. Specially, mitochondrial fission can serve as an upstream signal in the regulation of cortical inflammation and neural pyroptosis. Our study was designed to verify the existence of neuroinflammation in the pathogenesis of HFD-induced cognitive dysfunction and demonstrated that resveratrol (RSV) attenuated neural deficits via regulation of cortical mitochondrial fission. A total of 50 male Sprague Dawley rats were randomly divided into five groups: control (Cont, 26 weeks on normal rodent diet); high-fat diet (HFD); dietary adjustments (HFD + ND); resveratrol intervention (HFD + R); joint intervention (HFD + ND + R) for 26 weeks. The spatial learning and memory function, spine density, NLRP3 inflammasome associated protein, mRNA and protein expression involved in mitochondrial dynamics and SIRT1/PGC-1α signaling pathway in brain were measured. Furthermore, reactive oxygen species (ROS) accumulation and resultant mitochondrial membrane potential (MMP) alteration in PC12 cells exposed to palmitic acid (PA) or Drp1 inhibitor (Mdivi-1) were detected to reflect mitochondrial function. The findings suggested that prolonged treatment of RSV improved cognitive deficits and neuronal damage induced by HFD, potentially attributed to activation of the SIRT1/PGC-1α axis. We further indicated that the activation of the NLRP3 inflammasome in PA (200 μM) treated PC12 cells could be inhibited by Mdivi-1. More importantly, Mdivi-1 (10 μM) reduced intracellular ROS levels and enhanced MMP by reversing Drp1-mediated aberrant mitochondrial fission. To summarize, those results clearly indicated that a HFD inhibited the SIRT1/PGC-1α pathway, which contributed to an imbalance in mitochondrial dynamics and the onset of NLRP3-mediated pyroptosis. This effect was mitigated by the RSV possibly through triggering the SIRT1/PGC-1α axis, prevented aberrant mitochondrial fission and thus inhibited the activation of the NLRP3 inflammatory pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号