PPARγ

PPAR γ
  • 文章类型: Journal Article
    背景:无法正确识别结直肠癌(CRC)病理生理学的复杂性,对开发新药物和靶向治疗方法的进展产生了重大影响。脂质氧化和脂肪酸生物合成过程中的不平衡是CRC发展的重要风险因素。特异性靶向过氧化物酶体增殖物激活受体γ(PPARγ)及其下游反应元件的治疗干预,响应脂质代谢,已经发现促进肿瘤的生长,并在癌症患者中显示出显著的临床优势。
    方法:进行了临床CRC样本和广泛的体外和体内实验,以通过一系列生化测定确定ZDHHC6及其下游靶标的作用,分子分析方法和脂质代谢组学测定,等。结果:为了研究ZDHHC6对CRC进展的影响,并确定ZDHHC6是否是调节脂肪酸合成的棕榈酰转移酶,直接棕榈酰化并稳定PPARγ,而这种稳定又激活了ACLY转录相关的代谢途径。在这项研究中,我们证明PPARγ在其DNA结合域(DBD)部分经历棕榈酰化。这种与脂质相关的修饰通过防止PPARγ蛋白的去稳定化来增强其稳定性。因此,棕榈酰化的PPARγ抑制其由溶酶体诱导的降解,并促进其转运到细胞核中。此外,我们已经确定锌指-天冬氨酸-组氨酸-半胱氨酸6(ZDHHC6)是脂肪酸生物合成的关键控制因子。ZDHHC6直接与棕榈酰基相互作用并添加棕榈酰基以在PPARγ的DBD结构域内的Cys-313位点稳定PPARγ。因此,这种棕榈酰化导致ATP柠檬酸裂解酶(ACLY)的表达增加。此外,我们的发现表明,ZDHHC6积极刺激脂肪酸的产生,并在结直肠癌的发展中起作用。然而,在体内试验中,当ZDHHC6的表达被抑制时,我们观察到致癌效应显著降低.重要的是,在CRC,ZDHHC6的高表达与PPARγ的表达呈正相关。此外,ZDHHC6的高表达与CRC的严重程度有关,提示预后不良.
    结论:我们发现了脂质生物合成受ZDHHC6控制的机制,包括PPARγ-ACLY在CRC进展中的信号传导。这一发现为通过阻断ZDHHC6作为潜在的治疗方法来靶向脂质合成提供了理由。
    BACKGROUND: The failure of proper recognition of the intricate nature of pathophysiology in colorectal cancer (CRC) has a substantial effect on the progress of developing novel medications and targeted therapy approaches. Imbalances in the processes of lipid oxidation and biosynthesis of fatty acids are significant risk factors for the development of CRC. Therapeutic intervention that specifically targets the peroxisome proliferator-activated receptor gamma (PPARγ) and its downstream response element, in response to lipid metabolism, has been found to promote the growth of tumors and has shown significant clinical advantages in cancer patients.
    METHODS: Clinical CRC samples and extensive in vitro and in vivo experiments were carried out to determine the role of ZDHHC6 and its downstream targets via a series of biochemical assays, molecular analysis approaches and lipid metabolomics assay, etc. RESULTS: To study the effect of ZDHHC6 on the progression of CRC and identify whether ZDHHC6 is a palmitoyltransferase that regulates fatty acid synthesis, which directly palmitoylates and stabilizes PPARγ, and this stabilization in turn activates the ACLY transcription-related metabolic pathway. In this study, we demonstrate that PPARγ undergoes palmitoylation in its DNA binding domain (DBD) section. This lipid-related modification enhances the stability of PPARγ protein by preventing its destabilization. As a result, palmitoylated PPARγ inhibits its degradation induced by the lysosome and facilitates its translocation into the nucleus. In addition, we have identified zinc finger-aspartate-histidine-cysteine 6 (ZDHHC6) as a crucial controller of fatty acid biosynthesis. ZDHHC6 directly interacts with and adds palmitoyl groups to stabilize PPARγ at the Cys-313 site within the DBD domain of PPARγ. Consequently, this palmitoylation leads to an increase in the expression of ATP citrate lyase (ACLY). Furthermore, our findings reveals that ZDHHC6 actively stimulates the production of fatty acids and plays a role in the development of colorectal cancer. However, we have observed a significant reduction in the cancer-causing effects when the expression of ZDHHC6 is inhibited in in vivo trials. Significantly, in CRC, there is a strong positive correlation between the high expression of ZDHHC6 and the expression of PPARγ. Moreover, this high expression of ZDHHC6 is connected with the severity of CRC and is indicative of a poor prognosis.
    CONCLUSIONS: We have discovered a mechanism in which lipid biosynthesis is controlled by ZDHHC6 and includes the signaling of PPARγ-ACLY in the advancement of CRC. This finding provides a justification for targeting lipid synthesis by blocking ZDHHC6 as a potential therapeutic approach.
    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
    肾纤维化是几乎所有进行性形式的慢性肾病(CKD)的共同结果,一个重大的社会健康问题。谷氨酸脱氢酶(GDH)1是谷氨酰胺代谢中催化谷氨酸可逆转化为α-酮戊二酸和氨的关键酶之一。然而,其在肾纤维化中的功能尚未得到证实。在这项研究中,GDH1表达在患有肾脏疾病的儿童和CKD动物模型的肾组织中显著下调。在体内,R162(GDH1抑制剂)的使用显着改善了肾脏纤维化,如天狼星红和马森三色染色所示。这些发现与单侧输尿管梗阻(UUO)和5/6肾切除术(5/6Nx)模型中肾脏纤维化指标的受损表达一致。体外,沉默GDH1或用R162预处理抑制了用转化生长因子β1(TGF-β1)处理的组织肾脏近端肾小管细胞(TKPTS)的纤维化指标的诱导,而激活GDH1则恶化了TGF-β1的诱导影响。使用RNA序列,荧光素酶报告分析和Biacore分析,我们证明GDH1与过氧化物酶体增殖物激活受体γ(PPARγ)相互作用并阻断其转录活性,独立于蛋白质的表达。此外,R162处理增强PPARγ转录活性,阻断该信号通路可逆转R162的保护作用。最后,我们发现R162治疗或沉默GDH1大大降低了活性氧(ROS)和脂质积累。这些发现得出结论,抑制GDH1或R162治疗可以通过增加PPARγ转录活性以控制脂质积累和氧化还原平衡来预防肾纤维化。
    Renal fibrosis is the common outcome of practically all progressive forms of chronic kidney disease (CKD), a significant societal health concern. Glutamate dehydrogenase (GDH) 1 is one of key enzymes in glutamine metabolism to catalyze the reversible conversion of glutamate to α-ketoglutarate and ammonia. However, its function in renal fibrosis has not yet been proven. In this study, GDH1 expression was significantly downregulated in kidney tissues of both children with kidney disease and animal models of CKD. In vivo, the use of R162 (a GDH1 inhibitor) significantly improved renal fibrosis, as indicated by Sirius red and Masson trichrome staining. These findings are consistent with the impaired expression of fibrosis indicators in kidneys from both the unilateral ureteral obstruction (UUO) and 5/6 nephrectomy (5/6 Nx) models. In vitro, silencing GDH1 or pretreatment with R162 inhibited the induction of fibrosis indicators in tissue kidney proximal tubular cells (TKPTS) treated with Transforming growth factor Beta 1 (TGF-β1), whereas activating GDH1 worsened TGF-β1\'s induction impact. Using RNA-sequence, luciferase reporter assays and Biacore analysis, we demonstrated that GDH1 interacts with Peroxisome proliferator-activated receptor gamma (PPARγ) and blocks its transcriptional activity, independent of the protein\'s expression. Additionally, R162 treatment boosted PPARγ transcriptional activity, and blocking of this signaling pathway reversed R162\'s protective effect. Finally, we discovered that R162 treatment or silencing GDH1 greatly lowered reactive oxygen species (ROS) and lipid accumulation. These findings concluded that suppressing GDH1 or R162 treatment could prevent renal fibrosis by augmenting PPARγ transcriptional activity to control lipid accumulation and redox balance.
    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
    脓毒症是由宿主对感染的反应失调引起的全身性炎症反应综合征。CD4+T细胞减少对脓毒症诱导的免疫抑制至关重要。焦亡,程序性坏死,与淋巴细胞减少有关.过氧化物酶体增殖物激活受体γ(PPARγ)受上游mTOR调节,发挥抗焦亡作用。探讨mTOR-PPARγ对脓毒症诱导的CD4+T细胞耗竭的潜在影响及其机制。我们通过盲肠结扎穿孔(CLP)脓毒症小鼠模型观察到mTOR激活和焦亡与PPARγ-Nrf抑制。进一步的机制研究使用具有T细胞特异性敲除mTOR或结节性硬化症复合物1(TSC1)的基因修饰小鼠。提示mTOR通过负向调控PPARγ-Nrf2信号通路,介导脓毒症小鼠CD4+T细胞凋亡。一起来看,mTOR-PPARγ-Nrf2信号介导脓毒症CD4+T细胞凋亡,有助于CD4+T细胞消耗和免疫抑制。
    Sepsis is a systemic inflammatory response syndrome caused by a dysregulated host response to infection. CD4+T cell reduction is crucial to sepsis-induced immunosuppression. Pyroptosis, a programmed necrosis, is concerned with lymphocytopenia. Peroxisome proliferator-activated receptor gamma (PPARγ) regulated by upstream mTOR, exerts anti-pyroptosis effects. To investigate the potential effects of mTOR-PPARγ on sepsis-induced CD4+T cell depletion and the underlying mechanisms, we observed mTOR activation and pyroptosis with PPARγ-Nrf suppression through cecal ligation and puncture (CLP) sepsis mouse model. Further mechanism research used genetically modified mice with T cell-specific knockout mTOR or Tuberous Sclerosis Complex1 (TSC1). It revealed that mTOR mediated CD4 + T cell pyroptosis in septic mice by negatively regulating the PPARγ-Nrf2 signaling pathway. Taken together, mTOR-PPARγ-Nrf2 signaling mediated the CD4+ T cell pyroptosis in sepsis, contributing to CD4+T cell depletion and immunosuppression.
    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
    慢性前列腺炎引起的过度炎症和氧化应激(OS)损害严重影响男性的生活质量。然而,它的治疗仍然是一个主要的临床挑战。因此,确定可以减少慢性前列腺炎和氧化应激目标的药物是紧迫和必要的。CXCR4是一种经典的趋化因子受体,与炎症的发生和发展密切相关。这项研究旨在阐明CXCR4如何影响前列腺炎的消退和进展。HE染色评价CXCR4对慢性前列腺炎的疗效,免疫组织化学,免疫荧光,PCR,和TUNEL分析。此外,CXCR4对代谢的影响也通过监测体重来评估,体温,食物摄入量,和LC/MS此外,染色质免疫沉淀,蛋白质印迹,进行了双荧光素酶报告基因测定,以阐明CXCR4通过PPARγ调节Fads2转录的机制。最后,ROS,DHE,mito-tracker,和ATP用于验证α-亚麻酸对前列腺上皮细胞OS的保护作用。结果表明,抑制CXCR4可以有效缓解小鼠前列腺炎。此外,下调CXCR4的表达可以显著减少小鼠前列腺组织的炎症细胞浸润,降低DNA损伤标记水平的升高,MDA和4-HNE,减轻前列腺上皮细胞凋亡。此外,用PPARγ抑制剂处理CXCR4敲低小鼠显示出上述表型的不同程度的变化。机械上,PPARγ蛋白转位到细胞核,并作为转录因子调节Fads2的表达,从而改变PUFA代谢。此外,体外实验表明,α-亚麻酸可通过保护线粒体功能和增强前列腺上皮细胞的抗氧化能力,有效减轻OS损伤和RWPE-1细胞凋亡。总之,降低CXCR4水平可减轻慢性前列腺炎的炎症和OS损伤。
    Chronic prostatitis-induced excessive inflammation and oxidative stress (OS) damage substantially affect men\'s quality of life. However, its treatment remains a major clinical challenge. Therefore, the identification of drugs that can decrease chronic prostatitis and oxidative stress targets is urgent and essential. CXCR4 is a classic chemokine receptor that is crucially associated with the occurrence and development of inflammation. This investigation aimed to elucidate how CXCR4 affects prostatitis regression and progression. The effect of CXCR4 on chronic prostatitis was evaluated by HE staining, immunohistochemistry, immunofluorescence, PCR, and TUNEL analyses. Furthermore, CXCR4 influence on metabolism was also evaluated by monitoring body weight, body temperature, food intake, and LC/MS. Additionally, chromatin immunoprecipitation, Western blot, and double luciferase reporter gene assays were carried out to elucidate the mechanism by which CXCR4 modulates Fads2 transcription by PPARγ. Lastly, ROS, DHE, mito-tracker, and ATP were utilized to validate the α-linolenic acid\'s protective effect against OS in prostate epithelial cells. It was revealed that the inhibition of CXCR4 can effectively alleviate prostatitis in mice. Furthermore, downregulating CXCR4 expression can markedly reduce the inflammatory cell infiltration in mouse prostates, decrease the elevated levels of DNA damage markers,MDA and 4-HNE, and mitigate apoptosis of prostatic epithelial cells. Moreover, treatment of CXCR4 knockdown mice with a PPARγ inhibitor revealed different degrees of changes in the above phenotypes. Mechanistically, the PPARγ protein translocates to the nucleus and serves as a transcription factor to regulate Fads2 expression, thereby altering PUFA metabolism. Additionally, in vitro experiments indicated that α-linolenic acid can effectively alleviate OS damage and RWPE-1 cell apoptosis by protecting mitochondrial function and enhancing the antioxidant capacity of prostatic epithelial cells. In conclusion, reducing the levels of CXCR4 can alleviate inflammation and OS damage in chronic prostatitis.
    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
    本课题组前期研究表明,川芎人参汤(CRD)具有治疗AD的作用,但其作用的确切机制尚不清楚。本研究旨在探讨CRD在AD神经炎症中的作用及其机制。
    Morris水迷宫(MWM)测试用于评估AD小鼠的记忆和学习能力。HE和Nissl染色观察小鼠神经细胞。免疫组化染色检测Iba-1和CD86的表达。利用UHPLC-MS/MS代谢组学技术和KEGG分析CRD抗AD的代谢途径。脂多糖(LPS)引诱BV2小胶质细胞构建神经炎模子。免疫荧光和流式细胞术检测Iba-1和CD86的表达。ELISA法检测TNF-α和IL-1β的含量。Westernblot法检测PPARγ的表达,p-NF-κBp65,NF-κBp65蛋白和炎症细胞因子iNOS和COX-2在PPARγ/NF-κB途径中有或没有PPARγ抑制剂GW9662。
    CRD改善了3×Tg-AD小鼠的学习记忆能力,修复了海马中受损的神经细胞,减少了海马和皮质区域Iba-1和CD86阳性区域的面积,以及降低小鼠血清IL-1β和TNF-α水平。含CRD的血清中Iba-1的表达水平明显降低,TNF-α和IL-1β水平明显降低,显著增加PPARγ的蛋白表达,显著降低了iNOS的蛋白表达,BV2小胶质细胞的COX-2和p-NF-κBp65。添加PPARγ抑制剂GW9662后,含CRD的血清对NF-κB活化的抑制作用明显减弱。
    CRD可以激活PPARγ,调节PPARγ/NF-κB信号通路,抑制小胶质细胞过度激活并减少AD神经炎症。
    UNASSIGNED: Previous studies of our research group have shown that Chuanxiong Renshen Decoction (CRD) has the effect of treating AD, but the exact mechanism of its effect is still not clarified. The aim of this study was to investigate the effect and mechanism of CRD on AD neuroinflammation.
    UNASSIGNED: Morris Water Maze (MWM) tests were employed to assess the memory and learning capacity of AD mice. HE and Nissl staining were used to observe the neural cells of mice. The expression of Iba-1 and CD86 were detected by immunohistochemical staining. Utilize UHPLC-MS/MS metabolomics techniques and the KEGG to analyze the metabolic pathways of CRD against AD. Lipopolysaccharide (LPS) induced BV2 microglia cells to construct a neuroinflammatory model. The expression of Iba-1 and CD86 were detected by immunofluorescence and flow cytometry. The contents of TNF-α and IL-1β were detected by ELISA. Western blot assay was used to detect the expression of PPARγ, p-NF-κB p65, NF-κB p65 proteins and inflammatory cytokines iNOS and COX-2 in PPARγ/NF-κB pathway with and without PPARγ inhibitor GW9662.
    UNASSIGNED: CRD ameliorated the learning and memory ability of 3×Tg-AD mice, repaired the damaged nerve cells in the hippocampus, reduced the area of Iba-1 and CD86 positive areas in both the hippocampus and cortex regions, as well as attenuated serum levels of IL-1β and TNF-α in mice. CRD-containing serum significantly decreased the expression level of Iba-1, significantly reduced the levels of TNF-α and IL-1β, significantly increased the protein expression of PPARγ, and significantly decreased the proteins expression of iNOS, COX-2 and p-NF-κB p65 in BV2 microglia cells. After addition of PPARγ inhibitor GW9662, the inhibitory effect of CRD-containing serum on NF-κB activation was significantly weakened.
    UNASSIGNED: CRD can activate PPARγ, regulating PPARγ/NF-κB signaling pathway, inhibiting microglia over-activation and reducing AD neuroinflammation.
    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
    白色脂肪组织特异性适体Adipo8可以特异性结合成熟脂肪细胞或组织并抑制脂肪生成。在这项研究中,我们使用mRNA水平测序研究了Adipo8干预对脂肪形成过程中转录组的影响,分析了Adipo8抑制脂肪生成的机制。结果表明,Adipo8可以抑制分化的3T3-L1细胞的脂质形成,并下调PPARγ和C/EBPα。Adipo8干预后3个T3-L1细胞的转录组mRNA测序显示,Adipo8可能通过下调Acsl1和Plin1抑制脂肪酸代谢和PPAR信号通路来抑制脂肪生成的生物学功能。之后,利用Spacer18连接优化后截短的Adipo8,我们构建了一个二价适配体Adipo8cBand,生物效应,分化和成熟3T3-L1细胞中适体之间的生物学稳定性。在细胞层面,亲和力,生物效应,在3T3-L1细胞中,Adipo8cB的血清稳定性优于Adipo8。然后,我们研究了Adipo8cB作为体内脂质抑制药物的生物学特性,使用C57BL/6J小鼠饮食诱导的肥胖。体重,血糖,脂质水平,肝功能,葡萄糖耐量,观察并比较各组小鼠干预后的二价适配体Adipo8cB和Adipo8等相关指标。Adipo8cB和Adipo8都有效地防止了由饮食引起的肥胖引起的小鼠脂肪积累引起的体重增加。同时也降低了血脂水平,改善葡萄糖耐量,防止肝脏脂肪变性,此外,Adipo8cB比Adipo8具有更好的效果。
    The white adipose tissue-specific aptamer Adipo8 can specificity bindwith mature adipocytes or tissues and inhibit adipogenesis.In this research, we exploredthe effect of Adipo8 intervention on the transcriptome in the process of adipogenesis using mRNA-level sequencing,analyzed the mechanism ofAdipo8 ininhibiting adipogenesis. The results showed that Adipo8 can inhibit lipid formation and downregulate PPARγ and C/EBPα in differentiated 3 T3-L1 cells. Transcriptome mRNA sequencing of 3 T3-L1 cells after Adipo8 interventionrevealed that Adipo8 might inhibit the biological function of adipogenesis by downregulating Acsl1 and Plin1 to inhibit fatty acid metabolism and PPAR signaling pathways.After that, using Spacer18 to connect the optimized and truncated Adipo8, we constructed a bivalent aptamer Adipo8cBand compared the affinity, biological effects, and biological stability between the aptamers in differentiated and mature 3 T3-L1 cells. At the cellular level,the affinity, biological effects, and serum stability of Adipo8cB were verified to be superior to those of Adipo8in 3 T3-L1 cells.We then investigated the biological properties of Adipo8cB as a lipid-inhibiting drug invivo, using C57BL/6J mice with diet-induced obesity. The body weight, blood sugar, lipid levels, liver function, glucose tolerance, and other related indicators in each group of mice were observed and compared after intervention with the bivalent aptamers Adipo8cB and Adipo8. Both Adipo8cB and Adipo8 effectively prevented weight gain caused by fat accumulation in micewith diet induced obesity, while also reducing blood lipid levels, improving glucose tolerance, and protecting against liver steatosis, moreover, Adipo8cB has a better effect than Adipo8.
    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
    背景:我们以前的研究证明了痤疮酸杆菌之间的密切关系(C.痤疮),氧化应激,和痤疮炎症。丁香酸(SA)是一种广泛用于抗菌的植物,抗炎,和抗氧化活性,但缺乏痤疮的数据。本研究旨在探讨SA对痤疮C.acnes诱导的痤疮炎症的作用及机制。
    方法:使用SA暴露HaCaT角质形成细胞后,我们重新评估了SA对细胞活力的影响,细胞凋亡,ROS,CAT,SOD,以及热灭活的痤疮梭菌处理的HaCaT细胞中的其他炎性变量。接下来,诱导小鼠痤疮炎症,向ICR小鼠的右耳皮内注射活痤疮梭菌。然后检查SA对该炎症的作用。此外,我们通过ELISA法探讨了SA对PPARγ/Nrf2和NLRP3/caspase-1/IL-1β通路的作用机制,免疫荧光显微镜,和蛋白质印迹分析。
    结果:热杀伤痤疮丙酸杆菌引发显著的细胞凋亡,ROS生产,白细胞介素(IL)-1β,IL-18、IL-6和TNF-α释放,降低SOD和CAT活性,并上调HaCaT细胞中蛋白质的表达,包括上调IL-1β,PPARγ,Nrf2,HO-1,NQO1,NLRP3和caspase-1,而SA通过部分削弱PPARγ激活来抑制这些作用。此外,PPARγ沉默减少痤疮杆菌诱导的IL-1β分泌和细胞内ROS的产生,下调Nrf2的表达。Nrf2激活剂(SFN)通过抗氧化机制增强抗炎活性,促进细胞内ROS的产生,降低SOD和CAT活性,促进ROS的增加,HO-1、NQO1和IL-1β水平,而PPARγ抑制剂(GW662)在热杀死的痤疮梭菌处理的细胞中有效地抑制这种作用。最后,SA还表现出耳朵发红的显着改善,肿胀,和PPARγ的表达,体内NLRP3和IL-1β。
    结论:SA通过激活PPARγ/Nrf2-抗氧化途径调节NLRP3/caspase-1/IL-1β信号轴,从而抑制痤疮梭菌诱导的炎症,提出了一种新的治疗寻常痤疮的可能性。
    BACKGROUND: Our previous studies have demonstrated a strong relationship betweenCutibacterium acnes(C. acnes), oxidative stress, and acne inflammation. Syringic acid (SA) is a plant widely used for its antimicrobial, anti-inflammatory, and antioxidant activities, but lacking data on acne. This study aims to investigate the effect and mechanism of SA on acne inflammation induced by C. acnes in vitro and in vivo.
    METHODS: After using the SA to expose HaCaT keratinocytes, we reevaluated the effect of the SA on cell viability, cell apoptosis, ROS, CAT, SOD, and other inflammatory variables in the heat-killed C. acnes-treated HaCaT cells. Next, to induce mice with acne inflammation, ICR mice were given an intradermal injection of live C. acnes into their right ears. The effect of SA on this inflammation was then examined. Moreover, we explored the mechanism of SA on PPARγ/Nrf2 and NLRP3/caspase-1/IL-1β pathways by ELISA, immunofluorescence microscopy, and western blot assay.
    RESULTS: Heat-killed C. acnes triggered remarkable cell apoptosis, ROS production, interleukin (IL)-1β, IL-18, IL-6, and TNF-α release, reduced SOD and CAT activity, and upregulated the expression of proteins in HaCaT cells, including up-regulating IL-1β, PPARγ, Nrf2, HO-1, NQO1, NLRP3, and caspase-1, whereas SA inhibited these effects by partially impairing PPARγ activation. In addition, PPARγ silencing decreased C. acnes-induced IL-1β secretion and the production of intracellular ROS, down-regulating the expression of Nrf2. Nrf2 activator (SFN) enhanced anti-inflammatory activity through antioxidant mechanisms, boosting intracellular ROS production, reducing SOD and CAT activity, and promoting the increase in ROS, HO-1, NQO1, and IL-1β levels, while PPARγ inhibitor (GW662) effectively inhibited this effect in heat-killed C. acnes-treated cells. Finally, SA also exhibited notable improvements in ear redness, swelling, and the expression of PPARγ, NLRP3, and IL-1β in vivo.
    CONCLUSIONS: SA inhibited C. acnes-induced inflammation via regulating the NLRP3/caspase-1/IL-1β signaling axis by activating the PPARγ/Nrf2-antioxidant pathway, suggesting a new treatment possibility for acne vulgaris.
    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
    目的:非酒精性脂肪性肝病(NAFLD)是目前全球最常见的慢性肝病之一,以存在脂滴为特征。Rab18是一种重要的脂滴蛋白;然而,其对NAFLD的影响和作用机制尚不清楚.
    方法:使用游离脂肪酸刺激的AML-12细胞和高脂饮食(HFD)喂养的小鼠作为NAFLD模型。使用过表达Rab18(Rab18-OE)或敲低(Rab18-KD)的慢病毒来产生用于遗传分析的稳定细胞系。血清丙氨酸转氨酶水平,天冬氨酸转氨酶,总胆固醇,甘油三酯,高密度脂蛋白胆固醇,低密度脂蛋白胆固醇,葡萄糖,使用生化自动分析仪测量瘦素。进行苏木精和伊红染色以检测对肝脏的病理损伤。通过油红O染色评估细胞中的脂质积累。使用qPCR测量靶表达,西方印迹,和免疫细胞化学.
    结果:Rab18mRNA和蛋白质表达水平在游离脂肪酸刺激的AML-12细胞和HFD喂养小鼠的肝脏中增加。Rab18-OE在体外增加脂质积累,被Rab18-KD减毒。在体内,Rab18-OE增强肝脏病理损害,血清丙氨酸氨基转移酶/天冬氨酸氨基转移酶活性,和甘油三酯,总胆固醇,和低密度脂蛋白水平,而Rab18-KD降低了这些指标。Rab18-KD还下调HFD喂养小鼠的血糖水平。机械上,Rab18-OE和Rab18-KD在体外和体内调节perilipin2(PLIN2)和过氧化物酶体增殖物激活受体γ(PPARγ)的mRNA和蛋白表达水平,分别。免疫细胞化学显示Rab18在AML-12细胞中与PLIN2和PPARγ共定位。
    结论:在NAFLD小鼠模型中,Rab18的表达在体外和体内均升高。Rab18调节PLIN2和PPARγ的表达以增加NAFLD患者的肝损伤和脂质积累。因此,Rab18可能是该疾病中的关键蛋白和潜在的治疗靶标。
    OBJECTIVE: Nonalcoholic fatty liver disease (NAFLD) is currently one of the most common chronic liver diseases worldwide, characterized by the presence of lipid droplets. Rab18 is an important lipid droplet protein; however, its effects and mechanisms of action on NAFLD remain unclear.
    METHODS: Free fatty acid-stimulated AML-12 cells and high-fat diet (HFD)-fed mice were used as NAFLD models. Lentiviruses overexpressing Rab18 (Rab18-OE) or knockdown (Rab18-KD) were used to generate stable cell lines for genetic analysis. Blood serum levels of alanine aminotransferase, aspartate aminotransferase, total cholesterol, triglycerides, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, glucose, and leptin were measured using a biochemical autoanalyzer. Hematoxylin and eosin staining was performed to detect pathological damage to the liver. Lipid accumulation in the cells was assessed by Oil Red O staining. Target expression was measured using qPCR, western blotting, and immunocytochemistry.
    RESULTS: Rab18 mRNA and protein expression levels increased in free fatty acid-stimulated AML-12 cells and the livers of HFD-fed mice. Rab18-OE increased lipid accumulation in vitro, which was attenuated by Rab18-KD. In vivo, Rab18-OE augmented liver pathological damage, serum alanine aminotransferase/aspartate aminotransferase activity, and triglyceride, total cholesterol, and low-density lipoprotein levels, whereas Rab18-KD decreased these indicators. Rab18-KD also downregulated blood glucose levels in HFD-fed mice. Mechanistically, Rab18-OE and Rab18-KD regulated the mRNA and protein expression levels of perilipin 2 (PLIN2) and peroxisome proliferator-activated receptor gamma (PPARγ) in vitro and in vivo, respectively. Immunocytochemistry revealed that Rab18 colocalized with PLIN2 and PPARγ in AML-12 cells.
    CONCLUSIONS: Rab18 expression was elevated in vitro and in vivo in the NAFLD mouse model. Rab18 regulates PLIN2 and PPARγ expression to exaggerate liver injury and lipid accumulation in patients with NAFLD. Thus, Rab18 may be a crucial protein in this disease and a potential therapeutic target.
    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
    子宫内膜炎是危害人类和动物健康的常见疾病。花青素-3-O-葡萄糖苷(C3G),一种花青素,存在于多种植物中并表现出许多生物活性。这里,我们研究了C3G对LPS诱导的小鼠子宫内膜炎的作用和机制。结果表明,C3G显著降低了子宫的湿重/干重(W/D)比,改善子宫病理损伤,并抑制MPO活性。进一步的机制研究表明,NFκB通路的激活和TNF-α的水平,IL-1β,C3G治疗后IL-6明显抑制。相反,C3G促进LPS诱导的PPARγ/ABCA1途径的激活。有趣的是,PPARγ抑制剂GW9662显著削弱了C3G的抗炎作用。此外,还发现了C3G的抗氧化应激作用。第一次,我们的结果表明,C3G治疗可能是治疗子宫内膜炎的新策略.
    Endometritis is a common disease that endangers human and animal health. Cyanidin-3-O-glucoside (C3G), a kind of anthocyanin, exists in a variety of plants and shows many biological activities. Here, we investigated the effect and mechanism of C3G on LPS-induced endometritis in mice. The results showed that C3G significantly decreased wet to dry weight (W/D) ratio of uterine, improved uterine pathological injury, and inhibited MPO activity. Further mechanism investigation showed that the activation of NFκB pathway and the levels of TNF-a, IL-1β, and IL-6 were significantly suppressed after C3G treatment. Conversely, C3G promoted LPS-induced the activation of the PPARγ/ABCA1 pathway. Interestingly, the anti-inflammatory effect of C3G was significantly weakened by GW9662, a PPARγ inhibitor. In addition, the anti-oxidative stress effect of C3G was also found. For the first time, our results showed that treatment with C3G might be a new strategy for treating endometritis.
    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
    骨关节炎(OA),以慢性疼痛为特征,显著影响受影响个体的生活质量。OA发病的关键因素包括软骨降解和炎症。信号转导和转录激活因子3(STAT3),STAT蛋白家族的一员,在介导炎症中起关键作用。STX-0119已被证实为可以特异性抑制STAT3的小分子化合物。然而,STX-0119治疗OA的疗效尚待评估.因此,本研究旨在探讨STX-0119治疗OA的疗效及分子机制。我们发现磷酸化STAT3的表达在人OA软骨以及OA小鼠模型的软骨中上调。在体内,STX-0119联合注射到OA小鼠中减轻软骨退变而不影响软骨下骨。此外,STX-0119可抑制软骨中STAT3的磷酸化。体外,STX-0119在白细胞介素-1β诱导的软骨细胞炎症模型中抑制软骨细胞的炎症反应并促进合成代谢。此外,转录组测序和慢病毒感染测定的结果表明,在软骨细胞中,STX-0119通过抑制STAT3磷酸化诱导过氧化物酶体增殖物激活受体γ(PPARγ)表达上调。最后,在人软骨样品的离体培养中,重申STX-0119通过STAT3/PPARγ信号通路抑制软骨退变。一起,我们的研究结果支持STX-0119作为靶向STAT3治疗OA的治疗药物的开发潜力.
    Osteoarthritis (OA), characterized by chronic pain, significantly affects the quality of life of affected individuals. Key factors in OA pathogenesis include cartilage degradation and inflammation. Signal transducer and activator of transcription 3 (STAT3), a member of the STAT protein family, plays a pivotal role in mediating inflammation. STX-0119 has been verified as a small molecular compound that can specifically inhibit STAT3. However, the efficacy of STX-0119 in the treatment of OA remains to be evaluated. Therefore, the aim of this study was to explore the therapeutic effects and molecular mechanisms of STX-0119 in the treatment of OA. We found that the expression of phosphorylated STAT3 is upregulated in human OA cartilage as well as in the cartilage of a mouse model of OA. In vivo, joint injection of STX-0119 into OA mice alleviated cartilage degeneration without affecting the subchondral bone. Additionally, STX-0119 could inhibit the phosphorylation of STAT3 in the cartilage. In vitro, STX-0119 suppressed inflammatory responses in chondrocytes and promoted anabolic metabolism in an interleukin-1β-induced chondrocyte inflammation model. Additionally, the results of transcriptome sequencing and lentiviral infection assays demonstrated that in chondrocytes, STX-0119 induces the upregulation of peroxisome proliferators-activated receptor gamma (PPARγ) expression by inhibiting STAT3 phosphorylation. Finally, in ex vivo cultures of human cartilage samples, STX-0119 was reaffirmed to inhibit cartilage degeneration via the STAT3/PPARγ signaling pathway. Together, our findings support the potential of STX-0119 for development as a therapeutic agent targeting STAT3 for the treatment of OA.
    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