HSCs, hepatic stellate cells

HSC,肝星状细胞
  • 文章类型: Journal Article
    未经证实:酒精相关性肝病(ALD)的进展是由遗传易感性驱动的。脂蛋白脂肪酶(LPL)基因中的rs13702变体与非酒精性脂肪肝疾病有关。我们旨在阐明其在ALD中的作用。
    未经证实:酒精相关性肝硬化患者,有(n=385)和无肝细胞癌(HCC)(n=656),肝癌可归因于病毒性丙型肝炎(n=280),酒精滥用而无肝脏损害的对照(n=366),和健康对照(n=277)进行LPLrs13702多态性的基因分型。此外,对英国生物银行队列进行了分析.在人肝标本和肝细胞系中研究LPL表达。
    UNASSIGNED:与无HCC的ALD相比,有HCC的ALD中LPLrs13702CC基因型的频率较低(3.9%vs.9.3%)和验证队列(4.7%与9.5%;各p<0.05),与病毒性HCC患者(11.4%)相比,酒精滥用无肝硬化(8.7%),或健康对照(9.0%)。这种保护作用(比值比[OR]=0.5)在多变量分析中得到证实,包括年龄(OR=1.1/年),男性(OR=3.0),糖尿病(OR=1.8),并携带PNPLA3I148M风险变体(OR=2.0)。在英国生物银行队列中,LPLrs13702C等位基因被复制为HCC的危险因素.LPLmRNA的肝脏表达依赖于LPLrs13702基因型,与对照组和酒精相关的HCC相比,ALD肝硬化患者的肝脏表达明显更高。尽管肝细胞系显示可忽略的LPL蛋白表达,肝星状细胞和肝窦内皮细胞表达LPL。
    未经证实:酒精相关性肝硬化患者肝脏中LPL上调。LPLrs13702高生产者变体在ALD中赋予对HCC的保护,这可能有助于对人们进行HCC风险分层。
    UASSIGNED:肝细胞癌是受遗传易感性影响的肝硬化的严重并发症。我们发现,编码脂蛋白脂肪酶的基因中的遗传变异可降低酒精相关性肝硬化中患肝细胞癌的风险。这种遗传变异可能直接影响肝脏,因为,与健康的成人肝脏不同,脂蛋白脂肪酶是由酒精相关肝硬化的肝细胞产生的。
    UNASSIGNED: Progression of alcohol-associated liver disease (ALD) is driven by genetic predisposition. The rs13702 variant in the lipoprotein lipase (LPL) gene is linked to non-alcoholic fatty liver disease. We aimed at clarifying its role in ALD.
    UNASSIGNED: Patients with alcohol-associated cirrhosis, with (n = 385) and without hepatocellular carcinoma (HCC) (n = 656), with HCC attributable to viral hepatitis C (n = 280), controls with alcohol abuse without liver damage (n = 366), and healthy controls (n = 277) were genotyped regarding the LPL rs13702 polymorphism. Furthermore, the UK Biobank cohort was analysed. LPL expression was investigated in human liver specimens and in liver cell lines.
    UNASSIGNED: Frequency of the LPL rs13702 CC genotype was lower in ALD with HCC in comparison to ALD without HCC both in the initial (3.9% vs. 9.3%) and the validation cohort (4.7% vs. 9.5%; p <0.05 each) and compared with patients with viral HCC (11.4%), alcohol misuse without cirrhosis (8.7%), or healthy controls (9.0%). This protective effect (odds ratio [OR] = 0.5) was confirmed in multivariate analysis including age (OR = 1.1/year), male sex (OR = 3.0), diabetes (OR = 1.8), and carriage of the PNPLA3 I148M risk variant (OR = 2.0). In the UK Biobank cohort, the LPL rs13702 C allele was replicated as a risk factor for HCC. Liver expression of LPL mRNA was dependent on LPL rs13702 genotype and significantly higher in patients with ALD cirrhosis compared with controls and alcohol-associated HCC. Although hepatocyte cell lines showed negligible LPL protein expression, hepatic stellate cells and liver sinusoidal endothelial cells expressed LPL.
    UNASSIGNED: LPL is upregulated in the liver of patients with alcohol-associated cirrhosis. The LPL rs13702 high producer variant confers protection against HCC in ALD, which might help to stratify people for HCC risk.
    UNASSIGNED: Hepatocellular carcinoma is a severe complication of liver cirrhosis influenced by genetic predisposition. We found that a genetic variant in the gene encoding lipoprotein lipase reduces the risk for hepatocellular carcinoma in alcohol-associated cirrhosis. This genetic variation may directly affect the liver, because, unlike in healthy adult liver, lipoprotein lipase is produced from liver cells in alcohol-associated cirrhosis.
    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
    未经证实:氧化应激被认为是非酒精性脂肪性肝炎(NASH)进展的主要驱动因素。转录因子NRF2及其负调节因子KEAP1是氧化还原的主调节因子,代谢和蛋白质稳态,以及排毒,因此似乎是治疗NASH的有吸引力的靶标。
    UNASSIGNED:分子建模和X射线晶体学用于设计S217879-一种可以破坏KEAP1-NRF2相互作用的小分子。使用各种分子和细胞测定高度表征S217879。然后在两个不同的NASH相关临床前模型中进行评估,即蛋氨酸和胆碱缺乏饮食(MCDD)和饮食诱导的肥胖NASH(DIONASH)模型。
    UNASSIGNED:基于分子和细胞的检测证实S217879是一种高效和选择性的NRF2激活剂,具有明显的抗炎特性,如原代人外周血单核细胞所示。在MCDD小鼠中,S217879治疗2周导致NAFLD活性评分的剂量依赖性降低,同时显着增加肝脏Nqo1mRNA水平,一种特定的NRF2靶参与生物标志物。在DIONASH小鼠中,S217879治疗导致已建立的肝损伤的显着改善,NAS和肝纤维化均明显减少。αSMA和Col1A1染色,以及肝脏羟脯氨酸水平的定量,证实了响应S217879的肝纤维化的减少。RNA测序分析揭示了响应S217879的肝脏转录组中的主要变化,NRF2依赖性基因转录的激活和驱动疾病进展的关键信号通路的显著抑制。
    UNASSIGNED:这些结果突出了NRF2-KEAP1相互作用选择性破坏治疗NASH和肝纤维化的潜力。
    UNASSIGNED:我们报告了S217879的发现——一种具有良好药代动力学特性的有效和选择性的NRF2激活剂。通过破坏KEAP1-NRF2相互作用,S217879触发抗氧化反应的上调和涉及NASH疾病进展的广谱基因的协调调节,最终导致小鼠NASH和肝纤维化进展的减少。
    UNASSIGNED: Oxidative stress is recognized as a major driver of non-alcoholic steatohepatitis (NASH) progression. The transcription factor NRF2 and its negative regulator KEAP1 are master regulators of redox, metabolic and protein homeostasis, as well as detoxification, and thus appear to be attractive targets for the treatment of NASH.
    UNASSIGNED: Molecular modeling and X-ray crystallography were used to design S217879 - a small molecule that could disrupt the KEAP1-NRF2 interaction. S217879 was highly characterized using various molecular and cellular assays. It was then evaluated in two different NASH-relevant preclinical models, namely the methionine and choline-deficient diet (MCDD) and diet-induced obesity NASH (DIO NASH) models.
    UNASSIGNED: Molecular and cell-based assays confirmed that S217879 is a highly potent and selective NRF2 activator with marked anti-inflammatory properties, as shown in primary human peripheral blood mononuclear cells. In MCDD mice, S217879 treatment for 2 weeks led to a dose-dependent reduction in NAFLD activity score while significantly increasing liver Nqo1 mRNA levels, a specific NRF2 target engagement biomarker. In DIO NASH mice, S217879 treatment resulted in a significant improvement of established liver injury, with a clear reduction in both NAS and liver fibrosis. αSMA and Col1A1 staining, as well as quantification of liver hydroxyproline levels, confirmed the reduction in liver fibrosis in response to S217879. RNA-sequencing analyses revealed major alterations in the liver transcriptome in response to S217879, with activation of NRF2-dependent gene transcription and marked inhibition of key signaling pathways that drive disease progression.
    UNASSIGNED: These results highlight the potential of selective disruption of the NRF2-KEAP1 interaction for the treatment of NASH and liver fibrosis.
    UNASSIGNED: We report the discovery of S217879 - a potent and selective NRF2 activator with good pharmacokinetic properties. By disrupting the KEAP1-NRF2 interaction, S217879 triggers the upregulation of the antioxidant response and the coordinated regulation of a wide spectrum of genes involved in NASH disease progression, leading ultimately to the reduction of both NASH and liver fibrosis progression in mice.
    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
    未经证实:非酒精性脂肪性肝病(NAFLD)的患病率及其严重形式,非酒精性脂肪性肝炎(NASH),正在增加。患有NASH的个体通常发展为肝纤维化,并且晚期肝纤维化是患有NASH的个体的死亡率的主要决定因素。我们和其他人报道STAT3有助于小鼠的肝纤维化和肝细胞癌。
    未经评估:这里,我们探讨了肝细胞和非肝细胞区域的STAT3激活,通过磷酸-STAT3(pSTAT3)测量,与133例NAFLD患者的肝纤维化进展相关。通过整合纤维化NAFLD肝脏中的空间分布和转录组变化,我们进一步表征了STAT3激活的分子和细胞决定因素。结果:非肝细胞区域的pSTAT3评分随着纤维化严重程度而逐渐增加(r=0.53,p<0.001)。pSTAT3评分与1,540个免疫和癌症相关基因的表达之间的相关性分析揭示了STAT3激活对非肝细胞区域基因表达变化的巨大影响,并证实了STAT3激活在纤维形成中的主要作用。数字空间转录组分析也在肝细胞和非肝细胞区域选择从四个NAFLD肝活检与晚期纤维化的13个区域进行,使用定制的标记物组,包括pSTAT3、PanCK+CK8/18和CD45。基于阳性或阴性pSTAT3染色进一步分割区域。细胞去卷积分析显示活化的STAT3富集在肝祖细胞(HPCs)和窦内皮细胞中。在NASH小鼠中STAT3抑制后肝纤维化的回归导致HPCs减少,证明STAT3在HPC扩展中的直接作用。
    UNASSIGNED:增加对NASH和肝纤维化进展中STAT3信号传导的空间依赖性的理解可能导致新的靶向治疗方法。
    未经证实:晚期肝纤维化是NASH患者死亡率的主要决定因素。这项研究表明,使用来自133名NAFLD患者的肝活检,非肝细胞区域的STAT3激活与纤维化严重程度密切相关,炎症,并发展到NASH。STAT3激活富集在肝祖细胞(HPCs)和肝窦内皮细胞(SECs),由研究pSTAT3空间分布的创新技术决定。最后,小鼠中的STAT3抑制导致肝纤维化减少和HPCs的消耗,表明HPCs中的STAT3激活有助于其在NAFLD中的扩张和纤维化形成。
    UNASSIGNED: The prevalence of non-alcoholic fatty liver disease (NAFLD) and its severe form, non-alcoholic steatohepatitis (NASH), is increasing. Individuals with NASH often develop liver fibrosis and advanced liver fibrosis is the main determinant of mortality in individuals with NASH. We and others have reported that STAT3 contributes to liver fibrosis and hepatocellular carcinoma in mice.
    UNASSIGNED: Here, we explored whether STAT3 activation in hepatocyte and non-hepatocyte areas, measured by phospho-STAT3 (pSTAT3), is associated with liver fibrosis progression in 133 patients with NAFLD. We further characterized the molecular and cellular determinants of STAT3 activation by integrating spatial distribution and transcriptomic changes in fibrotic NAFLD livers.Results: pSTAT3 scores in non-hepatocyte areas progressively increased with fibrosis severity (r = 0.53, p <0.001). Correlation analyses between pSTAT3 scores and expression of 1,540 immune- and cancer-associated genes revealed a large effect of STAT3 activation on gene expression changes in non-hepatocyte areas and confirmed a major role for STAT3 activation in fibrogenesis. Digital spatial transcriptomic profiling was also performed on 13 regions selected in hepatocyte and non-hepatocyte areas from four NAFLD liver biopsies with advanced fibrosis, using a customized panel of markers including pSTAT3, PanCK+CK8/18, and CD45. The regions were further segmented based on positive or negative pSTAT3 staining. Cell deconvolution analysis revealed that activated STAT3 was enriched in hepatic progenitor cells (HPCs) and sinusoidal endothelial cells. Regression of liver fibrosis upon STAT3 inhibition in mice with NASH resulted in a reduction of HPCs, demonstrating a direct role for STAT3 in HPC expansion.
    UNASSIGNED: Increased understanding of the spatial dependence of STAT3 signaling in NASH and liver fibrosis progression could lead to novel targeted treatment approaches.
    UNASSIGNED: Advanced liver fibrosis is the main determinant of mortality in patients with NASH. This study showed using liver biopsies from 133 patients with NAFLD, that STAT3 activation in non-hepatocyte areas is strongly associated with fibrosis severity, inflammation, and progression to NASH. STAT3 activation was enriched in hepatic progenitor cells (HPCs) and sinusoidal endothelial cells (SECs), as determined by innovative technologies interrogating the spatial distribution of pSTAT3. Finally, STAT3 inhibition in mice resulted in reduced liver fibrosis and depletion of HPCs, suggesting that STAT3 activation in HPCs contributes to their expansion and fibrogenesis in NAFLD.
    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
    非酒精性脂肪性肝病(NAFLD)正在达到流行病的比例,全球成人患病率为25%。非酒精性脂肪性肝炎(NASH),会导致肝硬化,已经成为欧洲和美国肝移植的主要适应症。肝纤维化是持续的结果,迭代肝损伤,以及NASH结局的主要决定因素。肝脏具有显著的内在可塑性,肝纤维化可以在去除有害物质后消退,从而提供了通过治疗干预改变长期结局的机会.尽管肝细胞损伤是NASH的关键驱动因素,肝纤维化小生境内的多个其他细胞系在炎症的延续中起主要作用,间充质细胞活化,细胞外基质积累以及纤维化分辨率。这种细胞相互作用的成分,以及纤维化小生境中的各种亚群如何相互作用以驱动纤维发生是一个活跃的研究领域。纤维化小生境的重要细胞成分包括内皮细胞,巨噬细胞,传代免疫细胞群和肌成纤维细胞。在这次审查中,我们将描述单细胞基因组学等技术的快速发展,空间转录组学和单细胞配体受体分析正在改变我们对NAFLD/NASH细胞相互作用组的理解,以及这个新的,利用高分辨率信息为NASH患者开发合理的新疗法.
    Non-alcoholic fatty liver disease (NAFLD) is reaching epidemic proportions, with a global prevalence of 25% in the adult population. Non-alcoholic steatohepatitis (NASH), which can lead to cirrhosis, has become the leading indication for liver transplantation in both Europe and the USA. Liver fibrosis is the consequence of sustained, iterative liver injury, and the main determinant of outcomes in NASH. The liver possesses remarkable inherent plasticity, and liver fibrosis can regress when the injurious agent is removed, thus providing opportunities to alter long-term outcomes through therapeutic interventions. Although hepatocyte injury is a key driver of NASH, multiple other cell lineages within the hepatic fibrotic niche play major roles in the perpetuation of inflammation, mesenchymal cell activation, extracellular matrix accumulation as well as fibrosis resolution. The constituents of this cellular interactome, and how the various subpopulations within the fibrotic niche interact to drive fibrogenesis is an area of active research. Important cellular components of the fibrotic niche include endothelial cells, macrophages, passaging immune cell populations and myofibroblasts. In this review, we will describe how rapidly evolving technologies such as single-cell genomics, spatial transcriptomics and single-cell ligand-receptor analyses are transforming our understanding of the cellular interactome in NAFLD/NASH, and how this new, high-resolution information is being leveraged to develop rational new therapies for patients with NASH.
    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
    高剂量照射是帮助控制肝肿瘤生长的重要工具,但它可以导致辐射诱发的肝脏疾病(RILD)。这种危及生命的并发症在放射治疗后数月表现出来,其特征是中心周围窦纤维化。在这项研究中,我们旨在建立RILD小鼠模型,以探讨放射性肝纤维化的潜在机制。
    使用小动物图像引导放射治疗平台,设计了一种将50Gy作为单剂量输送到小鼠肝脏焦点的照射方案。对组织进行了1天和6天的分析,照射后6周和20周。通过组织学评估辐照的肝脏,免疫组织化学,成像质量细胞计数和RNA测序。使用高分辨率呼吸测量法评估线粒体功能。
    在照射后6周和20周,在高度照射的区域可见中心周围纤维化,同时存在免疫细胞浸润和红细胞外渗。RNA测序分析显示与急性DNA损伤相关的基因特征,p53激活,衰老及其相关的分泌表型和纤维化。此外,检测到线粒体损伤的基因谱和线粒体DNA异质性的增加。体外肝细胞的呼吸测定证实了辐射诱导的线粒体功能障碍。最后,高度辐照的纤维化区域显示出活性氧的标志物,例如谷胱甘肽减少,脂质过氧化物增加和衰老样表型。
    基于我们的RILD小鼠模型,我们认为,辐射诱导的线粒体DNA不稳定性有助于纤维化的发展通过产生过量的活性氧,p53途径激活和衰老样表型。
    照射是一种有效的癌症疗法,然而,其对肝脏的适用性受到威胁生命的辐射诱导的肝纤维化的限制。我们开发了一种新的小鼠辐射诱导肝纤维化模型,概括了人类疾病。我们的模型强调了线粒体DNA不稳定性在辐射诱导的肝纤维化发展中的作用。这个新模型和随后的发现将有助于增加我们对肝脏对辐射的反应的理解,并找到保护肝脏的策略,使放射治疗的扩大使用,以治疗肝肿瘤。
    UNASSIGNED: High-dose irradiation is an essential tool to help control the growth of hepatic tumors, but it can cause radiation-induced liver disease (RILD). This life-threatening complication manifests itself months following radiation therapy and is characterized by fibrosis of the pericentral sinusoids. In this study, we aimed to establish a mouse model of RILD to investigate the underlying mechanism of radiation-induced liver fibrosis.
    UNASSIGNED: Using a small animal image-guided radiation therapy platform, an irradiation scheme delivering 50 Gy as a single dose to a focal point in mouse livers was designed. Tissues were analyzed 1 and 6 days, and 6 and 20 weeks post-irradiation. Irradiated livers were assessed by histology, immunohistochemistry, imaging mass cytometry and RNA sequencing. Mitochondrial function was assessed using high-resolution respirometry.
    UNASSIGNED: At 6 and 20 weeks post-irradiation, pericentral fibrosis was visible in highly irradiated areas together with immune cell infiltration and extravasation of red blood cells. RNA sequencing analysis showed gene signatures associated with acute DNA damage, p53 activation, senescence and its associated secretory phenotype and fibrosis. Moreover, gene profiles of mitochondrial damage and an increase in mitochondrial DNA heteroplasmy were detected. Respirometry measurements of hepatocytes in vitro confirmed irradiation-induced mitochondrial dysfunction. Finally, the highly irradiated fibrotic areas showed markers of reactive oxygen species such as decreased glutathione and increased lipid peroxides and a senescence-like phenotype.
    UNASSIGNED: Based on our mouse model of RILD, we propose that irradiation-induced mitochondrial DNA instability contributes to the development of fibrosis via the generation of excessive reactive oxygen species, p53 pathway activation and a senescence-like phenotype.
    UNASSIGNED: Irradiation is an efficient cancer therapy, however, its applicability to the liver is limited by life-threatening radiation-induced hepatic fibrosis. We have developed a new mouse model of radiation-induced liver fibrosis, that recapitulates the human disease. Our model highlights the role of mitochondrial DNA instability in the development of irradiation-induced liver fibrosis. This new model and subsequent findings will help increase our understanding of the hepatic reaction to irradiation and to find strategies that protect the liver, enabling the expanded use of radiotherapy to treat hepatic tumors.
    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
    Ferroptosis是受调节的细胞死亡的一种形式,以铁和ROS依赖性方式过度的膜脂质过氧化为特征。雷公藤多,一种从雷公藤中提取的天然生物活性三萜,在多种肝病中显示出有效的抗纤维化和抗炎活性。然而,雷公藤红素在肝纤维化治疗中的确切分子作用机制和直接蛋白靶点仍然难以捉摸。这里,我们发现雷公藤红素通过促进活性氧(ROS)的产生和诱导活化肝星状细胞(HSC)的铁凋亡而发挥抗纤维化作用。通过使用基于活性的蛋白质谱分析(ABPP)结合生物正交点击化学反应和细胞热转移测定(CETSA),我们显示雷公藤多酚类直接与过氧化物酶(PRDXs)结合,包括PRDX1,PRDX2,PRDX4和PRDX6,通过活性半胱氨酸位点,并抑制其抗氧化活性。雷公藤红素还靶向血红素加氧酶1(HO-1),并上调其在活化HSC中的表达。HSC中PRDX1、PRDX2、PRDX4、PRDX6或HO-1的敲除,在不同程度上,细胞ROS水平升高并诱导铁凋亡。一起来看,我们的发现揭示了雷公藤红素改善肝纤维化的直接蛋白靶点和分子机制,从而支持雷公藤红素作为肝纤维化有前途的治疗剂的进一步发展。
    Ferroptosis is a form of regulated cell death, characterized by excessive membrane lipid peroxidation in an iron- and ROS-dependent manner. Celastrol, a natural bioactive triterpenoid extracted from Tripterygium wilfordii, shows effective anti-fibrotic and anti-inflammatory activities in multiple hepatic diseases. However, the exact molecular mechanisms of action and the direct protein targets of celastrol in the treatment of liver fibrosis remain largely elusive. Here, we discover that celastrol exerts anti-fibrotic effects via promoting the production of reactive oxygen species (ROS) and inducing ferroptosis in activated hepatic stellate cells (HSCs). By using activity-based protein profiling (ABPP) in combination with bio-orthogonal click chemistry reaction and cellular thermal shift assay (CETSA), we show that celastrol directly binds to peroxiredoxins (PRDXs), including PRDX1, PRDX2, PRDX4 and PRDX6, through the active cysteine sites, and inhibits their anti-oxidant activities. Celastrol also targets to heme oxygenase 1 (HO-1) and upregulates its expression in activated-HSCs. Knockdown of PRDX1, PRDX2, PRDX4, PRDX6 or HO-1 in HSCs, to varying extent, elevated cellular ROS levels and induced ferroptosis. Taken together, our findings reveal the direct protein targets and molecular mechanisms via which celastrol ameliorates hepatic fibrosis, thus supporting the further development of celastrol as a promising therapeutic agent for liver fibrosis.
    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
    脂质是一类复杂多样的分子,在许多生理过程中起着至关重要的作用。以及在发病中,programming,和癌症的维持。脂肪酸和胆固醇是脂质的组成部分,协调这些关键的代谢过程。在肝脏中,脂质改变是普遍的原因和慢性乙型肝炎和丙型肝炎病毒感染的后果,酒精性肝炎,非酒精性脂肪性肝病和脂肪性肝炎。脂质组学的最新发展也揭示了三酰甘油的动态变化,磷脂,鞘脂,神经酰胺,脂肪酸,和胆固醇参与原发性肝癌的发展和进展。因此,脂质代谢的转录景观表明增加脂肪酸和固醇合成的致癌作用。然而,迄今为止,对肝脂质组复杂性的机制见解有限,阻碍了有效疗法的发展。
    Lipids are a complex and diverse group of molecules with crucial roles in many physiological processes, as well as in the onset, progression, and maintenance of cancers. Fatty acids and cholesterol are the building blocks of lipids, orchestrating these crucial metabolic processes. In the liver, lipid alterations are prevalent as a cause and consequence of chronic hepatitis B and C virus infections, alcoholic hepatitis, and non-alcoholic fatty liver disease and steatohepatitis. Recent developments in lipidomics have also revealed that dynamic changes in triacylglycerols, phospholipids, sphingolipids, ceramides, fatty acids, and cholesterol are involved in the development and progression of primary liver cancer. Accordingly, the transcriptional landscape of lipid metabolism suggests a carcinogenic role of increasing fatty acids and sterol synthesis. However, limited mechanistic insights into the complex nature of the hepatic lipidome have so far hindered the development of effective therapies.
    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
    肝细胞癌(HCC)是一种侵袭性人类癌症,在全球范围内发病率不断上升。已经做出了许多努力来探索治疗HCC的药物疗法。如靶向酪氨酸激酶抑制剂,基于免疫的疗法和联合化疗。然而,目前的策略存在局限性,包括例如化学抗性。肿瘤的启动和进展是由代谢的重新编程驱动的,特别是在HCC发展过程中。最近,代谢相关脂肪性肝病(MAFLD),非酒精性脂肪性肝病(NAFLD)新命名法的重新评估,表明对肝脏疾病发病机制中代谢的认识日益提高,包括HCC,从而提出了针对异常代谢的肝癌治疗新策略。在这次审查中,我们通过突出葡萄糖的代谢目标来介绍方向,脂肪酸,氨基酸和谷氨酰胺代谢,适用于HCC药物干预。我们还总结和讨论了目前针对HCC治疗过程中代谢失调的药物和研究。此外,讨论了肝癌靶向代谢治疗的发现和发展的机遇和挑战。
    Hepatocellular carcinoma (HCC) is an aggressive human cancer with increasing incidence worldwide. Multiple efforts have been made to explore pharmaceutical therapies to treat HCC, such as targeted tyrosine kinase inhibitors, immune based therapies and combination of chemotherapy. However, limitations exist in current strategies including chemoresistance for instance. Tumor initiation and progression is driven by reprogramming of metabolism, in particular during HCC development. Recently, metabolic associated fatty liver disease (MAFLD), a reappraisal of new nomenclature for non-alcoholic fatty liver disease (NAFLD), indicates growing appreciation of metabolism in the pathogenesis of liver disease, including HCC, thereby suggesting new strategies by targeting abnormal metabolism for HCC treatment. In this review, we introduce directions by highlighting the metabolic targets in glucose, fatty acid, amino acid and glutamine metabolism, which are suitable for HCC pharmaceutical intervention. We also summarize and discuss current pharmaceutical agents and studies targeting deregulated metabolism during HCC treatment. Furthermore, opportunities and challenges in the discovery and development of HCC therapy targeting metabolism are discussed.
    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
    转化生长因子-β(TGF-β)是肝脏中的有效效应,它参与了肝损伤后启动的过多过程。TGF-β影响实质,非实质,和炎症细胞高度依赖环境的方式。其生物利用度对于对各种侮辱的快速反应是至关重要的。在肝脏中-可能在其他器官中-通过将大部分TGF-β作为称为潜伏TGF-β(L-TGF-β)的灭活前体形式在细胞外基质中沉积而成为可能。几种基质蛋白参与基质沉积,潜在的复杂稳定,和L-TGF-β的激活。细胞外基质蛋白1(ECM1)最近被认为是维持健康肝脏中沉积的L-TGF-β潜伏期的关键因素。的确,它的耗竭导致自发的TGF-β信号激活,对肝脏结构和功能产生有害影响.这篇综述文章介绍了细胞内L-TGF-β复合物形成的最新知识,分泌,基质沉积,和激活,并描述了所涉及的蛋白质和过程。Further,我们强调了下调肝纤维化和肝癌中L-TGF-β活化的治疗潜力。
    Transforming growth factor-β (TGF-β) is a potent effector in the liver, which is involved in a plethora of processes initiated upon liver injury. TGF-β affects parenchymal, non-parenchymal, and inflammatory cells in a highly context-dependent manner. Its bioavailability is critical for a fast response to various insults. In the liver - and probably in other organs - this is made possible by the deposition of a large portion of TGF-β in the extracellular matrix as an inactivated precursor form termed latent TGF-β (L-TGF-β). Several matrisomal proteins participate in matrix deposition, latent complex stabilisation, and activation of L-TGF-β. Extracellular matrix protein 1 (ECM1) was recently identified as a critical factor in maintaining the latency of deposited L-TGF-β in the healthy liver. Indeed, its depletion causes spontaneous TGF-β signalling activation with deleterious effects on liver architecture and function. This review article presents the current knowledge on intracellular L-TGF-β complex formation, secretion, matrix deposition, and activation and describes the proteins and processes involved. Further, we emphasise the therapeutic potential of toning down L-TGF-β activation in liver fibrosis and liver cancer.
    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
    本研究调查了二甲双胍治疗对高脂饮食(HFD)喂养大鼠肝脏氧化应激和非酒精性脂肪性肝病(NADLD)相关炎症的影响。
    用HFD或实验室食物饮食喂养Wistar大鼠8周。二甲双胍以200mg/kg的剂量口服给药。体重,每天记录食物和水的摄入量。口服葡萄糖耐量试验(OGTT),对血浆和组织样本进行生化分析和组织学检查.使用逆转录聚合物链反应(RT-PCR)分析抗氧化剂和抗炎mRNA表达。
    二甲双胍治疗8周可防止HFD诱导的体重增加并减少HFD喂养大鼠的脂肪沉积。生化分析表明,二甲双胍治疗显着减弱硝基氧化应激标志物丙二醛(MDA),高级蛋白质氧化产物(APOP),HFD喂养大鼠肝脏中一氧化氮(NO)水平过高。基因表达分析表明,二甲双胍治疗与HFD喂养大鼠肝脏中Nrf-2,HO-1,SOD和过氧化氢酶等抗氧化基因表达增强有关。二甲双胍治疗还发现调节脂肪代谢和抗炎基因的表达,包括PPAR-γ,C/EBP-α,SREBP1c,FAS,AMPK和GLUT-4。与生化和基因表达数据一致,组织病理学检查显示,二甲双胍治疗减轻了炎症细胞浸润,脂肪变性,肝细胞坏死,胶原蛋白沉积,HFD喂养大鼠肝脏纤维化。
    总而言之,这项研究表明,二甲双胍可能通过降低肝脏氧化应激和肝脏炎症来预防和治疗HFD诱导的脂肪变性.
    UNASSIGNED: This current study investigated the effect of metformin treatment on hepatic oxidative stress and inflammation associated with nonalcoholic fatty liver disease (NADLD) in high fat diet (HFD) fed rats.
    UNASSIGNED: Wistar rats were fed with a HFD or laboratory chow diet for 8 weeks. Metformin was administered orally at a dose of 200 mg/kg. Body weight, food and water intake were recorded on daily basis. Oral glucose tolerance test (OGTT), biochemical analysis and histological examinations were conducted on plasma and tissue samples. Antioxidant and anti-inflammatory mRNA expression was analyzed using reverse transcription polymeric chain reaction (RT-PCR).
    UNASSIGNED: Metformin treatment for 8 weeks prevented HFD-induced weight gain and decreased fat deposition in HFD fed rats. Biochemical analysis revealed that metformin treatment significantly attenuated nitro-oxidative stress markers malondialdehyde (MDA), advanced protein oxidation product (APOP), and excessive nitric oxide (NO) levels in the liver of HFD fed rats. Gene expression analysis demonestrated that metformin treatment was associated with an enhanced expression of antioxidant genes such as Nrf-2, HO-1, SOD and catalase in liver of HFD fed rats. Metformin treatment also found to modulate the expression of fat metabolizing and anti-inflammatory genes including PPAR--γ, C/EBP-α, SREBP1c, FAS, AMPK and GLUT-4. Consistent with the biochemical and gene expression data, the histopathological examination unveiled that metformin treatment attenuated inflammatory cells infiltration, steatosis, hepatocyte necrosis, collagen deposition, and fibrosis in the liver of HFD fed rats.
    UNASSIGNED: In conclusion, this study suggests that metformin might be effective in the prevention and treatment of HFD-induced steatosis by reducing hepatic oxidative stress and inflammation in the liver.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号