首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 390 毫秒
1.
肝纤维化是肝脏损伤后的病理性修复反应,其特征是肝脏中肝星状细胞(HSC)激活、扩增和细胞外基质蛋白过度沉积。CCN蛋白因子是具信号传递功能的细胞基质蛋白,参与组织修复反应并涉及肝纤维化发生发展。本文制备了高纯度的大鼠原代HSC并通过体外培养进行激活,同步定量分析了6个CCN因子在激活过程中的表达,以期对CCN因子在HSC激活过程中的作用进行更全面的评价。  相似文献   

2.
CTNNB1编码的β-连环蛋白(β-catenin)是细胞膜上钙黏蛋白复合物的主要组成成分,参与Wnt介导的细胞内信号传递。除了Wnt通路,β-catenin还参与其他信号通路。β-catenin与转录因子如T细胞因子(T-cell factor,TCF)4、叉头框转录因子O亚族(Fork head box protein O,FOXO)及缺氧诱导因子(Hypoxia inducible factor1α,HIF1α)结合调控靶基因的转录表达。β-catenin信号的改变可激活肝星状细胞(Hepatic stellate cell,HSC),而HSC是肝纤维化形成过程中的主要效应细胞。因此,对β-catenin的调控有望成为抗肝纤维化的治疗靶点。本文就有关β-catenin信号及其在肝纤维化中的作用予以简要综述。  相似文献   

3.
肺纤维化中TGF-β信号传导通路及其靶点治疗   总被引:1,自引:0,他引:1  
转化生长因子β(TGF-β)是最重要的致肺纤维化细胞因子.TGF-β通过下游的信号转导因子激活成纤维细胞等效应细胞.对TGF信号传导通路各靶点进行干预可能为治疗纤维化疾病提供一种新手段.  相似文献   

4.
肝纤维化是由各种病因所导致的肝脏病理性反应,是发展成肝硬化甚至肝癌的必经途径。以往研究发现,肝纤维化甚至是肝硬化早期都可以通过一定的干预治疗抑制与逆转病情,该过程有多种肝实质以及非实质细胞参与,肝星状细胞(hepatic stellate cell, HSC)与肝巨噬细胞是肝纤维化进程中关键的细胞类型。HSCs是肝纤维化的核心细胞,而肝巨噬细胞是肝纤维化进程中的主要调控细胞,HSCs与巨噬细胞间可通过分泌趋化因子、炎症因子以及凋亡因子诱导双方细胞的活化、分化、增殖和凋亡,并且能够调节细胞外基质(ECM)的生成与降解,进而影响肝纤维化的发生发展与抑制逆转。该文立足于HSCs与肝巨噬细胞的各自特征性功能,通过对它们之间的相互影响的阐述,探究两者在促进与逆转肝纤维化中的作用,以期探究肝纤维化复杂病理过程中的机制,为治疗逆转肝纤维化提供新的思路和有效靶点。  相似文献   

5.
人肝癌细胞与成纤维细胞共育时几种生长因子的表达   总被引:1,自引:0,他引:1  
本文采用定位细胞培养技术.将人肝癌细胞和成纤维细胞有序地排列在同一容器内。采用过氧化物酶标记链霉卵白素染色法(LSAB法).观察人肝癌细胞和成纤维细胞表达碱性成纤维细胞生长因子(bFGF)、表皮细胞生长因子(EGF)和转化生长因子β1(TGFβ1).研究肝癌细胞和成纤维细胞之间的相互调控作用。结果显示,bFGF、EGF和TGFβ1在两类细胞中的表达量随着细胞增殖率的变化也发生着动态变化。在细胞增殖率增高的时相,正性生长因子bFGF、EGF在细胞中的表达量也增高.而负性生长因子TGFβ1的表达量则降低;在细胞增殖率降低的时机.其表达量正好相反。表明肝癌细胞和成纤维细胞共育培养时,自分泌、旁分泌生长因子对细胞的增殖起着近程调控作用。  相似文献   

6.
肝纤维化发生发展的中心环节是肝星状细胞的激活和增生。活化的肝星状细胞(HSC)引起肝纤维化细胞外基质(ECM)组分改变的同时又被ECM所调控。活化HSC中过量表达的微小RNA29家族能减少ECM胶原I等基质蛋白分泌。先前未报道的ECM蛋白CYR61能使活化的HSC衰老,wnt-5a可能作为HSC增殖调节器在肝纤维化中对抗细胞凋亡。因此探索肝纤维化ECM和HSC之间的调控方式及作用机制能为早期干预甚至逆转肝纤维化提供有力靶点。  相似文献   

7.
CD8+T淋巴细胞与高血压心肌纤维化的研究进展   总被引:2,自引:0,他引:2  
血管生长因子增多,血管平滑肌细胞增殖和炎症在血管重塑方面起到了关键的作用。这种低级的炎症反应导致粘附分子表达,白细胞的侵入,细胞因子的产生,氧化应激的增加,从而激活免疫细胞和血管炎症信号通路,使T淋巴细胞及巨噬细胞等细胞活化,产生和释放多种活性因子,激活心肌的细胞外基质生成细胞,引起胶原形成及代谢异常,并可导致心肌实质细胞的变性、坏死或亚细胞结构变化等,从而引起心肌纤维化一系列病理生理变化。本文主要就CD8+T淋巴细胞在高血压心肌纤维化炎症反应中的细胞毒性作用、诱导细胞凋亡作用、分泌大量的炎症因子、增加MMPs的活性从而影响心肌纤维化的形成等方面做一综述!  相似文献   

8.
肺纤维化(pulmonary fibrosis,PF)是许多肺损伤的共同过程,病理表现为肌成纤维细胞的大量集聚,细胞外基质的沉积。近年来研究表明,转化生长因子β1(transforming growth factor-β1,TGF-β1)/Smad通路在肺纤维化中有重要作用。核转录共抑制因子SnoN(Ski-related novel protein N)能通过Smads蛋白抑制TGF-β1信号通路从而调节肺纤维化的发生发展。本文就SnoN在肺纤维化TGF-β1/Smad通路中的作用作一综述,为治疗肺纤维化找到新方向。  相似文献   

9.
目的:研究肝星状细胞(HSC)中smad2特异性小干扰RNA(siRNA)对Ⅰ型胶原表达的抑制作用,探讨抗肝纤维化的基因治疗新方法。方法:设计合成靶向Smad2基因的siRNA,将筛选成功的siRNA瞬时转染入体外培养的肝星状细胞(HSC),并给予转化生长因子β(TGF-β)刺激,应用RT-PCR和Western blot技术检测对照组与实验组Ⅰ型胶原mRNA水平和蛋白水平表达差异,研究siRNA对Ⅰ型胶原表达的抑制作用。结果:siRNA能明显降低肝星状细胞中Smad2的RNA和蛋白的表达水平,证实筛选的siRNA有效,能特异性抑制Smad2的基因表达;TGF-β刺激肝星状细胞后,与对照组比较,siRNA转染组细胞外基质(ECM)成分Ⅰ型胶原的表达水平明显降低(P<0.05)。结论:siRNA能够抑制TGFβ对肝星状细胞的激活,阻断TGFβ-Smads传导通路,使Ⅰ型胶原分泌下调,有效抑制TGFβ诱导的肝纤维化。  相似文献   

10.
肝纤维化是慢性肝病向肝硬化发展的必经之路,是细胞外基(Extracellular matrix,ECM)在肝内过多沉积所致。ECM主要由激活的肝星状细胞(Hepatic stellate cell,HSC)合成,同时它的降解受到基质金属蛋白酶组织抑制剂-1(Tissue inhibitor of metal protease-1,TIMP-1)的调控,因此HSC及TIMP-1对肝纤维化形成至关重要。近年来针对HSC及TIMP-1抗肝纤维化的研究已成为热点,就HSC及TIMP-1的生物学特性及其对肝纤维化的作用作一综述。  相似文献   

11.
Liver fibrosis is a wound-healing response to chronic injuries, characterized by the excessive accumulation of extracellular matrix or scar tissue within the liver; in addition, its formation is associated with multiple cytokines as well as several cell types and a variety of signaling pathways. When liver fibrosis is not well controlled, it can progress to liver cirrhosis, but it is reversible in principle. Thus far, no efficient therapy is available for treatment of liver fibrosis. Although liver transplantation is the preferred strategy, there are many challenges remaining in this approach, such as shortage of donor organs, immunological rejection, and surgical complications. Hence, there is a great need for an alternative therapeutic strategy. Currently, mesenchymal stem cell (MSC) therapy is considered a promising therapeutic strategy for the treatment of liver fibrosis; advantageously, the characteristics of MSCs are continuous self-renewal, proliferation, multipotent differentiation, and immunomodulatory activities. The human umbilical cord-derived (hUC)-MSCs possess not only the common attributes of MSCs but also more stable biological characteristics, relatively easy accessibility, abundant source, and no ethical issues (e.g., bone marrow being the adult source), making hUC-MSCs a good choice for treatment of liver fibrosis. In this review, we summarize the biological characteristics of hUC-MSCs and their paracrine effects, exerted by secretion of various cytokines, which ultimately promote liver repair through several signaling pathways. Additionally, we discuss the capacity of hUC-MSCs to differentiate into hepatocyte-like cells for compensating the function of existing hepatocytes, which may aid in amelioration of liver fibrosis. Finally, we discuss the current status of the research field and its future prospects.  相似文献   

12.
Liver fibrosis is a disease caused by long‐term damage that is related to a number of factors. The current research on the treatment of liver fibrosis mainly focuses on the activation of hepatic stellate cell, in addition to protecting liver cells. byakangelicin has certain anti‐inflammatory ability, but its effect on liver fibrosis is unclear. This study aims to explore whether byakangelicin plays a role in the development of liver fibrosis and to explore its mechanism. We determined that byakangelicin has a certain ability to resist fibrosis and reduce liver cell damage in a model of carbon tetrachloride–induced liver fibrosis in mice. Thereafter, we performed further verification in vitro. The signalling pathways of two important pro‐fibrotic cytokines, transforming growth factor‐β and platelet‐derived growth factor, were studied. Results showed that byakangelicin can inhibit related pathways. According to the hepatoprotective effect of byakangelicin observed in animal experiments, we studied the effect of byakangelicin on 4‐HNE–induced hepatocyte (HepG2) apoptosis and explored its related pathways. The results showed that byakangelicin could attenuate 4‐HNE–induced hepatocyte apoptosis via inhibiting ASK‐1/JNK signalling. In conclusion, byakangelicin could improve carbon tetrachloride–induced liver fibrosis and liver injury by inhibiting hepatic stellate cell proliferation and activation and suppressing hepatocyte apoptosis.  相似文献   

13.
14.
关于肝纤维化形成的复杂的细胞和分子联系已经有了相当多的研究进展。最近的数据表明,纤维化进程的终止和纤维分解途径的恢复可以逆转晚期肝纤维化甚至肝硬化。因此,需要更好地阐明参与肝纤维化的细胞和分子机制。HSC(肝星状细胞)的激活是肝纤维化发生的中心事件,此外还有产生基质的其他细胞来源,包括肝门区的成纤维细胞,纤维细胞和骨髓来源的肌纤维母细胞。这些细胞与其邻近细胞通过多种联系聚集产生纤维疤痕并造成持续性损伤。阐明不同类型的细胞的相互作用,揭示细胞因子对这些细胞的影响,理清活化HSC基因表达的调控,将有助于我们探索新的肝纤维化治疗靶点。此外,不同的病因有不同的致病途径,弄清这一点有助于针对特异性疾病治疗方法的发现。本文概述了肝纤维化的细胞和分子机制的最新研究进展,可能为未来治疗方法带来新的突破。  相似文献   

15.
Leucocyte infiltration into human liver tissue is a common process in all adult inflammatory liver diseases. Chronic infiltration can drive the development of fibrosis and progression to cirrhosis. Understanding the molecular mechanisms that mediate leucocyte recruitment to the liver could identify important therapeutic targets for liver disease. The key interaction during leucocyte recruitment is that of inflammatory cells with endothelium under conditions of shear stress. Recruitment to the liver occurs within the low shear channels of the hepatic sinusoids which are lined by hepatic sinusoidal endothelial cells (HSEC). The conditions within the hepatic sinusoids can be recapitulated by perfusing leucocytes through channels lined by human HSEC monolayers at specific flow rates. In these conditions leucocytes undergo a brief tethering step followed by activation and firm adhesion, followed by a crawling step and subsequent transmigration across the endothelial layer. Using phase contrast microscopy, each step of this ''adhesion cascade'' can be visualized and recorded followed by offline analysis. Endothelial cells or leucocytes can be pretreated with inhibitors to determine the role of specific molecules during this process.  相似文献   

16.
恶性肿瘤已成为危害人类健康的重要杀手,针对肿瘤的研究也成为当今医学界的热点.含有MARVEL跨膜结构域的趋化素样因子基因家族(CKLF-like MARVEL transmembrane domain containing family of genes,CMTM family),原名人类趋化素样因子超家族(chemo...  相似文献   

17.
Liver fibrosis, an important health condition associated with chronic liver injury that provides a permissive environment for cancer development, is characterized by the persistent deposition of extracellular matrix components that are mainly derived from activated hepatic stellate cells (HSCs). CDH11 belongs to a group of transmembrane proteins that are principally located in adherens junctions. CDH11 mediates homophilic cell-to-cell adhesion, which may promote the development of cirrhosis. The goal of this study was to determine whether CDH11 regulates liver fibrosis and to examine its mechanism by focusing on HSC activation. Here we demonstrate that CDH11 expression is elevated in human and mouse fibrotic liver tissues and that CDH11 mediates the profibrotic response in activated HSCs. Our data indicate that CDH11 regulates the TGFβ-induced activation of HSCs. Moreover, cells from CDH11 deficient mice displayed decreased HSC activation in vitro, and CDH11 deficient mice developed liver fibrogenesis in response to chronic damage induced by CCl4 administration. In addition, CDH11 expression was positively correlated with liver fibrosis in patients with cirrhosis, and could therefore be a prognostic factor in patients with liver fibrosis. Collectively, our findings demonstrate that CDH11 promotes liver fibrosis by activating HSCs and may represent a potential target for anti-fibrotic therapies.  相似文献   

18.
《遗传学报》2022,49(4):338-349
Liver fibrosis is a key transformation stage and also a reversible pathological process in various types of chronic liver diseases. However, the pathogenesis of liver fibrosis still remains elusive. Here, we report that the calcium binding protein A11 (S100A11) is consistently upregulated in the integrated data from GSE liver fibrosis and tree shrew liver proteomics. S100A11 is also experimentally activated in liver fibrosis in mouse, rat, tree shrew, and human with liver fibrosis. While overexpression of S100A11 in vivo and in vitro exacerbates liver fibrosis, the inhibition of S100A11 improves liver fibrosis. Mechanistically, S100A11 activates hepatic stellate cells (HSCs) and the fibrogenesis process via the regulation of the deacetylation of Smad3 in the TGF-β signaling pathway. S100A11 physically interacts with SIRT6, a deacetylase of Smad2/3, which may competitively inhibit the interaction between SIRT6 and Smad2/3. The subsequent release and activation of Smad2/3 promote the activation of HSCs and fibrogenesis. Additionally, a significant elevation of S100A11 in serum is observed in clinical patients. Our study uncovers S100A11 as a novel profibrogenic factor in liver fibrosis, which may represent both a potential biomarker and a promising therapy target for treating liver fibrosis and fibrosis-related liver diseases.  相似文献   

19.
The pathogenesis of hepatic fibrosis involves significant deposition of fibrilar collagen and other extracellular matrix proteins. It is a rather dynamic process of wound healing in response to a variety of persistent liver injury caused by factors such as ethanol intake, viral infection, drugs, toxins, cholestasis, and metabolic disorders. Liver fibrosis distorts the hepatic architecture, decreases the number of endothelial cell fenestrations and causes portal hypertension. Key events are the activation and transformation of quiescent hepatic stellate cells into myofibroblast-like cells with the subsequent up-regulation of proteins such as α-smooth muscle actin, interstitial collagens, matrix metalloproteinases, tissue inhibitor of metalloproteinases, and proteoglycans. Oxidative stress is a major contributing factor to the onset of liver fibrosis and it is typically associated with a decrease in the antioxidant defense. Currently, there is no effective therapy for advanced liver fibrosis. In its early stages, liver fibrosis is reversible upon cessation of the causative agent. In this review, we discuss some aspects on the etiology of liver fibrosis, the cells involved, the molecular pathogenesis, and the current therapeutic approaches.  相似文献   

20.
Trans-differentiation of quiescent hepatic stellate cells (Q-HSCs), which exhibit epithelial and adipocytic features, into myofibroblastic-HSC (MF-HSCs) is a key event in liver fibrosis. Culture models demonstrated that Hedgehog (Hh) pathway activation is required for transition of epithelioid/adipocytic Q-HSCs into MF-HSCs. Hh signaling inhibits adiposity and promotes epithelial-to-mesenchymal transitions (EMTs). Leptin (anti-adipogenic, pro-EMT factor) promotes HSC trans-differentiation and liver fibrosis, suggesting that the pathways may interact to modulate cell fate. This study aimed to determine whether leptin activates Hh signaling and whether this is required for the fibrogenic effects of leptin. Cultures of primary HSCs from lean and fa/fa rats with an inherited ObRb defect were examined. Inhibitors of PI3K/Akt, JAK/STAT, and Hh signaling were used to delineate how ObRb activation influenced Hh signaling and HSC trans-differentiation. Fibrogenesis was compared in wild type and db/db mice (impaired ObRb function) to assess the profibrotic role of leptin. The results demonstrate that leptin-ObR interactions activate Hh signaling with the latter necessary to promote trans-differentiation. Leptin-related increases in Hh signaling required ObR induction of PI3K/Akt, which was sufficient for leptin to repress the epithelioid/adipocytic program. Leptin-mediated induction of JAK/STAT was required for mesenchymal gene expression. Leptin-ObRb interactions were not necessary for HSC trans-differentiation to occur in vitro or in vivo but are important because liver fibrogenesis was attenuated in db/db mice. These findings reveal that leptin activates Hh signaling to alter gene expression programs that control cell fate and have important implications for liver fibrosis and other leptin-regulated processes involving EMTs, including development, obesity, and cancer metastasis.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号