Caveolin 1

小窝蛋白 1
  • 文章类型: Journal Article
    青光眼是一种退行性疾病,其特征是眼内压(IOP)升高导致的视网膜神经节细胞(RGC)死亡和视力损害。IOP升高可以激活小胶质细胞,参与神经节细胞损伤。基于caveolin-1(Cav-1)在青光眼中的研究,我们旨在探讨Cav-1对急性高眼压(AOH)小鼠RGC凋亡的影响及其机制。建立AOH小鼠,并玻璃体内注射Cav-1。从新生小鼠中分离视网膜小胶质细胞和RGC。TUNEL染色,苏木精-伊红染色,免疫组织化学,流式细胞术,采用PCR和免疫印迹法观察Cav-1对RGCs和小鼠视网膜的影响。整个视网膜和内视网膜亚层的厚度明显下降,AOH损伤后视网膜细胞凋亡增加,Cav-1治疗逆转了AOH损伤的作用。此外,Cav-1治疗促进了促炎M1小胶质细胞向抗炎M2小胶质细胞的转化。从新生小鼠中分离小胶质细胞和RGC。Cav-1通过改变体外视网膜小胶质细胞的极化状态保护RGCs免受OGD/R诱导的损伤。进一步研究发现Cav-1激活Akt/PTEN信号通路并抑制TLR4。我们的研究提供了证据,表明Cav-1可能是青光眼的有希望的治疗靶标。
    Glaucoma is a degenerative disease characterized by retinal ganglion cell (RGC) death and visual impairment caused by elevated intraocular pressure (IOP). Elevated IOP can activate microglia, which participate in ganglion cell injury. Based on the study of caveolin-1 (Cav-1) in glaucoma, we aimed to explore the effect and mechanism of Cav-1 on RGC apoptosis in mice with acute ocular hypertension (AOH). AOH mice were established, and Cav-1 was intravitreally injected. Retinal microglia and RGCs were isolated from neonatal mice. TUNEL staining, hematoxylin-eosin staining, immunohistochemistry, flow cytometry, PCR and western blotting were used to observe the effect of Cav-1 on RGCs and mouse retinas. The thickness of the whole retina and the inner retinal sublayer decreased significantly, retinal cell apoptosis increased after AOH injury, and Cav-1 treatment reversed the effect of AOH injury. In addition, Cav-1 treatment promoted the conversion of proinflammatory M1 microglia to anti-inflammatory M2 microglia. Microglia and RGCs were isolated from neonatal mice. Cav-1 protects RGCs from OGD/R-induced injury by changing the polarization status of retinal microglia in vitro. Further studies revealed that Cav-1 activated the Akt/PTEN signaling pathway and inhibited TLR4. Our study provides evidence that Cav-1 may be a promising therapeutic target for glaucoma.
    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
    腹膜转移是导致胃癌预后不良的最常见危险因素之一。我们先前报道了来自胃癌细胞的细胞外囊泡可以促进腹膜转移。然而,它们对缺氧条件下胃癌腹膜转移的影响尚不清楚.本研究旨在阐明耐缺氧胃癌细胞来源的细胞外囊泡如何影响常氧胃癌细胞的腹膜转移。蛋白质组学分析显示,耐缺氧胃癌细胞及其相应的细胞外囊泡中Caveolin1和层粘连蛋白β2的水平升高。重要的是,发现Caveolin1在介导层粘连蛋白β2分选源自缺氧抗性胃癌细胞的细胞外囊泡中起核心作用,随后,细胞外囊泡相关层粘连蛋白β2通过激活AKT通路促进常氧胃癌细胞的腹膜转移。进一步的研究证实,Rho相关卷曲激酶1介导的Y14残基磷酸化激活Caveolin1是促进层粘连蛋白β2分选细胞外囊泡的关键因素。此外,Y14磷酸化-Caveolin1通过激活Rab11增强层粘连蛋白β2分选。最后,我们的研究表明,血浆细胞外囊泡相关Caveolin1和细胞外囊泡相关层粘连蛋白β2的联合评估可为胃癌腹膜转移的发生提供准确的预测工具.
    Peritoneal metastasis is one of the most common risk factors contributing to the poor prognosis of gastric cancer. We previously reported that extracellular vesicles from gastric cancer cells could facilitate peritoneal metastasis. However, their impact on gastric cancer-induced peritoneal metastasis under hypoxic conditions remains unclear. This study aims to elucidate how hypoxia-resistant gastric cancer cell-derived extracellular vesicles affect the peritoneal metastasis of normoxic gastric cancer cells. Proteomic analysis revealed elevated levels of Caveolin1 and Laminin β2 in hypoxia-resistant gastric cancer cells and their corresponding extracellular vesicles. Importantly, Caveolin1 was found to play a central role in mediating Laminin β2 sorting into extracellular vesicles derived from hypoxia-resistant gastric cancer cells, and subsequently, extracellular vesicle-associated Laminin β2 promoted peritoneal metastasis in normoxic gastric cancer cells by activating the AKT pathway. Further investigation confirmed that Caveolin1 activation by Rho-related Coiled-coil kinase 1-mediated phosphorylation of Y14 residue is a key factor facilitating Laminin β2 sorting into extracellular vesicles. Moreover, Y14 phosphorylated- Caveolin1 enhanced Laminin β2 sorting by activating Rab11. Finally, our study demonstrated that a combined assessment of plasma extracellular vesicle-associated Caveolin1 and extracellular vesicle-associated Laminin β2 could provide an accurate predictive tool for peritoneal metastasis occurrence in gastric cancer.
    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:胆固醇代谢重编程被认为是癌症的一个新特征。胰腺导管腺癌(PDAC)是一种对胆固醇需求高的癌症。探讨了PDAC中胆固醇代谢抑制的潜在机制。
    方法:在TCGA数据库中证实了PDAC与胆固醇之间的相关性。在TCGA和GEO数据集中发现了表达和临床关联。采用敲低和过表达AGFG1进行功能研究。RNA测序,胆固醇检测,透射电子显微镜,免疫共沉淀,和免疫荧光等。被用来揭示潜在的机制。
    结果:生物信息学分析显示,AGFG1基因与PDAC中的胆固醇代谢呈正相关。然后发现AGFG1表达与PDAC的不良预后相关。AGFG1敲低导致肿瘤细胞在体外和体内的增殖降低。通过RNA测序,我们发现AGFG1上调表达导致细胞内胆固醇生物合成增强。AGFG1敲低抑制胆固醇生物合成和胆固醇在ER中的积累。机械上,我们证实AGFG1与CAV1相互作用以重新定位胆固醇进行胆固醇生物合成,因此导致细胞内胆固醇代谢紊乱。
    结论:我们的研究证明了AGFG1通过干扰PDAC中胆固醇代谢引起的肿瘤促进作用。我们的研究提出了基于PDAC中胆固醇代谢的癌症治疗方法的新视角。
    OBJECTIVE: Cholesterol metabolism reprograming has been acknowledged as a novel feature of cancers. Pancreatic ductal adenocarcinoma (PDAC) is a cancer with a high demand of cholesterol for rapid growth. The underlying mechanism of how cholesterol metabolism homestasis are disturbed in PDAC is explored.
    METHODS: The relevance between PDAC and cholesterol was confirmed in TCGA database. The expression and clinical association were discovered in TCGA and GEO datasets. Knockdown and overexpression of AGFG1 was adopted to perform function studies. RNA sequencing, cholesterol detection, transmission electron microscope, co-immunoprecipitation, and immunofluorescence et al. were utilized to reveal the underlying mechanism.
    RESULTS: AGFG1 was identified as one gene positively correlated with cholesterol metabolism in PDAC as revealed by bioinformatics analysis. AGFG1 expression was then found associated with poor prognosis in PDAC. AGFG1 knockdown led to decreased proliferation of tumor cells both in vitro and in vivo. By RNA sequencing, we found AGFG1 upregulated expression leads to enhanced intracellular cholesterol biosynthesis. AGFG1 knockdown suppressed cholesterol biosynthesis and an accumulation of cholesterol in the ER. Mechanistically, we confirmed that AGFG1 interacted with CAV1 to relocate cholesterol for the proceeding of cholesterol biosynthesis, therefore causing disorders in intracellular cholesterol metabolism.
    CONCLUSIONS: Our study demonstrates the tumor-promoting role of AGFG1 by disturbing cholesterol metabolism homestasis in PDAC. Our study has present a new perspective on cancer therapeutic approach based on cholerstrol metabolism in PDAC.
    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
    动静脉瘘(AVF)是尿毒症患者血液透析的首选血管通路,然而,其功能障碍构成了重大的临床挑战。静脉狭窄,主要由静脉新生内膜增生引起,是血管通路失败的关键因素。在血管通路功能障碍期间,内皮细胞(EC)将机械刺激转化为细胞内信号并与血管平滑肌细胞相互作用。丹参酮IIA,一种来自丹参的重要化合物,已被广泛用于治疗心血管疾病。然而,其在尿毒症条件下调节ECs的作用尚不完全清楚。在这项研究中,将EC暴露于丹参酮IIA磺酸钠(STS)并经受剪切应力和尿毒症条件。结果表明,STS可以降低尿毒症诱导的ECs对NF-κBp65,JNK和I型胶原表达的抑制作用。此外,通过抑制ERK1/2和上调Caveolin-1,可以增强NF-κBp65,JNK和胶原蛋白I的下调。这些结果表明,丹参酮IIA可能通过靶向小窝蛋白-1/ERK1/2途径改善尿毒症条件下的EC功能,提出丹参酮IIA作为一种潜在的治疗药物,用于治疗由EC功能障碍引起的AVF不成熟。
    An arteriovenous fistula (AVF) is the preferred vascular access for hemodialysis in uremic patients, yet its dysfunction poses a significant clinical challenge. Venous stenosis, primarily caused by venous neointimal hyperplasia, is a key factor in the failure of vascular access. During vascular access dysfunction, endothelial cells (ECs) transform mechanical stimuli into intracellular signals and interact with vascular smooth muscle cells. Tanshinone IIA, an important compound derived from Salvia miltiorrhiza, has been widely used to treat cardiovascular diseases. However, its role in modulating ECs under uremic conditions remains incompletely understood. In this research, ECs were exposed to sodium tanshinone IIA sulfonate (STS) and subjected to shear stress and uremic conditions. The results indicate that STS can reduce the suppressive effects on the expression of NF-κB p65, JNK and Collagen I in uremia-induced ECs. Moreover, the downregulation of NF-κB p65, JNK and Collagen I can be enhanced through the inhibition of ERK1/2 and the upregulation of Caveolin-1. These findings suggest that tanshinone IIA may improve EC function under uremic conditions by targeting the Caveolin-1/ERK1/2 pathway, presenting tanshinone IIA as a potential therapeutic agent against AVF immaturity caused by EC dysfunction.
    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:小窝蛋白-1(Cav-1)和Wnt/β-catenin信号通路在脑缺血再灌注(I/R)损伤中的作用已得到证实。β-catenin易位到细胞核是调节神经元凋亡的关键,修复,和缺血脑内的神经发生。据报道,小窝蛋白-1(Cav-1)的支架结构域(残基95-98)与β-连环蛋白(残基330-337)相互作用。然而,Cav-1/β-catenin复合物对I/R损伤的具体作用尚不清楚。
    结果:为了研究Cav-1/β-catenin复合物参与β-catenin亚细胞易位的机制及其对脑I/R损伤的后续影响,我们用ASON(Cav-1反义寡脱氧核苷酸)或FTVT(Cav-1和β-catenin相互作用的竞争性肽拮抗剂)治疗缺血性脑。我们的研究表明,I/R损伤后Cav-1与β-catenin的结合阻止了β-catenin的核积累。I/R损伤后用ASON或FTVT治疗可显着增加核β-连环蛋白的水平。此外,ASON降低了β-catenin在Ser33,Ser37和Thr41的磷酸化,这有助于其蛋白酶体降解,而FTVT在Tyr333处增加磷酸化,这与其核易位有关。
    结论:上述结果表明,在I/R损伤后,Cav-1/β-catenin复合物的形成在细胞质中锚定了β-catenin。此外,ASON和FTVT治疗均减轻了缺血性脑中的神经元死亡。我们的研究表明,靶向Cav-1和β-catenin之间的相互作用可作为一种新型的治疗策略,以防止脑损伤期间的神经元损伤。
    BACKGROUND: The roles of Caveolin-1 (Cav-1) and the Wnt/β-catenin signaling pathways in cerebral ischemia-reperfusion (I/R) injury are well established. The translocation of β-catenin into the nucleus is critical for regulating neuronal apoptosis, repair, and neurogenesis within the ischemic brain. It has been reported that the scaffold domain of Caveolin-1 (Cav-1) (residues 95-98) interacts with β-catenin (residues 330-337). However, the specific contribution of the Cav-1/β-catenin complex to I/R injury remains unknown.
    RESULTS: To investigate the mechanism underlying the involvement of the Cav-1/β-catenin complex in the subcellular translocation of β-catenin and its subsequent effects on cerebral I/R injury, we treated ischemic brains with ASON (Cav-1 antisense oligodeoxynucleotides) or FTVT (a competitive peptide antagonist of the Cav-1 and β-catenin interaction). Our study demonstrated that the binding of Cav-1 to β-catenin following I/R injury prevented the nuclear accumulation of β-catenin. Treatment with ASON or FTVT after I/R injury significantly increased the levels of nuclear β-catenin. Furthermore, ASON reduced the phosphorylation of β-catenin at Ser33, Ser37, and Thr41, which contributes to its proteasomal degradation, while FTVT increased phosphorylation at Tyr333, which is associated with its nuclear translocation.
    CONCLUSIONS: The above results indicate that the formation of the Cav-1/β-catenin complex anchors β-catenin in the cytoplasm following I/R injury. Additionally, both ASON and FTVT treatments attenuated neuronal death in ischemic brains. Our study suggests that targeting the interaction between Cav-1 and β-catenin serve as a novel therapeutic strategy to protect against neuronal damage during cerebral injury.
    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
    基因治疗的兴起,解决了许多常规方法无法有效治疗的疾病。基因载体对于保护治疗基因并将其递送到靶位点非常重要。甘露醇修饰的聚乙烯亚胺(PEI)可以提高我们小组先前报道的基因转染效率。为了进一步控制和提高有效基因释放到作用位点,将二硫键引入甘露醇修饰的PEI中以构建新的非病毒基因载体PeiSM。筛选甘露醇与二硫键连接的程度。其中,中度甘露醇修饰的具有二硫键的聚乙烯亚胺(M-PeiSM)显示出最佳的转染效率,和显着增强长期系统性转基因表达72小时在体内即使在单剂量给药,并且可以通过上调caveolin-1的磷酸化来促进caveole介导的摄取,并增加高GSH细胞内环境中纳米复合物的负载基因释放。这种功能化的基因递送系统可以用作进一步基因治疗的潜在和安全的非病毒纳米载体。
    The rise of gene therapy has solved many diseases that cannot be effectively treated by conventional methods. Gene vectors is very important to protect and deliver the therapeutic genes to the target site. Polyethyleneimine (PEI) modified with mannitol could enhance the gene transfection efficiency reported by our group previously. In order to further control and improve the effective gene release to action site, disulfide bonds were introduced into mannitol-modified PEI to construct new non-viral gene vectors PeiSM. The degrees of mannitol linking with disulfide bonds were screened. Among them, moderate mannitol-modified PEI with disulfide bonds showed the best transfection efficiency, and significantly enhanced long-term systemic transgene expression for 72 hin vivoeven at a single dose administration, and could promote caveolae-mediated uptake through up-regulating the phosphorylation of caveolin-1 and increase the loaded gene release from the nanocomplexes in high glutathione intracellular environment. This functionalized gene delivery system can be used as an potential and safe non-viral nanovector for further gene therapy.
    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
    迁徙者,在迁移细胞的回缩纤维上产生的囊泡细胞器,在迁徙过程中起着至关重要的作用,介导细胞间通讯。货物决定了迁移体的功能特异性。Migrasomes藏有许多管腔内囊泡,他们货物的关键组成部分。这些腔内囊泡运输到迁移体的潜在机制仍然是神秘的。在这项研究中,我们发现Rab10和Caveolin-1(CAV1)标记了迁移体中的腔内囊泡。Rab10-CAV1囊泡向迁移体的转运需要运动蛋白肌球蛋白Va和衔接蛋白RILPL2。值得注意的是,激酶LRRK2对Rab10的磷酸化调节了这一过程。此外,CSF-1可以通过这种机制转运到迁移体,随后促进皮肤伤口愈合中的单核细胞-巨噬细胞分化,这证明了这种运输机制的生理重要性。
    Migrasomes, vesicular organelles generated on the retraction fibers of migrating cells, play a crucial role in migracytosis, mediating intercellular communication. The cargoes determine the functional specificity of migrasomes. Migrasomes harbor numerous intraluminal vesicles, a pivotal component of their cargoes. The mechanism underlying the transportation of these intraluminal vesicles to the migrasomes remains enigmatic. In this study, we identified that Rab10 and Caveolin-1 (CAV1) mark the intraluminal vesicles in migrasomes. Transport of Rab10-CAV1 vesicles to migrasomes required the motor protein Myosin Va and adaptor proteins RILPL2. Notably, the phosphorylation of Rab10 by the kinase LRRK2 regulated this process. Moreover, CSF-1 can be transported to migrasomes through this mechanism, subsequently fostering monocyte-macrophage differentiation in skin wound healing, which served as a proof of the physiological importance of this transporting mechanism.
    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
    肺部疾病中的气道重塑可以通过抑制过度的平滑肌细胞增殖来治疗。Zedoarondiol(Zed)是从中药姜黄中分离出的天然化合物。小窝蛋白-1(CAV-1)在肺细胞中广泛表达,并在血小板衍生生长因子(PDGF)信号传导和细胞增殖中起关键作用。本研究旨在研究Zed对人支气管平滑肌细胞(HBSMC)增殖的影响,并探讨其潜在的分子机制。我们评估了Zed对PDGF刺激的HBSMC增殖的影响,并进行了蛋白质组学分析以鉴定潜在的分子靶标和途径。CAV1siRNA用于在体外验证我们的发现。在PDGF刺激的HBSMC中,Zed显著抑制HBSMC的过度增殖。对zedoarondiol处理的HBSMC的蛋白质组学分析显示,在细胞增殖相关途径和生物学过程中差异表达的蛋白质显着富集。Zed抑制HBSMC增殖与CAV1的上调,CAV-1/PDGF途径的调节以及MAPK和PI3K/AKT信号通路激活的抑制有关。用CAV1siRNA处理HBSMC部分逆转了Zed对HBSMC增殖的抑制作用。因此,这项研究表明,zedoarondiol通过上调CAV-1表达有效抑制HBSMC增殖,强调其在气道重塑和相关疾病中的潜在价值。
    Airway remodelling in lung diseases can be treated by inhibiting excessive smooth muscle cell proliferation. Zedoarondiol (Zed) is a natural compound isolated from the Chinese herb Curcuma longa. The caveolin-1 (CAV-1) is widely expressed in lung cells and plays a key role in platelet-derived growth factor (PDGF) signalling and cell proliferation. This study aims to investigate the effect of Zed on human bronchial smooth muscle cell (HBSMC) proliferation and explore its potential molecular mechanisms. We assessed the effect of Zed on the proliferation of PDGF-stimulated HBSMCs and performed proteomic analysis to identify potential molecular targets and pathways. CAV1 siRNA was used to validate our findings in vitro. In PDGF-stimulated HBSMCs, Zed significantly inhibited excessive proliferation of HBSMCs. Proteomic analysis of zedoarondiol-treated HBSMCs revealed significant enrichment of differentially expressed proteins in cell proliferation-related pathways and biological processes. Zed inhibition of HBSMC proliferation was associated with upregulation of CAV1, regulation of the CAV-1/PDGF pathway and inhibition of MAPK and PI3K/AKT signalling pathway activation. Treatment of HBSMCs with CAV1 siRNA partly reversed the inhibitory effect of Zed on HBSMC proliferation. Thus, this study reveals that zedoarondiol potently inhibits HBSMC proliferation by upregulating CAV-1 expression, highlighting its potential value in airway remodelling and related diseases.
    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
    肥胖已成为全球主要的健康问题,对骨形成的影响越来越受到关注。然而,肥胖和骨代谢之间的相互作用是复杂的,仍然没有完全理解。这里,我们发现caveolin-1(Cav1),参与调节多种细胞过程的膜支架蛋白,作为连接肥胖和骨代谢的桥梁,起着关键的调节作用。高脂饮食(HFD)诱导的肥胖C57BL/6J小鼠表现出Cav1表达的显着增加和成骨活性的降低;用1mM游离脂肪酸(FFA)体外处理成骨细胞MC3T3-E1细胞显着促进Cav1表达,PINK1/Parkin调节线粒体自噬,但抑制成骨标记基因的表达。相反,Cav1基因表达的降低阻止了这些影响。在FFA处理的细胞中,Cav1敲低后,内源性氧化应激和Sirt1途径也显着降低。最后,Cav1-Sirt1对接和免疫共沉淀结果表明,Cav1与Sirt1和FFA相互作用增强了相互作用。一起来看,这些结果表明,肥胖通过上调Cav1基因损害骨骼发育和形成,通过与Sirt1分子的相互作用抑制Sirt1/FOXO1和Sirt1/PGC-1α信号通路,和线粒体自噬水平的增加。
    Obesity has become a major global health problem and the effect on bone formation has received increasing attention. However, the interaction between obesity and bone metabolism is complex and still not fully understood. Here, we show that caveolin-1 (Cav1), a membrane scaffold protein involved in regulating a variety of cellular processes, plays a key regulatory role as a bridge connecting obesity and bone metabolism. High-fat diet (HFD)-induced obese C57BL/6J mouse displayed a significant increase in Cav1 expression and lower osteogenic activity; In vitro treatment of osteoblastic MC3T3-E1 cells with 1 mM free fatty acids (FFA) significantly promoted Cav1 expression and PINK1/Parkin regulated mitophagy, but inhibited the expression of osteogenic marker genes. Conversely, reduced expression of the Cav1 gene prevented these effects. Both endogenous oxidative stress and Sirt1 pathway were also significantly reduced after Cav1 knockdown in FFA-treated cells. Finally, Cav1-Sirt1 docking and co-immunoprecipitation results showed that Cav1 interacted with Sirt1 and FFA enhanced the interaction. Taken together, these results suggest that obesity impairs bone development and formation through up-regulation of the Cav1 gene, which lead to inhibition of Sirt1/FOXO1 and Sirt1/PGC-1α signaling pathways through interacting with Sirt1 molecule, and an increase of mitophagy level.
    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:创伤导致的伤口不愈合,手术,慢性疾病每年影响全球数百万人,由于对控制组织修复和再生的分子过程的理解不完全,因此可用的治疗策略有限。丹酚酸B(SalB)在促进血管生成和抑制炎症方面显示出有希望的生物活性。然而,其在组织再生中的调控机制尚不清楚。
    目的:本研究旨在研究SalB对伤口愈合和再生过程的影响,连同其潜在的分子机制,通过使用斑马鱼作为模型生物。
    方法:在本研究中,我们采用了多方面的方法来评估SalB对斑马鱼尾鳍再生的影响。我们利用了全鱼免疫荧光,TUNEL染色,线粒体膜电位(MMP),和吖啶橙(AO)探针分析SalB治疗下的组织修复和再生。此外,我们利用转基因斑马鱼菌株研究了在鳍再生的不同阶段炎症细胞的迁移。为了验证小窝蛋白-1(Cav1)在组织再生中的重要性,我们使用分子对接和基于吗啉代的基因敲低技术探讨了其功能作用。此外,我们通过应用原位杂交定量Cav1表达水平。
    结果:我们的研究结果表明,SalB通过促进细胞增殖的多方面机制加速斑马鱼尾鳍再生,抑制细胞凋亡,和增强MMP。此外,发现SalB在组织再生过程中对免疫细胞的动态聚集和随后的消退施加调节控制。重要的是,我们观察到Cav1的敲低显著损害了组织再生,导致免疫细胞过度浸润和凋亡水平增加。此外,Cav1的击倒也会影响胚层的形成,Cav1影响组织再生的关键过程。
    结论:这项研究的结果表明,SalB通过调节免疫细胞迁移和Cav1介导的成纤维细胞活化来促进组织修复和再生,促进胚芽的形成和发展。这项研究强调了SalB在促进组织再生方面的潜在药理作用。这些发现促进了再生医学研究的发展和创伤新治疗方法的发展。
    BACKGROUND: Non-healing wounds resulting from trauma, surgery, and chronic diseases annually affect millions of individuals globally, with limited therapeutic strategies available due to the incomplete understanding of the molecular processes governing tissue repair and regeneration. Salvianolic acid B (Sal B) has shown promising bioactivities in promoting angiogenesis and inhibiting inflammation. However, its regulatory mechanisms in tissue regeneration remain unclear.
    OBJECTIVE: This study aims to investigate the effects of Sal B on wound healing and regeneration processes, along with its underlying molecular mechanisms, by employing zebrafish as a model organism.
    METHODS: In this study, we employed a multifaceted approach to evaluate the impact of Sal B on zebrafish tail fin regeneration. We utilized whole-fish immunofluorescence, TUNEL staining, mitochondrial membrane potential (MMP), and Acridine Orange (AO) probes to analyze the tissue repair and regenerative under Sal B treatment. Additionally, we utilized transgenic zebrafish strains to investigate the migration of inflammatory cells during different phases of fin regeneration. To validate the importance of Caveolin-1 (Cav1) in tissue regeneration, we delved into its functional role using molecular docking and Morpholino-based gene knockdown techniques. Additionally, we quantified Cav1 expression levels through the application of in situ hybridization.
    RESULTS: Our findings demonstrated that Sal B expedites zebrafish tail fin regeneration through a multifaceted mechanism involving the promotion of cell proliferation, suppression of apoptosis, and enhancement of MMP. Furthermore, Sal B was found to exert regulatory control over the dynamic aggregation and subsequent regression of immune cells during tissue regenerative processes. Importantly, we observed that the knockdown of Cav1 significantly compromised tissue regeneration, leading to an excessive infiltration of immune cells and increased levels of apoptosis. Moreover, the knockdown of Cav1 also affects blastema formation, a critical process influenced by Cav1 in tissue regeneration.
    CONCLUSIONS: The results of this study showed that Sal B facilitated tissue repair and regeneration through regulating of immune cell migration and Cav1-mediated fibroblast activation, promoting blastema formation and development. This study highlighted the potential pharmacological effects of Sal B in promoting tissue regeneration. These findings contributed to the advancement of regenerative medicine research and the development of novel therapeutic approaches for trauma.
    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