cathepsin D

组织蛋白酶 D
  • 文章类型: Journal Article
    我们先前表达了一种嵌合蛋白,其中小的热休克蛋白αB-晶状体蛋白(αBC)在其N端融合到内质网(ER)蛋白mitsugumin23的第一个跨膜片段的C端,并确认其定位到ER。此外,该N末端修饰的αBC的过表达显示阻止共表达的R120GαBC变体的聚集,这是高度聚集的倾向,并与遗传性肌病αB晶体病有关。为了揭示ER锚定的αBC负调节蛋白质聚集的分子机制,我们分离了与ER锚定的αBC结合的蛋白质,并将溶酶体蛋白酶组织蛋白酶D(CTSD)鉴定为一种此类相互作用蛋白。蛋白水解活性CTSD是通过多步处理前组织蛋白酶D(proCTSD)产生的,最初在ER中合成并递送至溶酶体。当过度表达时,CTSD本身阻止共表达的R120GαBC变体聚集。这种抗聚集活性也在W383CCTSD变体的过表达后引发,主要被隔离在急诊室,因此仍未处理,表明proCTSD,而不是成熟的CTSD,用于抑制R120GαBC变体的聚集。同时,A58VCTSD变体的过表达,除了前肽内的Ala58Val取代外,它与野生型CTSD相同,没有抑制蛋白质聚集,表明前肽的完整性是proCTSD发挥其抗聚集活性所必需的。根据我们以前的发现,ER跨膜蛋白CLN6(宫颈脂褐变,神经元6),鉴定为ER锚定的αBC的相互作用蛋白,防止R120GαBC变体聚集,CLN6-proCTSD偶联被假设为支持proCTSD在ER内的功能.的确,CTSD,当在CLN6耗尽的细胞中过表达时,无法发挥其抗聚集活性,支持我们的观点。总的来说,我们在这里表明,proCTSD通过与CLN6在ER膜周围的微环境中的功能关联来防止蛋白质聚集,揭示了proCTSD的一个新方面及其在CTSD相关疾病中的潜在参与,其特征是异常蛋白聚集体的积累。
    We previously expressed a chimeric protein in which the small heat-shock protein αB-crystallin (αBC) is fused at its N-terminus to the C-terminus of the first transmembrane segment of the endoplasmic reticulum (ER) protein mitsugumin 23 and confirmed its localization to the ER. Moreover, overexpression of this N-terminally modified αBC was shown to prevent the aggregation of the coexpressed R120G αBC variant, which is highly aggregation-prone and associated with the hereditary myopathy αB-crystallinopathy. To uncover a molecular mechanism by which the ER-anchored αBC negatively regulates the protein aggregation, we isolated proteins that bind to the ER-anchored αBC and identified the lysosomal protease cathepsin D (CTSD) as one such interacting protein. Proteolytically active CTSD is produced by multi-step processing of pro-cathepsin D (proCTSD), which is initially synthesized in the ER and delivered to lysosomes. When overexpressed, CTSD itself prevented the coexpressed R120G αBC variant from aggregating. This anti-aggregate activity was also elicited upon overexpression of the W383C CTSD variant, which is predominantly sequestered in the ER and consequently remains unprocessed, suggesting that proCTSD, rather than mature CTSD, serves to suppress the aggregation of the R120G αBC variant. Meanwhile, overexpression of the A58V CTSD variant, which is identical to wild-type CTSD except for the Ala58Val substitution within the pro-peptide, did not suppress the protein aggregation, indicating that the integrity of the pro-peptide is required for proCTSD to exert its anti-aggregate activity. Based on our previous finding that overexpression of the ER transmembrane protein CLN6 (ceroid-lipofuscinosis, neuronal 6), identified as an interacting protein of the ER-anchored αBC, prevents the R120G αBC variant from aggregating, the CLN6-proCTSD coupling was hypothesized to underpin the functionality of proCTSD within the ER. Indeed, CTSD, when overexpressed in CLN6-depleted cells, was unable to exert its anti-aggregate activity, supporting our view. Collectively, we show here that proCTSD prevents the protein aggregation through the functional association with CLN6 in the microenvironment surrounding the ER membrane, shedding light on a novel aspect of proCTSD and its potential involvement in CTSD-related disorders characterized by the accumulation of aberrant protein aggregates.
    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:在工业化国家,纤维化导致45%的死亡,其特征是细胞外基质(ECM)的异常积累。没有针对肝纤维化的特异性抗纤维化治疗,以前在药物开发方面的失败尝试都集中在预防ECM沉积上。因为肝纤维化在很大程度上被认为是可逆的,调节纤维化分辨率可以提供新的治疗选择。然而,关于在分辨过程中控制ECM重塑的机制知之甚少。蛋白水解活性的变化对于ECM稳态是必需的,并且巨噬细胞是蛋白酶的重要来源。在这里,在这项研究中,我们评估了巨噬细胞衍生的组织蛋白酶D(CtsD)在肝纤维化中的作用。
    方法:在人类肝硬化的单细胞RNA测序和转录组数据集中对CtsD表达和相关通路进行了表征。肝纤维化进展,在新型髓样CtsD和肝细胞CtsD敲除小鼠中评估了逆转和功能表征。
    结果:单细胞RNA测序数据集的分析表明,CtsD在人肝硬化的巨噬细胞和肝细胞中表达。肝纤维化进展,在新型髓样CtsD(CtsDΔMyel)和肝细胞CtsD敲除小鼠中评估了逆转和功能表征。巨噬细胞中的CtsD缺失,但不是在肝细胞中,导致肝纤维化增强。在纤维化CtsDΔMyel肝中富含炎性和基质蛋白质组特征。此外,CtsDΔMyel肝巨噬细胞显示出功能性,表型和分泌组学变化,这导致了退化的表型转变,负责胶原蛋白I的体外蛋白水解加工缺陷和体内纤维化解决过程中胶原蛋白重塑受损。最后,肝硬化人肝脏中表达CtsD的单核吞噬细胞在溶酶体和ECM降解信号通路中富集。
    结论:我们的工作首次描述了CtsD驱动的溶酶体活性作为纤维化解决过程中恢复巨噬细胞功能的中心枢纽,并开辟了新的途径来探索其降解组景观,为药物开发提供信息。
    OBJECTIVE: Fibrosis contributes to 45% of deaths in industrialized nations and is characterized by an abnormal accumulation of extracellular matrix (ECM). There are no specific anti-fibrotic treatments for liver fibrosis, and previous unsuccessful attempts at drug development have focused on preventing ECM deposition. Because liver fibrosis is largely acknowledged to be reversible, regulating fibrosis resolution could offer novel therapeutical options. However, little is known about the mechanisms controlling ECM remodeling during resolution. Changes in proteolytic activity are essential for ECM homeostasis and macrophages are an important source of proteases. Herein, in this study we evaluate the role of macrophage-derived cathepsin D (CtsD) during liver fibrosis.
    METHODS: CtsD expression and associated pathways were characterized in single-cell RNA sequencing and transcriptomic datasets in human cirrhosis. Liver fibrosis progression, reversion and functional characterization were assessed in novel myeloid-CtsD and hepatocyte-CtsD knock-out mice.
    RESULTS: Analysis of single-cell RNA sequencing datasets demonstrated CtsD was expressed in macrophages and hepatocytes in human cirrhosis. Liver fibrosis progression, reversion and functional characterization were assessed in novel myeloid-CtsD (CtsDΔMyel) and hepatocyte-CtsD knock-out mice. CtsD deletion in macrophages, but not in hepatocytes, resulted in enhanced liver fibrosis. Both inflammatory and matrisome proteomic signatures were enriched in fibrotic CtsDΔMyel livers. Besides, CtsDΔMyel liver macrophages displayed functional, phenotypical and secretomic changes, which resulted in a degradomic phenotypical shift, responsible for the defective proteolytic processing of collagen I in vitro and impaired collagen remodeling during fibrosis resolution in vivo. Finally, CtsD-expressing mononuclear phagocytes of cirrhotic human livers were enriched in lysosomal and ECM degradative signaling pathways.
    CONCLUSIONS: Our work describes for the first-time CtsD-driven lysosomal activity as a central hub for restorative macrophage function during fibrosis resolution and opens new avenues to explore their degradome landscape to inform drug development.
    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
    未折叠蛋白反应(UPR)通过多种策略缓解内质网(ER)应激,包括减少蛋白质合成,增加蛋白质折叠能力,并增强错误折叠的蛋白质降解。经过多组学分析,我们发现信号识别粒子14(SRP14),SRP的重要组成部分,在经历内质网应激的细胞中显著减少。进一步的实验表明,SRP14的减少需要PRKR样ER激酶(PERK)介导的真核翻译起始因子2α(eIF2α)磷酸化,但与ATF4或ATF3转录因子无关。SRP14的减少与融合蛋白和内源性组织蛋白酶D的易位减少相关。具有延伸阻滞能力的SRP14变体的强制表达可防止组织蛋白酶D在应激细胞中的易位减少。而没有活性的SRP14突变体则没有。最后,SRP14的过表达增加了UPR并加重了ER应激诱导的细胞死亡。这些数据表明由PERK-SRP14轴介导的转位衰减是UPR减轻ER应激的保护性措施。
    The unfolded protein response (UPR) relieves endoplasmic reticulum (ER) stress through multiple strategies, including reducing protein synthesis, increasing protein folding capabilities, and enhancing misfolded protein degradation. After a multi-omics analysis, we find that signal recognition particle 14 (SRP14), an essential component of the SRP, is markedly reduced in cells undergoing ER stress. Further experiments indicate that SRP14 reduction requires PRKR-like ER kinase (PERK)-mediated eukaryotic translation initiation factor 2α (eIF2α) phosphorylation but is independent of ATF4 or ATF3 transcription factors. The decrease of SRP14 correlates with reduced translocation of fusion proteins and endogenous cathepsin D. Enforced expression of an SRP14 variant with elongation arrest capability prevents the reduced translocation of cathepsin D in stressed cells, whereas an SRP14 mutant without the activity does not. Finally, overexpression of SRP14 augments the UPR and aggravates ER-stress-induced cell death. These data suggest that translocational attenuation mediated by the PERK-SRP14 axis is a protective measure for the UPR to mitigate ER stress.
    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
    尽管血脑屏障(BBB)在阿尔茨海默病(AD)的病因和发病机制中起重要作用的概念已被越来越多的人接受。人们对它的实际贡献知之甚少。我们和其他人最近确定了一种新的功能不同的BBB周细胞(PC)子集。在本研究中,我们试图通过免疫组织化学和淀粉样蛋白β(Aβ)肽组学来确定这些PC亚群是否差异有助于AD相关病理。我们证明,与年龄匹配相比,AD患者的疾病相关PC子集(PC2)扩大,认知未受损的控制。令人惊讶的是,我们发现,在AD患者中,PC2百分比(%PC2)的增加与BBB击穿呈负相关,与自然衰老或其他报道的疾病状况不同。如通过免疫组织化学测定的,AD患者中较高的%PC2还与较低的Aβ42斑块负荷和较低的Aβ42:Aβ40比率相关。多色共聚焦免疫荧光显微镜图像的共定位分析表明,由于Aβ42被生理正常PC亚群(PC1)内化,具有低%PC2的AD患者具有更高的BBB分解,并且它们伴随的细胞死亡导致更多没有PC的血管和增加的斑块负荷。相反,似乎PC2可以分泌组织蛋白酶D,将PC2外部积累的Aβ裂解和降解成更可溶的形式,最终有助于减少BBB分解并减少Aβ斑块负荷。总的来说,我们的数据显示了PC1和PC2在高Aβ条件下的功能不同机制,证明在研究神经血管功能障碍对AD发病机制的贡献时正确识别这些人群的重要性。
    Although the concept that the blood-brain barrier (BBB) plays an important role in the etiology and pathogenesis of Alzheimer\'s disease (AD) has become increasingly accepted, little is known yet about how it actually contributes. We and others have recently identified a novel functionally distinct subset of BBB pericytes (PCs). In the present study, we sought to determine whether these PC subsets differentially contribute to AD-associated pathologies by immunohistochemistry and amyloid beta (Aβ) peptidomics. We demonstrated that a disease-associated PC subset (PC2) expanded in AD patients compared to age-matched, cognitively unimpaired controls. Surprisingly, we found that this increase in the percentage of PC2 (%PC2) was correlated negatively with BBB breakdown in AD patients, unlike in natural aging or other reported disease conditions. The higher %PC2 in AD patients was also correlated with a lower Aβ42 plaque load and a lower Aβ42:Aβ40 ratio in the brain as determined by immunohistochemistry. Colocalization analysis of multicolor confocal immunofluorescence microscopy images suggests that AD patient with low %PC2 have higher BBB breakdown due to internalization of Aβ42 by the physiologically normal PC subset (PC1) and their concomitant cell death leading to more vessels without PCs and increased plaque load. On the contrary, it appears that PC2 can secrete cathepsin D to cleave and degrade Aβ built up outside of PC2 into more soluble forms, ultimately contributing to less BBB breakdown and reducing Aβ plaque load. Collectively our data shows functionally distinct mechanisms for PC1 and PC2 in high Aβ conditions, demonstrating the importance of correctly identifying these populations when investigating the contribution of neurovascular dysfunction to AD pathogenesis.
    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
    DNAJC6基因的纯合突变导致常染色体隐性家族性19型帕金森病(PARK19)。为了验证PARK19DNAJC6突变通过减少功能性DNAJC6的蛋白表达并引起DNAJC6缺失而诱导多巴胺能细胞神经变性的假设,通过使用shRNA介导的内源性DANJC6基因沉默在分化的人SH-SY5Y多巴胺能神经元中构建体外PARK19模型.靶向DNAJC6的shRNA诱导多巴胺能细胞的神经变性。DNAJC6缺乏降低了多巴胺能神经元中胞质网格蛋白重链的水平和溶酶体的数量。DNAJC6缺乏诱导的溶酶体数量减少下调了溶酶体蛋白酶组织蛋白酶D的蛋白质水平和受损的巨自噬,导致内质网(ER)和线粒体中病理性α-突触核蛋白或磷酸-α-突触核蛋白Ser129的上调。α-突触核蛋白shRNA或组织蛋白酶D的表达阻断了DNAJC6缺陷引起的多巴胺能细胞变性。由DNAJC6缺乏激活的ER应激引起的ERα-突触核蛋白或磷酸-α-突触核蛋白Ser129的增加,未折叠的蛋白质反应和ER应激触发的凋亡信号。缺乏DNAJC6诱导的线粒体α-突触核蛋白上调会使线粒体膜电位去极化,并升高线粒体超氧化物水平。DNAJC6缺乏诱发的内质网应激相关凋亡级联反应,线粒体功能障碍和氧化应激通过激活线粒体促凋亡信号诱导多巴胺能神经元变性。与WTDNAJC6的神经保护功能相反,PARK19DNAJC6突变体(Q789X或R927G)未能减弱胞衣霉素或鱼藤酮诱导的病理性α-突触核蛋白上调和凋亡信号刺激。我们的数据表明,PARK19突变诱导的DNAJC6缺乏通过下调蛋白酶组织蛋白酶D和上调神经毒性α-突触核蛋白导致多巴胺能神经元变性。我们的结果还表明PARK19突变(Q789X或R927G)损害DNAJC6介导的神经保护功能。
    A homozygous mutation of the DNAJC6 gene causes autosomal recessive familial type 19 of Parkinson\'s disease (PARK19). To test the hypothesis that PARK19 DNAJC6 mutations induce the neurodegeneration of dopaminergic cells by reducing the protein expression of functional DNAJC6 and causing DNAJC6 paucity, an in vitro PARK19 model was constructed by using shRNA-mediated gene silencing of endogenous DANJC6 in differentiated human SH-SY5Y dopaminergic neurons. shRNA targeting DNAJC6 induced the neurodegeneration of dopaminergic cells. DNAJC6 paucity reduced the level of cytosolic clathrin heavy chain and the number of lysosomes in dopaminergic neurons. A DNAJC6 paucity-induced reduction in the lysosomal number downregulated the protein level of lysosomal protease cathepsin D and impaired macroautophagy, resulting in the upregulation of pathologic α-synuclein or phospho-α-synucleinSer129 in the endoplasmic reticulum (ER) and mitochondria. The expression of α-synuclein shRNA or cathepsin D blocked the DNAJC6 deficiency-evoked degeneration of dopaminergic cells. An increase in ER α-synuclein or phospho-α-synucleinSer129 caused by DNAJC6 paucity activated ER stress, the unfolded protein response and ER stress-triggered apoptotic signaling. The lack of DNAJC6-induced upregulation of mitochondrial α-synuclein depolarized the mitochondrial membrane potential and elevated the mitochondrial level of superoxide. The DNAJC6 paucity-evoked ER stress-related apoptotic cascade, mitochondrial malfunction and oxidative stress induced the degeneration of dopaminergic neurons via activating mitochondrial pro-apoptotic signaling. In contrast with the neuroprotective function of WT DNAJC6, the PARK19 DNAJC6 mutants (Q789X or R927G) failed to attenuate the tunicamycin- or rotenone-induced upregulation of pathologic α-synuclein and stimulation of apoptotic signaling. Our data suggest that PARK19 mutation-induced DNAJC6 paucity causes the degeneration of dopaminergic neurons via downregulating protease cathepsin D and upregulating neurotoxic α-synuclein. Our results also indicate that PARK19 mutation (Q789X or R927G) impairs the DNAJC6-mediated neuroprotective function.
    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
    Venetoclax和obinutuzumab正在成为慢性淋巴细胞白血病(CLL)患者的一线治疗方法。不幸的是,耐药性仍然存在,这种组合可能具有免疫抑制作用。溶酶体先前已被确定为CLL细胞中obinutuzumab细胞毒性的靶标,但机制尚不清楚。此外,研究表明,当与BTK抑制剂联合使用时,溶酶体激动剂可以在体外引起协同细胞死亡,伊布替尼,在原代CLL细胞中。这表明靶向溶酶体可能是CLL的治疗策略。在这项研究中,我们已经证明奥比妥珠单抗诱导CLL细胞中溶酶体膜透化(LMP)和组织蛋白酶D释放.组织蛋白酶的抑制减少了CLL细胞中奥比妥珠单抗诱导的细胞死亡。我们进一步确定,与维奈托克联合使用的溶菌药西拉米西汀通过增加活性氧(ROS)和组织蛋白酶的释放来增加原代CLL细胞的细胞死亡。当与维奈托克联用时,西拉美辛治疗也诱导协同细胞毒性。微环境因素IL4和CD40L或与HS-5基质细胞孵育未能显着保护CLL细胞免受西拉西汀和维奈托克诱导的凋亡。我们还发现西拉美辛治疗通过减少自体溶酶体抑制自噬。最后,自噬抑制剂氯喹未能进一步增加西拉美辛诱导的细胞死亡.一起来看,溶酶体靶向药物与维奈托克联合使用可能是克服CLL耐药性的有效策略。
    Venetoclax and obinutuzumab are becoming frontline therapies for chronic lymphocytic leukemia (CLL) patients. Unfortunately, drug resistance still occurs, and the combination could be immunosuppressive. Lysosomes have previously been identified as a target for obinutuzumab cytotoxicity in CLL cells, but the mechanism remains unclear. In addition, studies have shown that lysosomotropic agents can cause synergistic cell death in vitro when combined with the BTK inhibitor, ibrutinib, in primary CLL cells. This indicates that targeting lysosomes could be a treatment strategy for CLL. In this study, we have shown that obinutuzumab induces lysosome membrane permeabilization (LMP) and cathepsin D release in CLL cells. Inhibition of cathepsins reduced obinutuzumab-induced cell death in CLL cells. We further determined that the lysosomotropic agent siramesine in combination with venetoclax increased cell death in primary CLL cells through an increase in reactive oxygen species (ROS) and cathepsin release. Siramesine treatment also induced synergistic cytotoxicity when combined with venetoclax. Microenvironmental factors IL4 and CD40L or incubation with HS-5 stromal cells failed to significantly protect CLL cells from siramesine- and venetoclax-induced apoptosis. We also found that siramesine treatment inhibited autophagy through reduced autolysosomes. Finally, the autophagy inhibitor chloroquine failed to further increase siramesine-induced cell death. Taken together, lysosome-targeting drugs could be an effective strategy in combination with venetoclax to overcome drug resistance in CLL.
    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
    近apsilia念珠菌作为真菌感染的病原体的患病率越来越高,这突显了全面了解其毒力因素的必要性。分泌的天冬氨酸蛋白酶(Saps)在粘附事件中起重要作用,促进生物膜形成,造成组织损伤和逃避宿主的免疫反应。本研究使用各种方法调查了10个临床分离株中Sapp1和Sapp2的生产动态。每种真菌分离物都具有利用牛血清白蛋白(BSA)作为唯一氮源的能力,其在无细胞培养基中的降解证明,形成低分子量多肽。有趣的是,不同蛋白质底物的降解,如BSA,人血清白蛋白(HSA),明胶和血红蛋白,通常是孤立依赖的。值得注意的是,与BSA相比,HSA的蛋白水解更高,观察到明胶和血红蛋白。定量分析表明,肽荧光底物(组织蛋白酶D)的裂解是分离特异性的,范围从44.15到270.61FAU,平均蛋白水解为150.7FAU。通过采用特异性抗Sapp1和抗Sapp2抗体的免疫检测证实了这些真菌分离株的细胞表面上Sapp1和Sapp2抗原的存在。Sapp1的表面水平始终较高,多达四倍,与Sap2相比。同样,在真菌分泌物中检测到的Sapp1水平高于Sapp2。这项研究提供了有关Sapps在近融合梭菌中动态表达和调节的见解,强调了一种已知的毒力因子,该因子被认为是针对这种日益突出的病原体的药物开发的潜在靶标。
    真菌病原体近叶念珠菌可以分泌天冬氨酸蛋白酶(Sapps)作为其毒力因子的一部分。我们证明了萨普能够切割关键的宿主蛋白,当在浮游生物和生物膜形成细胞中生长时,Sapp1和Sapp2抗原的产生通常取决于真菌分离株。
    The increasing prevalence of Candida parapsilosis as a causative agent of fungal infections underscores the need to comprehensively understand its virulence factors. Secreted aspartic proteases (Saps) play a significant role in adhesion events, promoting biofilm formation, causing tissue damage and evading the host\'s immune response. In C. parapsilosis, three Saps have been identified: Sapp1, Sapp2 and Sapp3. The present study investigates the production dynamics of Sapp1 and Sapp2 across 10 clinical isolates of C. parapsilosis using various approaches. Each fungal isolate demonstrated the capability to utilize bovine serum albumin (BSA) as the sole nitrogen source, as evidenced by its degradation in a cell-free culture medium, forming low molecular mass polypeptides. Interestingly, the degradation of different proteinaceous substrates, such as BSA, human serum albumin (HSA), gelatin and hemoglobin, was typically isolate-dependent. Notably, higher proteolysis of HSA compared to BSA, gelatin and hemoglobin was observed. A quantitative assay revealed that the cleavage of a peptide fluorogenic substrate (cathepsin D) was isolate-specific, ranging from 44.15 to 270.61 fluorescence arbitrary units (FAU), with a mean proteolysis of 150.7 FAU. The presence of both Sapp1 and Sapp2 antigens on the cell surface of these fungal isolates was confirmed through immunological detection employing specific anti-Sapp1 and anti-Sapp2 antibodies. The surface levels of Sapp1 were consistently higher, up to fourfold, compared to Sapp2. Similarly, higher levels of Sapp1 than Sapp2 were detected in fungal secretions. This study provides insights into the dynamic expression and regulation of Sapps in C. parapsilosis, highlighting a known virulence factor that is considered a potential target for drug development against this increasingly prominent pathogen.
    The fungal pathogen Candida parapsilosis can secrete aspartic proteases (Sapps) as part of its arsenal of virulence factors. We demonstrated that Sapps were able to cleave key host proteins, and the production of Sapp1 and Sapp2 antigens was typically dependent on the fungal isolate when grown in both planktonic- and biofilm-forming cells.
    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
    Aurora激酶A(AURKA)是一种丝氨酸/苏氨酸蛋白激酶,可在神经元迁移和神经突形成过程中调节微管组织。在阿尔茨海默病(AD)脑样本中发现AURKA活性降低,但对AURKA在AD发病机制中的作用知之甚少。这里,我们证明AURKA在原代培养的大鼠神经元中表达,来自成年小鼠大脑的神经元,和死后人类AD大脑中的神经元。AURKA磷酸化,与它的活动呈正相关,在人类AD大脑中减少。在SH-SY5Y细胞中,AURKA的药理激活增加了AURKA磷酸化,酸化的内溶酶体,降低了淀粉样β蛋白(Aβ)生成酶β位点淀粉样前体蛋白裂解酶(BACE-1)的活性,增加Aβ降解酶组织蛋白酶D的活性,并降低Aβ的细胞内和分泌水平。相反,AURKA的药理学抑制降低了AURKA磷酸化,去酸化的内溶酶体,降低组织蛋白酶D的活性,细胞内和分泌的Aβ水平增加。因此,AD中AURKA活性的降低可能有助于Aβ的神经元内积累和细胞外淀粉样斑块的形成。
    Aurora kinase A (AURKA) is a serine/threonine-protein kinase that regulates microtubule organization during neuron migration and neurite formation. Decreased activity of AURKA was found in Alzheimer\'s disease (AD) brain samples, but little is known about the role of AURKA in AD pathogenesis. Here, we demonstrate that AURKA is expressed in primary cultured rat neurons, neurons from adult mouse brains, and neurons in postmortem human AD brains. AURKA phosphorylation, which positively correlates with its activity, is reduced in human AD brains. In SH-SY5Y cells, pharmacological activation of AURKA increased AURKA phosphorylation, acidified endolysosomes, decreased the activity of amyloid beta protein (Aβ) generating enzyme β-site amyloid precursor protein cleaving enzyme (BACE-1), increased the activity of the Aβ degrading enzyme cathepsin D, and decreased the intracellular and secreted levels of Aβ. Conversely, pharmacological inhibition of AURKA decreased AURKA phosphorylation, de-acidified endolysosomes, decreased the activity of cathepsin D, and increased intracellular and secreted levels of Aβ. Thus, reduced AURKA activity in AD may contribute to the development of intraneuronal accumulations of Aβ and extracellular amyloid plaque formation.
    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
    骨关节炎(OA)是普遍存在的退行性关节疾病,尽管它被广泛观察到,其确切原因尚不完全清楚。该研究的主要重点是评估补体C3和组织蛋白酶D在膝骨关节炎(OA)发展中的作用。这是最常见的退行性关节病。
    这项研究是在20名膝关节OA患者和20名健康对照组中进行的。OA膝盖(II/III级,放射学Kellgren和Lawrence(K/L)分类),年龄在40至65岁之间的人能够膝盖疼痛地行走。该研究还包括年龄匹配的健康对照。测定血清中补体C3和组织蛋白酶D的浓度。
    本研究的结果证明,与健康老年人匹配的对照组相比,OA患者中C3和组织蛋白酶D的浓度显著(P<0.001)更高。
    分析显示炎症标志物,补体C3以及组织蛋白酶D可用作膝OA的诊断标志物。观察结果表明,补体系统的激活主要影响关节内的过程,而C3似乎在产生全身性炎症反应中起核心作用。
    UNASSIGNED: Osteoarthritis (OA) stands as the prevailing degenerative joint condition, and although it is widely observed, its precise causes are not fully understood. The main focus of the study was to assess the role of Complement C3 and Cathepsin D in the development of knee osteoarthritis (OA), which is the most prevalent degenerative joint disease.
    UNASSIGNED: The study was carried out in 20 patients with knee OA and 20 healthy control group. OA knee (Grade II/III, Radiological Kellgren and Lawrence (K/L) classification), aged between 40 and 65 years were able to walk with a painful knee. The study also included healthy age-matched controls. The concentration of Complement C3 and Cathepsin D in serum was determined.
    UNASSIGNED: The results of the present study demonstrated significantly (P < 0.001) higher concentrations of C3 and Cathepsin D in OA patients in comparison to that of the healthy aged matched control group.
    UNASSIGNED: The analysis showed that inflammatory markers, Complement C3 as well as Cathepsin D may be used as diagnostic markers of knee OA. The observations suggest that the activation of the complement system mainly affects processes within the joints, while C3 appears to play a central role in generating a systemic inflammatory response.
    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
    隐性Stargardt病(STGD1)是由ABCA4基因突变引起的遗传性青少年黄斑病,没有合适的治疗方法。视网膜色素上皮(RPE)中功能性ABCA4的丢失,没有感光细胞的贡献,显示诱导STGD1病理学。这里,我们鉴定了组织蛋白酶D(CatD),原发性RPE溶酶体蛋白酶,作为导致STGD1内溶酶体功能障碍的关键分子参与者,使用新开发的“培养皿中疾病”RPE模型从证实的STGD1患者中获得。来自三名STGD1患者的诱导多能干细胞(iPSC)衍生的RPE表现出溶酶体pH升高,如先前在Abca4-/-小鼠中报道的。来自STGD1患者和Abca4-/-小鼠的RPE中CatD蛋白成熟和活性受损。因此,STGD1RPE细胞有减少的光感受器外节降解和α-突触核蛋白的异常积累,CatD的天然底物。此外,STGD1RPE细胞中功能失调的ABCA4导致自发荧光材料和磷脂酰乙醇胺(PE)的细胞内积累。与STGD1RPE细胞内膜相关的PE分布改变可能会损害LC3相关的吞噬作用,有助于延迟内溶酶体降解活性。STGD1的RPE中的溶酶体的药物介导的再酸化可恢复CatD功能活性并减少未成熟CatD蛋白负荷的积累。该临床前研究验证了CatD缺陷对STGD1病理学的贡献,并为靶向RPE细胞的有效治疗方法提供了证据。我们的发现支持细胞自主RPE驱动的病理学,未来的研究旨在靶向RPE细胞治疗ABCA4介导的视网膜病变。
    Recessive Stargardt disease (STGD1) is an inherited juvenile maculopathy caused by mutations in the ABCA4 gene, for which there is no suitable treatment. Loss of functional ABCA4 in the retinal pigment epithelium (RPE) alone, without contribution from photoreceptor cells, was shown to induce STGD1 pathology. Here, we identified cathepsin D (CatD), the primary RPE lysosomal protease, as a key molecular player contributing to endo-lysosomal dysfunction in STGD1 using a newly developed \"disease-in-a-dish\" RPE model from confirmed STGD1 patients. Induced pluripotent stem cell (iPSC)-derived RPE originating from three STGD1 patients exhibited elevated lysosomal pH, as previously reported in Abca4-/- mice. CatD protein maturation and activity were impaired in RPE from STGD1 patients and Abca4-/- mice. Consequently, STGD1 RPE cells have reduced photoreceptor outer segment degradation and abnormal accumulation of α-synuclein, the natural substrate of CatD. Furthermore, dysfunctional ABCA4 in STGD1 RPE cells results in intracellular accumulation of autofluorescent material and phosphatidylethanolamine (PE). The altered distribution of PE associated with the internal membranes of STGD1 RPE cells presumably compromises LC3-associated phagocytosis, contributing to delayed endo-lysosomal degradation activity. Drug-mediated re-acidification of lysosomes in the RPE of STGD1 restores CatD functional activity and reduces the accumulation of immature CatD protein loads. This preclinical study validates the contribution of CatD deficiencies to STGD1 pathology and provides evidence for an efficacious therapeutic approach targeting RPE cells. Our findings support a cell-autonomous RPE-driven pathology, informing future research aimed at targeting RPE cells to treat ABCA4-mediated retinopathies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号