legumain

Legumain
  • 文章类型: Journal Article
    靶向放射性核素治疗(TRT)是一种有效的肿瘤治疗方法。自缩合策略可以增强放射性核素在肿瘤中的保留并增强抗肿瘤效果。考虑到legumain在几种类型的人类癌症中过度表达,我们报道了一种131I标记的放射性药物([131I]MAAN),其基于2-氰基苯并噻唑(CBT)和半胱氨酸(Cys)的自缩合反应,用于体内治疗豆科蛋白酶过表达的肿瘤.然而,肝脏富集限制了它的应用。在这项研究中,通过将亲水肽序列His-Glu-His-Glu-His-Glu-Glu-Glu((HE)3)引入[131I]MAAN以优化药代动力学,合成了一种新的放射性药物[131I]IM(HE)3AAN.在还原环境下被生肉激活后,亲水性[131I]IM(HE)3AAN可以与其前体反应形成高度疏水的异源二聚体([131I]H-二聚体)。切伦科夫成像显示,与[131I]MAAN相比,[131I]IM(HE)3AAN显示出更高的肿瘤选择性和更长的肿瘤保留时间,肝脏摄取显着减少。在用[131I]IM(HE)3AAN治疗18天后,肿瘤增殖受到明显抑制,治疗过程中正常器官未见明显损伤。这些发现表明[131I]IM(HE)3AAN成为治疗豆科蛋白过表达肿瘤的有希望的候选者。
    Targeted radionuclide therapy (TRT) is an effective treatment for tumors. Self-condensation strategies can enhance the retention of radionuclides in tumors and enhance the anti-tumor effect. Considering legumain is overexpressed in several types of human cancers, we have reported a 131I-labeled radiopharmaceutical ([131I]MAAN) based on the self-condensation reaction between 2-cyanobenzothiazole (CBT) and cysteine (Cys) for treatment of legumain-overexpressed tumors in vivo. However, liver enrichment limits its application. In this study, a new radiopharmaceutical [131I]IM(HE)3AAN was synthesized by introducing a hydrophilic peptide sequence His-Glu-His-Glu-His-Glu ((HE)3) into [131I]MAAN to optimize the pharmacokinetics. Upon activation by legumain under a reducing environment, hydrophilic [131I]IM(HE)3AAN could react with its precursor to form heterologous dimer ([131I]H-Dimer) that is highly hydrophobic. Cerenkov imaging reveals that [131I]IM(HE)3AAN displayed superior tumor selectivity and longer tumor retention time as compared with [131I]MAAN, with a significant reduction in liver uptake. After an 18-day treatment with [131I]IM(HE)3AAN, the tumor proliferation was obviously inhibited, while no obvious injury was observed in the normal organs during treatment. These findings suggest [131I]IM(HE)3AAN emerges as a promising candidate for treatment of legumain-overexpressed tumors.
    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
    豆球蛋白是阿霉素和豆科菌素可裂解的肽接头的新型缀合物。已经开发了它来改善阿霉素的副作用。荷瘤小鼠的生物分布,急性耐受,并评估了豆类素对Sprague-Dawley大鼠和比格犬的潜在全身毒性作用。豆类蛋白进入循环后主要作为蛋白质复合物存在于血浆中。与小鼠等摩尔剂量的常规阿霉素相比,我们发现肿瘤中阿霉素的暴露量较高(约1.7倍增加),而正常组织中的暴露量较低(约3.26-,3.46-,心脏减少1.29倍,肾,和等离子体,分别)静脉注射豆类素后的荷瘤小鼠。在雌性大鼠中,豆类霉素的急性最大耐受剂量(MTD)>16mg/kg阿霉素当量,11mg/kg阿霉素在雄性大鼠中的当量(常规阿霉素的LD50为10.51mg/kg),和>8mg/kg多柔比星在狗中的当量(常规多柔比星的MTD为1.5mg/kg)。在大鼠(每周一次5、10和25mg/kg/剂)和狗(每周一次3/1.5、10/5和20/10mg/kg/剂)中进行了为期四周的静脉内豆球蛋白重复剂量毒性研究;由于不可耐受的豆球蛋白相关毒性为20mg/kg,因此剂量水平从第二剂量降低。毒性的主要器官包括胃肠道,淋巴和造血器官,肾,皮肤,肝脏,生殖器官,和周围神经,都与阿霉素有关.然而,仅在MTD剂量水平下观察到心脏毒性.总之,我们的研究结果证实,与常规阿霉素相比,豆霉素的安全性得到了改善,并支持了其在治疗癌症方面的临床获益.
    Legubicin is a novel conjugate of doxorubicin and a legumain-cleavable peptide linker. It has been developed to ameliorate the side effects of doxorubicin. Biodistribution in tumor-bearing mice, acute tolerance, and potential systemic toxic effects in Sprague-Dawley rats and beagle dogs of legubicin were assessed. Legubicin exists mainly as a protein complex in plasma after entering the circulation. Compared with conventional doxorubicin at an equal molar dose in mice, we found higher exposure to doxorubicin in tumor (approximately 1.7-fold increase) while lower exposure in normal tissues (an ~3.26-, 3.46-, and 1.29-fold reduction in heart, kidney, and plasma, respectively) in tumor-bearing mice after intravenous injection of legubicin. The acute maximum tolerance dose (MTD) of legubicin was >16 mg/kg doxorubicin equivalent in female rats, 11 mg/kg doxorubicin equivalent in male rats (LD50 of conventional doxorubicin is 10.51 mg/kg), and >8 mg/kg doxorubicin equivalent in dogs (MTD of conventional doxorubicin is 1.5 mg/kg). Four-week repeat-dose toxicity studies of intravenous legubicin were conducted in rats (5, 10, and 25 mg/kg/dose once weekly) and dogs (3/1.5, 10/5, and 20/10 mg/kg/dose once weekly); the dose levels were reduced from the second dose due to intolerable legubicin-associated toxicity at 20 mg/kg. Major organs of toxicity included the gastrointestinal tract, lymphoid and hematopoietic organs, kidney, skin, liver, reproductive organs, and peripheral nerves, which are all associated with doxorubicin. However, cardiotoxicity was only noted at MTD dose levels. Altogether, our results confirm an improved safety profile of legubicin over conventional doxorubicin and support its clinical benefit for treating 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
    Legubicin,一种基于阿霉素的新型前药,同时具有白蛋白结合和豆蔻素激活特性。本研究的目的是开发和验证一种UHPLC-MS/MS方法,用于研究静脉给药后大鼠和荷瘤小鼠体内豆球蛋白的体内药代动力学和组织分布特征,并将该前药与阳性对照药物阿霉素进行比较。该研究采用UHLC-MS/MS方法来确定血浆中白蛋白结合的豆球蛋白和两种代谢物(游离的Leu-DOX和DOX)的水平。肿瘤,和组织样本.该方法具有良好的选择性,高灵敏度,优良的提取回收率,运行时间短。结果表明,豆球蛋白主要以蛋白质结合形式存在于体内循环中,AUC值较大,清除率和分布较低,基本上释放出少量的阿霉素.与等摩尔剂量的阿霉素相比,legubicin显示肿瘤中活性药物的暴露增加,心脏和肾脏中活性药物的水平降低。这项研究提供了有价值的信息的药代动力学和组织分布,暗示其作为抗癌治疗的新型有效候选药物的潜力。
    Legubicin, a novel prodrug based on doxorubicin, has both albumin-binding and legumain-activating properties. The aim of this study was to develop and validate a UHPLC-MS/MS method for investigating the in vivo pharmacokinetics and tissue distribution profiles of legubicin in rats and tumor-bearing mice following intravenous administration, and to compare this prodrug with the positive control drug doxorubicin. The study employed a UHLC-MS/MS method to determine the levels of albumin-bound of legubicin and two metabolites (free Leu-DOX and DOX) in plasma, tumor, and tissue samples. This method was validated for good selectivity, high sensitivity, excellent extraction recovery, and short run time. The results showed that legubicin was present in the circulation in vivo mainly in a protein-bound form with larger AUC values and lower clearance and distribution, and essentially released small amounts of doxorubicin. Compared to administration of equimolar doses of doxorubicin, legubicin showed increased exposure of the active drug in the tumor and decreased the level of the active drug in the heart and kidney. This study provides valuable information on the pharmacokinetics and tissue distribution of legubicin, implicating its potential as a novel and effective drug candidate for anti-cancer 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
    Legumain在多种肿瘤中过度表达,作为一个重要的肿瘤生物标志物。我们的研究旨在开发一种新的正电子发射断层扫描(PET)探针[68Ga]Ga-NOTA-SF-AANM,用于在体内对豆球蛋白的表达水平进行成像。[68Ga]Ga-NOTA-SF-AANM的放射性标记在15分钟内完成。该探针具有良好的体外稳定性。NOTA-SF-AANM表现出对重组人豆蔻酶的快速反应,使分子内缩合环化。细胞摄取和溶酶体共定位实验表明,该探针能够特异性区分MDA-MB-468和PC-3癌细胞,并具有不同程度的豆蔻素表达。PET成像在MDA-MB-468肿瘤中显示出明显且持续的信号(60分钟时,为3.59±0.30%ID/mL),而PC-3肿瘤表现出较低的放射性(60分钟时1.08±0.35%ID/mL),进一步验证[68Ga]Ga-NOTA-SF-AANM对豆类的特异性靶向。[68Ga]Ga-NOTA-SF-AANM由于其在放射性标记和准确监测豆科蛋白表达水平方面的优势,是用于精确诊断豆科蛋白相关疾病的有前途的工具。
    Legumain is overexpressed in diverse tumors, serving as a significant tumor biomarker. Our study aimed to develop a new positron emission tomography (PET) probe [68Ga]Ga-NOTA-SF-AANM for imaging the expression level of legumain in vivo. The radio-labeling of [68Ga]Ga-NOTA-SF-AANM was accomplished within 15 min. The probe has good stability in vitro. NOTA-SF-AANM exhibited rapid response to recombinant human legumain enzyme, enabling intramolecular condensation cyclization. Cellular uptake and lysosomal co-localization experiments demonstrated that the probe was able to differentiate specifically between MDA-MB-468 and PC-3 cancer cells with varying degrees of legumain expression. PET imaging displayed a significant and persistent signal (3.59 ± 0.30 %ID/mL at 60 min) in MDA-MB-468 tumors, while PC-3 tumors exhibited lower radioactivity (1.08 ± 0.35 %ID/mL at 60 min), further validating the specific targeting of [68Ga]Ga-NOTA-SF-AANM towards legumain. [68Ga]Ga-NOTA-SF-AANM is a promising tool for precise diagnosis of legumain-related diseases due to its advantages in radio-labeling and accurate monitoring of legumain expression levels.
    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
    莱古曼,溶酶体半胱氨酸蛋白酶在多种肿瘤中过度表达,已被认为是各种癌症的有希望的生物标志物。精确检测溶酶体中的豆蔻素活性代表了肿瘤早期诊断和预后的重要策略。具有靶向自组装特性的小分子探针在分子成像中具有巨大的潜力。在这项研究中,我们报道了两种双靶向放射性示踪剂([18F]SF-AAN-M和[18F]SF-AAN-HEM),其具有用于正电子发射断层扫描(PET)成像的legumain介导的自组装特性.两种放射性示踪剂均以高标记产率(>50%)合成,并且通过一步直接的18F-标记,放射化学纯度超过99%。两种示踪剂均被还原剂和legumain有效激活,以自组装成聚集体,并在legumain过表达的MDA-MB-468细胞和肿瘤中显示出增强的保留,这表明溶酶体靶向吗啉的引入增加了肿瘤的摄取,并延长了在过表达的肿瘤中放射性示踪剂的保留。此外,[18F]SF-AAN-HEM与亲水(组氨酸-谷氨酸)3标签显示肝脏摄取显著降低,肿瘤摄取没有明显减少,提供高信噪比(肿瘤/肝脏和肿瘤/肌肉)。所有这些结果表明,双靶向示踪剂[18F]SF-AAN-HEM可以为体内监测肿瘤中的豆球蛋白活性提供有希望的工具。
    Legumain, a lysosomal cysteine protease overexpressed in a variety of tumors, has been considered a promising biomarker for various cancers. Precise detection of legumain activity in the lysosome represents an important strategy for early diagnosis and prognosis of tumors. Small-molecule probes with the property of target-enabled self-assembly hold great potential for molecular imaging. In this study, we reported two dual-targeting radiotracers ([18F]SF-AAN-M and [18F]SF-AAN-HEM) with a property of legumain-mediated self-assembly for positron emission tomography (PET) imaging. Both the radiotracers were synthesized with high labeling yield (>50%) and the radiochemical purity was over 99% via one-step straightforward 18F-labeling. Both tracers were efficiently activated by the reducing agent and legumain to self-assemble into aggregates and showed enhanced retention in legumain-overexpressed MDA-MB-468 cells and tumors, indicating that the introduction of lysosome-targeting morpholine increased the tumor uptake and extended the retention of radiotracers in legumain-overexpressed tumors. In addition, [18F]SF-AAN-HEM with a hydrophilic (histidine-glutamate)3 tag displayed significantly reduced liver uptake with no conspicuous reduction in tumor uptake, affording high signal-to-noise ratios (tumor/liver and tumor/muscle). All of these results suggest that dual-targeting tracer [18F]SF-AAN-HEM could provide a promising tool for in vivo monitoring legumain activity in tumors.
    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: Review
    动脉粥样硬化是一种脂质驱动的慢性炎性疾病,对健康构成严重威胁。Legumain(LGMN),也被称为天冬酰胺核酸内切酶,是一种新型的半胱氨酸蛋白酶,可以特异性水解含有天冬酰胺残基的底物分子。它在哺乳动物中具有抗凋亡作用,并在炎症反应中起抗原呈递作用。多项研究发现LGMN可激活多条信号通路促进细胞凋亡和迁移,炎症反应,和动脉粥样硬化的发展。重要的是,LGMN通过参与动脉粥样硬化的多种病理生理机制发挥促动脉粥样硬化作用。包括血管重塑,炎症反应,斑块稳定性,和细胞外基质的降解。在本次审查中,我们描述了LGMN分布,结构,代,和功能合作伙伴。此外,我们总结了LGMN与动脉粥样硬化的关系。基于LGMN与动脉粥样硬化的关系,LGMN可能是动脉粥样硬化的潜在生物标志物。
    Atherosclerosis is a lipid-driven chronic inflammatory disease that poses a serious threat to health. Legumain (LGMN), also known as asparagine endonuclease, is a new type of cysteine proteases that can specifically hydrolyze substrate molecules containing asparagine residues. It has anti-apoptotic effects in mammals and plays an antigen-presenting role in inflammatory response. Several studies have found that LGMN can activate multiple signal pathways to promote cell apoptosis and migration, inflammatory response, and the development of atherosclerosis. Importantly, LGMN exerts pro-atherogenic effects by participating in a variety of pathophysiological mechanisms of atherosclerosis, including vascular remodeling, inflammatory response, plaque stability, and the degradation of extracellular matrix. In the present review, we describe the LGMN distribution, structure, generation, and functional partners. Furthermore, we summarize the relationship between LGMN and atherosclerosis. Based on the relationship between LGMN and atherosclerosis, LGMN may be a potential biomarker for atherosclerosis.
    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
    乳酸盐由于活跃的糖酵解(又名Warburg效应)而在癌组织中丰富,并且介导肿瘤细胞与免疫微环境(TIME)之间的串扰以促进乳腺癌的进展。槲皮素(QU)是一种有效的单羧酸转运蛋白(MCT)抑制剂,可以减少肿瘤细胞的乳酸产生和分泌。多柔比星(DOX)可以诱导免疫原性细胞死亡(ICD),促进肿瘤特异性免疫激活。因此,我们提出了QU和DOX的联合治疗来抑制乳酸代谢和刺激抗肿瘤免疫。为了提高肿瘤靶向效率,我们开发了一种对KC26肽进行修饰的可激活脂质体系统(KC26-Lipo),用于共同递送QU和DOX,以调节乳腺癌的肿瘤代谢和时间.KC26肽是一种豆类反应性,发夹结构的细胞穿透肽(聚精氨酸)衍生物。Legumain是一种在乳腺肿瘤中过度表达的蛋白酶,允许选择性激活KC26-Lipo以随后促进肿瘤内和细胞内渗透。KC26-Lipo通过化疗和抗肿瘤免疫有效抑制4T1乳腺癌肿瘤的生长。此外,乳酸代谢的抑制抑制HIF-1α/VEGF通路和血管生成,并使肿瘤相关巨噬细胞(TAMs)复极化.这项工作通过调节乳酸代谢和时间提供了一种有前途的乳腺癌治疗策略。
    Lactate is abundant in cancer tissues due to active glycolysis (aka Warburg effect) and mediates crosstalk between tumor cells and the immune microenvironment (TIME) to promote the progression of breast cancer. Quercetin (QU) is a potent monocarboxylate transporters (MCT) inhibitor, which can reduce lactate production and secretion of tumor cells. Doxorubicin (DOX) can induce immunogenic cell death (ICD), which promotes tumor-specific immune activation. Thus, we propose a combination therapy of QU&DOX to inhibit lactate metabolism and stimulate anti-tumor immunity. To enhance tumor-targeting efficiency, we developed a legumain-activatable liposome system (KC26-Lipo) with modification of KC26 peptide for co-delivery of QU&DOX for modulation of tumor metabolism and TIME in breast cancer. The KC26 peptide is a legumain-responsive, hairpin-structured cell-penetrating peptide (polyarginine) derivative. Legumain is a protease overexpressed in breast tumors, allowing selective activation of the KC26-Lipo to subsequently facilitate intra-tumoral and intracellular penetration. The KC26-Lipo effectively inhibited 4T1 breast cancer tumor growth through chemotherapy and anti-tumor immunity. Besides, inhibition of lactate metabolism suppressed the HIF-1α/VEGF pathway and angiogenesis and repolarized the tumor-associated macrophages (TAM). This work provides a promising breast cancer therapy strategy by regulating lactate metabolism and TIME.
    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
    干细胞为基础的治疗是非常有前途的阿尔茨海默病(AD),但还没有成为现实。一个关键的挑战是移植微环境,影响干细胞的治疗效果。在AD大脑中,淀粉样β(Aβ)肽和炎症细胞因子持续毒害组织微环境,导致移植细胞的低存活率和有限的功效。有必要为移植细胞创建支持生长的微环境。AD研究的最新进展表明,天冬酰胺酰内肽酶(AEP)是修饰病理变化的潜在干预靶标。我们在这里选择APP/PS1小鼠作为AD模型,并采用AEP的药理学抑制一个月来改善大脑微环境。此后,我们将神经干细胞(NSC)移植到海马中,并维持治疗一个月。我们发现AEPs的抑制导致Aβ的显著降低,TNF-α,他们大脑中的IL-6和IL-1β。在仅接受NSC移植的AD小鼠中,神经干细胞的存活率很低,而与AEP抑制预处理组合,移植细胞的存活率加倍。在2个月的治疗期内,神经干细胞的植入和AEP的预抑制显著增强了海马的神经可塑性并挽救了认知障碍。单独的NSC移植或单独的AEP抑制均未达到显著功效。总之,药物抑制AEP改善AD小鼠的大脑微环境,从而提高了移植干细胞的存活率和治疗效果。
    Stem-cell-based therapy is very promising for Alzheimer\'s disease (AD), yet has not become a reality. A critical challenge is the transplantation microenvironment, which impacts the therapeutic effect of stem cells. In AD brains, amyloid-beta (Aβ) peptides and inflammatory cytokines continuously poison the tissue microenvironment, leading to low survival of grafted cells and restricted efficacy. It is necessary to create a growth-supporting microenvironment for transplanted cells. Recent advances in AD studies suggest that the asparaginyl endopeptidase (AEP) is a potential intervention target for modifying pathological changes. We here chose APP/PS1 mice as an AD model and employed pharmacological inhibition of the AEP for one month to improve the brain microenvironment. Thereafter, we transplanted neural stem cells (NSCs) into the hippocampus and maintained therapy for one more month. We found that inhibition of AEPs resulted in a significant decrease of Aβ, TNF-α, IL-6 and IL-1β in their brains. In AD mice receiving NSC transplantation alone, the survival of NSCs was at a low level, while in combination with AEP inhibition pre-treatment the survival rate of engrafted cells was doubled. Within the 2-month treatment period, implantation of NSCs plus pre-inhibition of the AEP significantly enhanced neural plasticity of the hippocampus and rescued cognitive impairment. Neither NSC transplantation alone nor AEP inhibition alone achieved significant efficacy. In conclusion, pharmacological inhibition of the AEP ameliorated brain microenvironment of AD mice, and thus improved the survival and therapeutic efficacy of transplanted stem cells.
    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
    Legumain(LGMN)已被证明不仅在乳房中过度表达,前列腺,和肝脏肿瘤细胞,而且在构成肿瘤微环境的巨噬细胞中。这支持LGMN是调节肿瘤发展的关键蛋白的观点。入侵,和传播。用siRNA或化疗药物和肽靶向LGMN可以抑制培养物中的癌细胞增殖并减少体内肿瘤生长。此外,由于与肿瘤或肿瘤相关巨噬细胞(TAMs)相比,其在正常细胞中的表达显着降低,因此可以用作癌症检测和靶向的标志物。肿瘤形成受蛋白质异常表达和细胞结构改变的影响。但肿瘤微环境是一个至关重要的决定因素。Legumain(LGMN)是一种体内活性半胱氨酸蛋白酶,可催化许多蛋白质的降解。其精确的生物学机制包括许多途径,包括对肿瘤微环境中肿瘤相关巨噬细胞和新生血管内皮的影响。这项工作的目的是通过回顾LGMN在肿瘤发生和发展中的功能及其与肿瘤环境的关系,为深入研究LGMN在肿瘤微环境中的功能并发现新的肿瘤早期诊断标志物和治疗靶标奠定基础。
    Legumain (LGMN) has been demonstrated to be overexpressed not just in breast, prostatic, and liver tumor cells, but also in the macrophages that compose the tumor microenvironment. This supports the idea that LGMN is a pivotal protein in regulating tumor development, invasion, and dissemination. Targeting LGMN with siRNA or chemotherapeutic medicines and peptides can suppress cancer cell proliferation in culture and reduce tumor growth in vivo. Furthermore, legumain can be used as a marker for cancer detection and targeting due to its expression being significantly lower in normal cells compared to tumors or tumor-associated macrophages (TAMs). Tumor formation is influenced by aberrant expression of proteins and alterations in cellular architecture, but the tumor microenvironment is a crucial deciding factor. Legumain (LGMN) is an in vivo-active cysteine protease that catalyzes the degradation of numerous proteins. Its precise biological mechanism encompasses a number of routes, including effects on tumor-associated macrophage and neovascular endothelium in the tumor microenvironment. The purpose of this work is to establish a rationale for thoroughly investigating the function of LGMN in the tumor microenvironment and discovering novel tumor early diagnosis markers and therapeutic targets by reviewing the function of LGMN in tumor genesis and progression and its relationship with tumor milieu.
    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
    介导tau在AD中聚集和传播的分子机制尚不清楚。这里,我们显示,肌动蛋白结合蛋白cofilin被半胱氨酸蛋白酶天冬酰胺内肽酶(AEP)在AD患者的大脑中N138处裂解。AEP生成的cofilin1-138片段与tau相互作用并促进其聚集。由cofilin1-138和tau组成的混合原纤维比纯tau原纤维对细胞更具致病性。此外,脑中cofilin1-138的过表达促进病理性tau聚集体的繁殖,并促进tauP301S小鼠的AD样认知障碍。然而,与注射编码野生型cofilin的AAV的小鼠相比,用编码AEP-不可切割的cofilin突变体的腺相关病毒(AAV)感染的小鼠显示出减弱的tau病理学和认知障碍。一起,这些观察结果支持cofilin1-138片段在AD发作和进展期间tau病理的聚集和传播中的作用.
    The molecular mechanisms mediating the aggregation and transmission of tau in AD remain unclear. Here, we show that the actin-binding protein cofilin is cleaved by a cysteine protease asparagine endopeptidase (AEP) at N138 in the brains of patients with AD. The AEP-generated cofilin 1-138 fragment interacts with tau and promotes its aggregation. The mixed fibrils consisting of cofilin 1-138 and tau are more pathogenic to cells than pure tau fibrils. Furthermore, overexpression of cofilin 1-138 in the brain facilitates the propagation of pathological tau aggregates and promotes AD-like cognitive impairments in tau P301S mice. However, mice infected with adeno-associated viruses (AAVs) encoding an AEP-uncleavable cofilin mutant show attenuated tau pathology and cognitive impairments compared with mice injected with AAVs encoding wild-type cofilin. Together, these observations support the role of the cofilin 1-138 fragment in the aggregation and transmission of tau pathology during the onset and progression of AD.
    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