HSPB8

HSPB8
  • 文章类型: Journal Article
    肝内胆管癌(ICC)的发病率稳步上升,它与高死亡率有关。收集临床标本检测ICC组织中HSPB8和BAG3的表达。培养ICC细胞并用过表达或沉默特定基因的质粒转染,以研究基因表达改变对细胞功能的影响。使用qPCR和Western印迹技术来测量基因和蛋白质表达水平。进行伤口愈合测定以评估细胞迁移能力。Transwell测定用于评估细胞侵袭能力。Co-IP用于验证HSPB8和BAG3之间的结合关系。经由过程构建肿瘤转移模子,验证了HSPB8和BAG3对肿瘤体内肺转移的影响。通过以上实验,我们发现在ICC组织和细胞中HSPB8和BAG3的表达上调,它们的表达呈正相关。通过上调或下调BAG3的表达可以促进或抑制ICC细胞的转移能力。此外,HSPB8-BAG3伴侣复合物通过激活自噬导致丝状蛋白A的异常降解。丝状蛋白A的表达增加抑制ICC细胞的迁移和侵袭。体内过表达HSPB8和BAG3促进ICC细胞的肺转移能力。HSPB8-BAG3伴侣复合物通过调节CASA介导的丝状蛋白A降解促进ICC细胞迁移和侵袭,为加强ICC治疗策略提供见解。
    The incidence of intrahepatic cholangiocarcinoma (ICC) is steadily rising, and it is associated with a high mortality rate. Clinical samples were collected to detect the expression of HSPB8 and BAG3 in ICC tissues. ICC cells were cultured and transfected with plasmids that overexpressed or silenced specific genes to investigate the impact of gene expression alterations on cell function. qPCR and Western blot techniques were utilized to measure gene and protein expression levels. A wound healing assay was conducted to assess cell migration ability. The Transwell assay was used to assess cell invasion ability. Co-IP was used to verify the binding relationship between HSPB8 and BAG3. The effects of HSPB8 and BAG3 on lung metastasis of tumors in vivo were verified by constructing a metastatic tumor model. Through the above experiments, we discovered that the expressions of HSPB8 and BAG3 were up-regulated in ICC tissues and cells, and their expressions were positively correlated. The metastatic ability of ICC cells could be promoted or inhibited by upregulating or downregulating the expression of BAG3. Furthermore, the HSPB8-BAG3 chaperone complex resulted in the abnormal degradation of Filamin A by activating autophagy. Increased expression of Filamin A inhibits the migration and invasion of ICC cells. Overexpression of HSPB8 and BAG3 in vivo promoted the lung metastasis ability of ICC cells. The HSPB8-BAG3 chaperone complex promotes ICC cell migration and invasion by regulating CASA-mediated degradation of Filamin A, offering insights for enhancing ICC therapeutic strategies.
    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
    过度的氧化应激引发脑血管和神经退行性疾病,导致急性和慢性脑损伤。然而,潜在的机制仍然未知。小热休克蛋白B8(HSPB8)的水平,在大脑中高度表达,已知在脑损伤模型中显著升高。外源性HSPB8保护大脑免受线粒体损伤。这种保护的潜在机制是HSPB8过表达减轻线粒体依赖性凋亡途径;线粒体生物发生,裂变,和线粒体自噬。因此,HSPB8的过表达可能具有作为脑血管和神经退行性疾病的临床治疗潜力。本文综述了HSPB8对脑过度氧化应激保护作用的研究进展。包括线粒体功能障碍和有效信号通路的调节。
    Excessive oxidative stress triggers cerebrovascular and neurodegenerative diseases resulting in acute and chronic brain injury. However, the underlying mechanisms remain unknown. Levels of small heat shock protein B8 (HSPB8), which is highly expressed in the brain, are known to be significantly elevated in cerebral injury models. Exogenous HSPB8 protects the brain against mitochondrial damage. One potential mechanism underlying this protection is that HSPB8 overexpression alleviates the mitochondria-dependent pathways of apoptosis; mitochondrial biogenesis, fission, and mitophagy. Overexpression of HSPB8 may therefore have potential as a clinical therapy for cerebrovascular and neurodegenerative diseases. This review provides an overview of advances in the protective effects of HSPB8 against excessive cerebral oxidative stress, including the modulation of mitochondrial dysfunction and potent signaling pathways.
    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
    HSPB8是属于称为HSPB的ATP非依赖性应激蛋白家族的热休克蛋白,其广泛存在于各种生物体的细胞中。他们致力于蛋白质质量控制(PQC),努力避免蛋白质聚集,并创造一个可以快速折叠的非天然蛋白质库。它们的基本表达或胁迫诱导性受到位于HSPB调节区中的各种顺式元件的调节。在目前的研究中,我们已经预测并证实了肝脏中HSPB8基因的两个可变剪接的新转录本,大脑,和心脏。这些剪接变体由于较小的N末端区域而具有较小的尺寸,并且在它们的细胞定位中显示出显著的变化。预测新的同种型(HSPB8-N1)主要位于核区域,而报道的同种型(HSPB8)和一种新的同种型(HSPB8-N2)被预测为本质上是细胞质。在新同种型的磷酸化位点中也观察到许多变化。新报道的同种型缺乏对HSPB8蛋白的各种功能努力至关重要的几种结构基序。使用各种生物信息学工具对概念上翻译的蛋白质进行计算机模拟分析,以了解其特性,从而探索其在治疗中的可能潜力。
    HSPB8 is a heat shock protein belonging to a family of ATP-independent stress proteins called HSPB which are present far and wide in the cells of various organisms. They are committed to protein quality control (PQC) and strive to avert protein aggregation and to procreate a pool of non-native proteins that can be swiftly folded. Their fundamental expression or stress inducibility is regulated by various cis-elements localized in the HSPB regulatory regions. In the current study we have predicted and confirmed two alternatively spliced novel transcripts of HSPB8 gene in liver, brain, and heart. These spliced variants have smaller sizes owing to smaller N terminal regions and showed remarkable changes in their cellular localization. Novel isoform (HSPB8-N1) was predicted to be majorly localized to nuclear region while the reported isoform (HSPB8) and one of the novel isoforms (HSPB8-N2) were predicted to be cytoplasmic in nature. There were many changes observed in the phosphorylation sites of the novel isoforms as well. The newly reported isoforms lack several structural motifs that are essential for various functional endeavors of the HSPB8 protein. In silico analysis of the conceptually translated protein was carried out using various bioinformatics tools to gain an understanding of their properties in order to explore their possible potential in therapeutics.
    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
    子痫前期(PE)是一种发病机制复杂的妊娠特异性综合征。本研究旨在使用缺氧/复氧(H/R)处理的HTR8/SVneo细胞模型探讨热休克蛋白B8(HSPB8)和c-Myc在滋养细胞功能障碍中的作用。使用基因表达综合数据库分析PE患者组织中的HSPB8表达。在H/R刺激的HTR8/SVneo细胞中检测到HSPB8表达后,通过用HSPB8特异性质粒转染该基因来过表达HSPB8。细胞计数试剂盒-8,伤口愈合和Transwell测定用于评估增殖,暴露于H/R条件的HTR8/SVneo细胞的迁移和侵袭。通过2,7-二氯二氢荧光素二乙酸染色确定活性氧(ROS)。5,5\',6,6'-四氯-1,1',应用3,3'-四乙基苯并咪唑碳碘化花青(JC-1)染色评估线粒体膜电位。使用相应的商业试剂盒检测丙二醛(MDA)和超氧化物歧化酶(SOD)水平。此外,通过末端脱氧核苷酸转移酶dUTP缺口末端标记(TUNEL)染色评估细胞凋亡的诱导。此外,Biogrid数据库预测HSPB8与c-Myc绑定,并且使用共免疫沉淀(Co-IP)测定来验证这种相互作用。随后,c-Myc表达被沉默以在暴露于H/R条件和上调HSPB8表达的HTR8/SVneo细胞中进行拯救实验。值得注意的是,PE组织和H/R刺激的HTR8/SVneo细胞中HSPB8表达降低。HSPB8强制表达促进增殖,HTR8/SVneo细胞的迁移和侵袭。此外,H/R导致ROS和MDA水平增加以及TUNEL染色和聚集的JC-1荧光和SOD活性水平降低,HSPB8过表达后恢复。Co-IP证实了HSPB8和c-Myc之间的相互作用。此外,c-Myc表达的敲低损害了HSPB8上调对H/R诱导后滋养细胞功能障碍的影响。总的来说,数据表明,HSPB8可以通过与c-Myc结合来改善线粒体氧化应激,从而减轻滋养细胞功能障碍。这些发现可能为PE的发病机制提供新的见解,并强调HSPB8/c-Myc在未来PE的预防和治疗中的作用。
    Preeclampsia (PE) is a pregnancy-specific syndrome with complex pathogenesis. The present study aimed to explore the role of heat shock protein B8 (HSPB8) and c-Myc in trophoblast cell dysfunction using a hypoxia/reoxygenation (H/R)-treated HTR8/SVneo cell model. HSPB8 expression in tissues of patients with PE was analyzed using the Gene Expression Omnibus database. Following detection of HSPB8 expression in H/R-stimulated HTR8/SVneo cells, HSPB8 was overexpressed by transfection of the gene with a HSPB8-specific plasmid. Cell Counting Kit-8, wound healing and Transwell assays were used to evaluate the proliferation, migration and invasion of HTR8/SVneo cells exposed to H/R conditions. Reactive oxygen species (ROS) were determined by 2,7-dichlorodihydrofluorescein diacetate staining. 5,5\',6,6\'-tetrachloro-1,1\',3,3\'-tetraethylbenzimidazolocarbo-cyanine iodide (JC-1) staining was applied to assess mitochondrial membrane potential. Malondialdehyde (MDA) and superoxide dismutase (SOD) levels were detected using the corresponding commercial kits. In addition, the induction of apoptosis was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. Moreover, the Biogrid database predicted that HSPB8 was bound to c-Myc, and a co-immunoprecipitation (Co-IP) assay was used to verify this interaction. Subsequently, c-Myc expression was silenced to conduct rescue experiments in HTR8/SVneo cells exposed to H/R conditions and upregulated HSPB8 expression. Notably, reduced HSPB8 expression was noted in PE tissues and H/R-stimulated HTR8/SVneo cells. HSPB8 enforced expression promoted the proliferation, migration and invasion of HTR8/SVneo cells. Moreover, H/R caused an increase in ROS and MDA levels as well as in TUNEL staining and a decrease in aggregated JC-1 fluorescence and SOD activity levels, which were restored following HSPB8 overexpression. Co-IP confirmed the interaction between HSPB8 and c-Myc. Moreover, knockdown of c-Myc expression compromised the effects of HSPB8 upregulation on trophoblast cell dysfunction following induction of H/R. Collectively, the data indicated that HSPB8 could improve mitochondrial oxidative stress by binding to c-Myc to alleviate trophoblast cell dysfunction. The findings may provide new insights into the pathogenesis of PE and highlight the role of HSPB8/c-Myc in the prevention and treatment of PE in the future.
    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
    膀胱癌(BCa)是泌尿生殖系统中的重要恶性肿瘤。值得注意的是,热休克蛋白(HSPs)在受到环境胁迫的细胞中表现出升高的表达,并且与许多人类恶性肿瘤的进展有关。其中,HSPB8在BCa中的功能意义和具体机制尚未得到充分探索。在这项研究中,我们测量了BCa组织和各种细胞系中HSPB8的表达,进一步深入研究其对细胞行为的影响。我们的观察指出了BCa中HSPB8的上调,与更高级的临床表现密切相关的趋势。抑制HSPB8表现出细胞增殖和迁移能力的显著降低,同时放大细胞凋亡和诱导细胞周期阻滞。加强这些发现,我们使用小鼠模型进行的体内分析显示出类似的趋势.值得注意的是,在HSPB8击倒后,特定蛋白质的水平,包括eNOS(S1177),Hsp27(S78/S82),PRAS40(T246),RSK1/2(S221/S227),和STAT3(S727)下降,Hsp27(S78/S82)和PRAS40(T246)的下降幅度最大。此外,HSP27抑制剂的应用有效逆转了由HSPB8表达增加引起的表型。总的来说,我们的结果表明,升高的HSPB8表达可以作为BCa的潜在预后标志物,靶向HSPB8可能为治疗这种恶性肿瘤开辟新的治疗途径。
    Bladder cancer (BCa) stands as a significant malignancy within the genitourinary system. Notably, heat shock proteins (HSPs) exhibit elevated expression in cells subjected to environmental stresses and have been linked to the progression of many human malignancies. Among these, the functional implications and specific mechanism of HSPB8 in BCa have yet to be fully explored. In this study, we measured HSPB8 expression in both BCa tissues and various cell lines, further delving into its influence on cellular behaviors. Our observations pinpoint an upregulation of HSPB8 in BCa, a trend strongly associated with more advanced clinical manifestations. Suppressing HSPB8 exhibited marked reductions in cell proliferation and migration capabilities, while simultaneously amplifying apoptosis and inducing cell cycle arrest. Reinforcing these findings, our in vivo analyses using mouse models showed similar trends. Notably, upon HSPB8 knockdown, levels of specific proteins including eNOS (S1177), Hsp27 (S78/S82), PRAS40(T246), RSK1/2(S221/S227), and STAT3 (S727) decreased, with Hsp27 (S78/S82) and PRAS40(T246) experiencing the most profound drops. Furthermore, the application of an HSP27 inhibitor effectively reversed the phenotypes caused by increased HSPB8 expression. Collectively, our results suggest that elevated HSPB8 expression could act as a potential prognostic marker for BCa, and targeting HSPB8 might open new therapeutic avenues for treating this malignancy.
    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
    据报道,右美托咪定(Dex)在阿尔茨海默病(AD)中起神经保护作用。然而,具体机制尚不清楚.在AD模型中阐明Dex调控神经细胞凋亡的分子机制。将SH-SY5Y细胞用Aβ1-42(10μM)处理24h后,建立了体外AD模型。通过RIP分析验证了UPF1,lncRNASNHG14和HSPB8之间的相互作用。细胞活力,凋亡,基因的水平,并通过CCK-8测定法检测蛋白质,流式细胞术,蛋白质印迹,和qRT-PCR,分别。Dex下调lncRNASNHG14水平并抑制神经细胞凋亡。LncRNASNHG14过表达逆转了Dex对AD模型神经细胞凋亡的抑制作用。LncRNASNHG14通过招募UPF1减弱HSPB8mRNA的稳定性。HSPB8过表达抑制AD模型神经细胞凋亡。此外,HSPB8敲低可逆转Dex对AD模型神经细胞凋亡的抑制作用。我们的研究表明,Dex通过抑制lncRNASNHG14/UPF1轴促进HSPB8的表达,从而抑制AD中神经细胞的凋亡。
    Dexmedetomidine (Dex) is reported to play a neuroprotective role in Alzheimer\'s disease (AD). However, the specific mechanism remains unclear. Figure out the underlying molecular mechanism of Dex regulating nerve cell apoptosis in the AD model. The AD model in vitro was established after SH-SY5Y cells were treated with Aβ1 - 42 at (10 μM) for 24 h. The interaction among UPF1, lncRNA SNHG14, and HSPB8 was verified by RIP assay. Cell viability, apoptosis, the level of genes, and proteins were detected by CCK-8 assay, flow cytometry, Western blot, and qRT-PCR, respectively. Dex downregulated lncRNA SNHG14 level and inhibited apoptosis of nerve cells. LncRNA SNHG14 overexpression reversed the inhibitory effect of Dex on nerve cell apoptosis in the AD model. LncRNA SNHG14 attenuated HSPB8 mRNA stability by recruiting UPF1. HSPB8 overexpression inhibited apoptosis of nerve cells in the AD model. Moreover, HSPB8 knockdown reversed the inhibitory effect of Dex on nerve cell apoptosis in the AD model. Our study demonstrated that Dex promoted HSPB8 expression via inhibiting the lncRNA SNHG14/UPF1 axis to inhibit nerve cell apoptosis in AD.
    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
    最近,不受控制的肌动蛋白聚合已被认为是早发性血脑屏障(BBB)破裂的引发剂。这里,使用体外模型,我们发现氧糖剥夺/再灌注(OGD/R)后,HSPB8的内皮过度表达抑制了异常肌动蛋白聚合,从而保留了BBB的完整性。我们进一步研究了HSPB8控制肌动蛋白组装的机制。HSPB8抑制bEnd.3细胞中的RhoA/ROCK2/p-MLC信号通路,RhoA激活剂消除了HSPB8对OGD/R后肌动蛋白重组的抑制作用。此外,OGD/R后内皮自噬通量受损HSPB8过表达减弱了这种作用。自噬抑制部分逆转了HSPB8对RhoA/ROCK2/p-MLC通路的影响。一起来看,本研究表明,通过抑制RhoA/ROCK2/p-MLC信号通路,过表达HSPB8可以恢复自噬通量,逆转内皮细胞骨架肌动蛋白的聚集,最终减轻OGD/R诱导的BBB损伤。
    Recently, uncontrolled actin polymerization has been recognized as an initiator of early-onset blood-brain barrier (BBB) rupture. Here, using in vitro models, we found that after oxygen-glucose deprivation/reperfusion (OGD/R), endothelial overexpression of HSPB8 suppressed aberrant actin polymerization and thus preserved the integrity of BBB. We further investigated the mechanisms of HSPB8 in the control of actin assembly. HSPB8 suppressed the RhoA/ROCK2/p-MLC signaling pathway in bEnd.3 cells and the RhoA activator abrogated the inhibitory action of HSPB8 on actin reorganization after OGD/R. In addition, endothelial autophagic flux was impaired after OGD/R. This effect was attenuated by HSPB8 overexpression. Autophagy inhibition partially reversed the effect of HSPB8 on the RhoA/ROCK2/p-MLC pathway. Taken together, the present study revealed that the restoration of autophagic flux by overexpressing HSPB8, via the inhibition of the RhoA/ROCK2/p-MLC signaling pathway, reverses the aggregation of endothelial cytoskeleton actin, eventually alleviating OGD/R-induced BBB injury.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • DOI:
    文章类型: Journal Article
    目的:真核延伸因子1alpha-2(EEF1A2)和热休克蛋白B8(HSPB8)在癌症发生和发展中的关键作用已得到充分证实。然而,EEF1A2/HSPB8在胃癌(GC)发生发展中的调控作用尚未完全明白。本研究旨在阐明EEF1A2/HSPB8对GC细胞恶性行为的生物学效应,并探讨EEF1A2/HSPB8参与GC的分子机制。
    方法:在目前的工作中,采用逆转录-定量聚合酶链反应(RT-qPCR)和westernblot检测GC细胞中EEF1A2和HSPB8的表达差异。细胞计数试剂盒-8(CCK-8),5-乙炔基-2'-脱氧尿苷(EdU)染色,伤口愈合和transwell分析用于检测增殖,GC细胞的迁移和侵袭。此外,采用试管形成试验来评估与GC细胞条件培养基(CM)孵育的HUVEC的体外血管生成。此外,EEF1A2和HSPB8之间的相互作用从BioGrid数据库预测和分析通过免疫共沉淀(Co-IP)。
    结果:本研究显示EEF1A2和HSPB8在GC细胞系中高表达。EEF1A2敲低明显抑制增殖,GC细胞的迁移和侵袭以及体外血管生成。此外,证实EEF1A2与HSPB8相互作用并正调控HSPB8的表达。HSPB8的过表达逆转了EEF1A2敲低对GC细胞增殖的抑制作用,迁移,侵袭和体外血管生成。
    结论:结论:EEF1A2可能通过促进HSPB8的表达而作为癌基因参与GC的发展。
    OBJECTIVE: The critical roles of eukaryotic elongation factor 1alpha-2 (EEF1A2) and heat shock protein B8 (HSPB8) in the carcinogenesis and progression of cancers have been well documented. However, the regulatory role of EEF1A2/HSPB8 in the development of gastric cancer (GC) have not been fully understood. This study was aimed at clarifying the biological effects of EEF1A2/HSPB8 on the malignant behaviors of GC cells and to investigate the molecular mechanism underlying the involvement of EEF1A2/HSPB8 in GC.
    METHODS: In the present work, expression differences of EEF1A2 and HSPB8 in GC cells were detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot assay. Cell counting kit -8 (CCK-8), 5-ethynyl-2\'-deoxyuridine (EdU) staining, wound healing and transwell assays were employed to detect the proliferation, migration and invasion of GC cells. In addition, tube formation assay was adopted to assess in vitro angiogenesis of HUVECs incubated with the conditioned media (CM) of GC cells. Moreover, the interaction between EEF1A2 and HSPB8 was predicted from BioGrid database and analyzed through co-immunoprecipitation (Co-IP).
    RESULTS: The present research revealed that EEF1A2 and HSPB8 were highly expressed in GC cell lines. EEF1A2 knockdown markedly suppressed the proliferation, migration and invasion of GC cells as well as in vitro angiogenesis. Furthermore, it was verified that EEF1A2 interacted with HSPB8 and positively regulated HSPB8 expression. Overexpression of HSPB8 reversed the suppressive effects of EEF1A2 knockdown on GC cell proliferation, migration, invasion and in vitro angiogenesis.
    CONCLUSIONS: In conclusion, EEF1A2 could act as an oncogene in the development of GC via promoting HSPB8 expression.
    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
    脓毒症相关性脑病(SAE)与认知缺陷的高风险相关;然而,其潜在机制尚不清楚。最近,研究表明,HSPB8是一个小的热休克蛋白家族,影响认知功能并改善脓毒症诱导的功能障碍。然而,HSPB8在SAE相关认知障碍中的作用尚未阐明.在这项研究中,我们发现在脂多糖诱导的脓毒症小鼠脑内HSPB8表达上调.HSPB8过表达减轻了SAE小鼠的认知能力下降。此外,外源性HSPB8通过调节NRF1/TFAM诱导的线粒体生物发生和DRP1介导的线粒体裂变在脂多糖诱导的小鼠模型中发挥神经保护作用和挽救突触功能。此外,HSPB8过表达抑制SAE模型中的IBA1和NLRP3活化。HSPB8的过表达可能是缓解SAE相关认知下降的有效治疗方法。
    Sepsis-associated encephalopathy (SAE) is associated with a higher risk of cognitive deficits; however, its potential mechanisms are still unknow. Recently, researches show that HSPB8, a family of small heat shock proteins, affects cognitive function and ameliorates sepsis-induced dysfunction. However, the role of HSPB8 in SAE-associated cognitive impairment has not been elucidated. In this study, we found that HSPB8 expression was up-regulated in the brain of mice with lipopolysaccharide-induced sepsis. HSPB8 overexpression alleviated cognitive decline in SAE mice. In addition, exogenous HSPB8 exerts neuroprotective effects and salvages synaptic function via regulating NRF1/TFAM-induced mitochondrial biogenesis and DRP1-mediate mitochondrial fission in a lipopolysaccharide-induced mouse model. Furthermore, HSPB8 overexpression inhibits IBA1 and NLRP3 activation in the SAE model. Overexpression of HSPB8 may be an efficient treatment for relieving SAE-related cognitive decline.
    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
    肌萎缩侧索硬化(ALS)是通过突变异常折叠蛋白的积累鉴定的神经元变性病症。多肽的天然折叠是由分子伴侣介导的,防止它们的致病性聚集。ALS中的突变蛋白表达与伴侣能力的截留和消耗有关。缺乏对伴侣参与ALS发病机制的透彻了解,对其治疗提出了重大挑战。这里,我们回顾了ALS相关突变体FUS的积累,TDP-43、SOD1和C9orf72蛋白破坏细胞稳态机制导致神经元丢失。Further,我们讨论了HSP70和DNAJ家族共同伴侣如何作为减少ALS中错误折叠蛋白积累的潜在靶标。此外,小HSPB1和HSPB8分子伴侣可以促进神经保护和防止应激相关的错误折叠蛋白凋亡。通过药理学增强细胞伴侣能力以减少突变蛋白对ALS病理机制的蛋白毒性作用来设计治疗策略可能是相当大的进步。监护人,除了直接与错误折叠的蛋白质相互作用以进行蛋白质质量控制外,还可以通过启动强烈的应激反应途径来过滤它们的毒性,调节转录表达谱,和促进抗凋亡功能。总的来说,分子伴侣的这些特性使它们成为获得错误折叠蛋白质疾病的基本见解并设计针对ALS的更有效疗法的有吸引力的靶标。
    Amyotrophic lateral sclerosis (ALS) is a neuronal degenerative condition identified via a build-up of mutant aberrantly folded proteins. The native folding of polypeptides is mediated by molecular chaperones, preventing their pathogenic aggregation. The mutant protein expression in ALS is linked with the entrapment and depletion of chaperone capacity. The lack of a thorough understanding of chaperones\' involvement in ALS pathogenesis presents a significant challenge in its treatment. Here, we review how the accumulation of the ALS-linked mutant FUS, TDP-43, SOD1, and C9orf72 proteins damage cellular homeostasis mechanisms leading to neuronal loss. Further, we discuss how the HSP70 and DNAJ family co-chaperones can act as potential targets for reducing misfolded protein accumulation in ALS. Moreover, small HSPB1 and HSPB8 chaperones can facilitate neuroprotection and prevent stress-associated misfolded protein apoptosis. Designing therapeutic strategies by pharmacologically enhancing cellular chaperone capacity to reduce mutant protein proteotoxic effects on ALS pathomechanisms can be a considerable advancement. Chaperones, apart from directly interacting with misfolded proteins for protein quality control, can also filter their toxicity by initiating strong stress-response pathways, modulating transcriptional expression profiles, and promoting anti-apoptotic functions. Overall, these properties of chaperones make them an attractive target for gaining fundamental insights into misfolded protein disorders and designing more effective therapies against ALS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号