cell membrane

细胞膜
  • 文章类型: Journal Article
    超长链脂肪酸(VLCFAs)调节细胞膜的生物物理特性,以确定真核生物的生长和发育,如稻瘟病菌稻瘟病菌的发病机理。脂肪酸延伸酶Elo1通过调节VLCFA生物合成来调节米曲霉的发病机理。然而,目前尚不清楚Elo1是否以及如何与其他因素联合调节真菌病原体中的VLCFA生物合成。这里,我们在米曲霉中通过邻近标记鉴定了Ifa38,Phs1和Tsc13为Elo1的相互作用蛋白。Elo1与内质网(ER)膜上的Ifa38,Phs1和Tsc13相关,以控制VLCFA的生物合成。靶向基因缺失突变体Δifa38,Δphs1和Δtsc13在营养生长中都与Δelo1相似地受损,分生孢子形态,ER中的应激反应,细胞壁和细胞膜。这些缺失突变体还显示出细胞膜完整性的严重损害,并且未能组织对穿透钉形成和致病性至关重要的隔膜环。我们的研究表明,米曲霉利用脂肪酸延伸酶复合物来调节VLCFAs以维持或重塑细胞膜结构,这对septin介导的宿主渗透很重要。
    Very-long-chain fatty acids (VLCFAs) regulate biophysical properties of cell membranes to determine growth and development of eukaryotes, such as the pathogenesis of the rice blast fungus Magnaporthe oryzae. The fatty acid elongase Elo1 regulates pathogenesis of M. oryzae by modulating VLCFA biosynthesis. However, it remains unknown whether and how Elo1 associates with other factors to regulate VLCFA biosynthesis in fungal pathogens. Here, we identified Ifa38, Phs1 and Tsc13 as interacting proteins of Elo1 by proximity labelling in M. oryzae. Elo1 associated with Ifa38, Phs1 and Tsc13 on the endoplasmic reticulum (ER) membrane to control VLCFA biosynthesis. Targeted gene deletion mutants Δifa38, Δphs1 and Δtsc13 were all similarly impaired as Δelo1 in vegetative growth, conidial morphology, stress responses in ER, cell wall and membrane. These deletion mutants also displayed severe damage in cell membrane integrity and failed to organize the septin ring that is essential for penetration peg formation and pathogenicity. Our study demonstrates that M. oryzae employs a fatty acid elongase complex to regulate VLCFAs for maintaining or remodelling cell membrane structure, which is important for septin-mediated host penetration.
    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
    聚合物介导的药物/基因递送的表面官能化对疾病治疗具有巨大的潜力。然而,聚合物表面官能化的设计原理仍然难以捉摸。在这项研究中,我们采用计算机模拟来演示刚度,长度,密度,和聚合物配体的分布影响它们穿过细胞膜的渗透能力。我们的模拟表明,聚合物配体的刚度影响它们跨膜运输货物的能力。增加聚合物配体的刚度可以促进它们跨膜的递送,特别是对于较大的货物。此外,适当增加聚合物配体的长度可以更有利于协助货物进入膜的下层。此外,聚合物配体在货物表面的分布在其运输中也起着至关重要的作用。具体来说,聚合物配体的四分之一模式和条纹模式分布表现出更高的渗透能力,协助货物穿透膜。这些发现为设计用于药物/基因递送的高效功能化聚合物配体提供了仿生灵感。
    The surface functionalization of polymer-mediated drug/gene delivery holds immense potential for disease therapy. However, the design principles underlying the surface functionalization of polymers remain elusive. In this study, we employed computer simulations to demonstrate how the stiffness, length, density, and distribution of polymer ligands influence their penetration ability across the cell membrane. Our simulations revealed that the stiffness of polymer ligands affects their ability to transport cargo across the membrane. Increasing the stiffness of polymer ligands can promote their delivery across the membrane, particularly for larger cargoes. Furthermore, appropriately increasing the length of polymer ligands can be more conducive to assisting cargo to enter the lower layer of the membrane. Additionally, the distribution of polymer ligands on the surface of the cargo also plays a crucial role in its transport. Specifically, the one-fourth mode and stripy mode distributions of polymer ligands exhibited higher penetration ability, assisting cargoes in penetrating the membrane. These findings provide biomimetic inspiration for designing high-efficiency functionalization polymer ligands for drug/gene delivery.
    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
    骨折愈合结果很大程度上取决于病变部位的间充质干细胞(MSC)的数量和成骨分化能力。在这里,巨噬细胞膜(MM)可逆掩蔽的纳米复合物(NCs)被设计用于短基质衍生因子1α肽(sSDF-1α)和Ckip-1小干扰RNA(Ckip-1siRNA,siCkip-1)通过同时促进内源性MSCs的募集和成骨分化来促进骨修复。为了建立NC,穿透膜的α-螺旋多肽首先与siCkip-1组装,然后将阳离子NC依次用过氧化氢酶和sSDF-1α锚定MM的外壳包被。由于MM辅助炎症归巢,静脉注射NC可以有效地积聚在股骨骨折处,其中过氧化氢酶分解局部过氧化氢以产生氧气气泡,从而驱使sSDF-1α锚定的MM在细胞外室中脱落。暴露的,因此,阳离子内核能够实现稳健的跨膜递送到MSC中以诱导Ckip-1沉默。因此,sSDF-1α引导的MSCs募集与siCkip-1介导的成骨分化协作以促进骨形成并加速骨折愈合。本研究为大分子药物分层共递送到不同的细胞区室提供了一种开明的策略,它也为骨折愈合的管理提供了一种有希望的方式。
    The fracture healing outcome is largely dependent on the quantities as well as osteogenic differentiation capacities of mesenchymal stem cells (MSCs) at the lesion site. Herein, macrophage membrane (MM)-reversibly cloaked nanocomplexes (NCs) are engineered for the lesion-targeted and hierarchical co-delivery of short stromal derived factor-1α peptide (sSDF-1α) and Ckip-1 small interfering RNA (Ckip-1 siRNA, siCkip-1) to promote bone repair by concurrently fostering recruitment and osteogenic differentiation of endogenous MSCs. To construct the NCs, a membrane-penetrating α-helical polypeptide first assembles with siCkip-1, and the cationic NCs are sequentially coated with catalase and an outer shell of sSDF-1α-anchored MM. Due to MM-assisted inflammation homing, intravenously injected NCs could efficiently accumulate at the fractured femur, where catalase decomposes the local hydrogen peroxide to generate oxygen bubbles that drives the shedding of sSDF-1α-anchored MM in the extracellular compartment. The exposed, cationic inner core thus enables robust trans-membrane delivery into MSCs to induce Ckip-1 silencing. Consequently, sSDF-1α-guided MSCs recruitment cooperates with siCkip-1-mediated osteogenic differentiation to facilitate bone formation and accelerate bone fracture healing. This study provides an enlightened strategy for the hierarchical co-delivery of macromolecular drugs into different cellular compartments, and it also renders a promising modality for the management of fracture healing.
    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
    平面细胞极性(PCP)的缺陷与多种人类病理有关。Vangl2是对PCP信令至关重要的核心PCP组件之一。Vangl2的失调与严重的神经管缺陷和癌症有关。然而,Vangl2蛋白是如何在翻译后水平上被调节的还没有被很好地理解。使用脂肪酰化和生化验证的化学报告基因,在这里,我们提出Vangl2亚细胞定位受可逆的S-硬脂酰化循环调节.其动态过程主要受ZDHHC9酰基转移酶和脱酰基酶酰蛋白硫酯酶1(APT1)调控。Vangl2的硬脂酰化缺陷型突变体显示出降低的质膜定位,导致细胞迁移过程中PCP建立的破坏。遗传或药理学抑制ZDHHC9表型对Vangl2的硬脂酰化损失的影响。此外,Vangl2硬脂酰化的缺失增强了致癌Yes相关蛋白1(YAP)的激活,丝氨酸-苏氨酸激酶AKT,和细胞外信号调节蛋白激酶(ERK)信号并促进乳腺癌细胞生长和HRasG12V突变体(HRasV12)诱导的致癌转化。我们的结果揭示了Vangl2的调节机制,并提供了有关脂肪酸代谢和蛋白质脂肪酰化如何通过核心PCP蛋白质脂化调节PCP信号传导和肿瘤发生的机制见解。
    Defects in planar cell polarity (PCP) have been implicated in diverse human pathologies. Vangl2 is one of the core PCP components crucial for PCP signaling. Dysregulation of Vangl2 has been associated with severe neural tube defects and cancers. However, how Vangl2 protein is regulated at the posttranslational level has not been well understood. Using chemical reporters of fatty acylation and biochemical validation, here we present that Vangl2 subcellular localization is regulated by a reversible S-stearoylation cycle. The dynamic process is mainly regulated by acyltransferase ZDHHC9 and deacylase acyl-protein thioesterase 1 (APT1). The stearoylation-deficient mutant of Vangl2 shows decreased plasma membrane localization, resulting in disruption of PCP establishment during cell migration. Genetically or pharmacologically inhibiting ZDHHC9 phenocopies the effects of the stearoylation loss of Vangl2. In addition, loss of Vangl2 stearoylation enhances the activation of oncogenic Yes-associated protein 1 (YAP), serine-threonine kinase AKT, and extracellular signal-regulated protein kinase (ERK) signaling and promotes breast cancer cell growth and HRas G12V mutant (HRasV12)-induced oncogenic transformation. Our results reveal a regulation mechanism of Vangl2, and provide mechanistic insight into how fatty acid metabolism and protein fatty acylation regulate PCP signaling and tumorigenesis by core PCP protein lipidation.
    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: English Abstract
    目的:研究携带人抗氧化蛋白Cu-Zn超氧化物歧化酶(Cu,Zn-SOD)并评估这些融合蛋白的抗氧化和抗炎活性。
    方法:融合蛋白hPP10-Cu,Zn-SOD是通过基因工程获得的,并通过Western印迹进行鉴定。用免疫荧光法评价融合蛋白的膜穿透性,荧光共定位测定和蛋白质印迹,使用商业试剂盒检测其SOD酶活性,用MTT法评估其对细胞活力的影响。在H2O2诱导的氧化应激的HEK293细胞模型中,HPP10-Cu的作用,用流式细胞术和RT-qPCR分析Zn-SOD对细胞凋亡的影响,并采用活性氧(ROS)法评估其抗氧化作用;通过RT-qPCR检测炎症因子的表达,在TPA诱导的小鼠耳部炎症模型中评估其抗炎作用。蛋白质印迹和免疫组织化学。
    结果:融合蛋白hPP10-Cu,成功获得Zn-SOD。免疫荧光实验证实了这种融合蛋白在HEK293细胞中的明显膜渗透,定位在细胞膜和细胞进入后的细胞核。hPP10-Cu,浓度为5μmol/L的Zn-SOD表现出较强的抗氧化活性,但浓度高达10μmol/L时对细胞活力的影响最小。在小鼠耳部炎症模型中,融合蛋白明显抑制细胞凋亡,降低细胞内ROS水平,显著降低炎症因子的mRNA和蛋白表达。
    结论:融合蛋白hPP10-Cu,能够穿透细胞膜的Zn-SOD具有很强的抗氧化和抗炎活性,细胞毒性很小,证明了hPP10作为有效药物递送载体的价值和hPP10-Cu的潜力,Zn-SOD在护肤品开发中的应用
    OBJECTIVE: To investigate the cell membrane-penetrating capacity of human cell-penetrating peptide hPP10 carrying human antioxidant protein Cu-Zn superoxide dismutase (Cu, Zn-SOD) and assess the antioxidant and anti-inflammatory activity of these fusion proteins.
    METHODS: The fusion protein hPP10-Cu, Zn-SOD was obtained by genetic engineering and identified by Western blotting. The membrane-penetrating ability of the fusion protein was evaluated by immunofluorescence assay, fluorescence colocalization assay and Western blotting, its SOD enzyme activity was detected using a commercial kit, and its effect on cell viability was assessed with MTT assay. In a HEK293 cell model of H2O2-induced oxidative stress, the effect of hPP10-Cu, Zn-SOD on cell apoptosis was analyzed with flow cytometry and RT-qPCR, and its antioxidant effect was assessed using reactive oxygen species (ROS) assay; its anti-inflammatory effect was evaluated in mouse model of TPA-induced ear inflammation by detecting expression of the inflammatory factors using RT-qPCR, Western blotting and immunohistochemistry.
    RESULTS: The fusion protein hPP10-Cu, Zn-SOD was successfully obtained. Immunofluorescence assay confirmed obvious membrane penetration of this fusion protein in HEK293 cells, localized both in the cell membrane and the cell nuclei after cell entry. hPP10-Cu, Zn-SOD at the concentration of 5 μmol/L exhibited strong antioxidant activity with minimal impact on cell viability at the concentration up to 10 μmol/L. The fusion protein obviously inhibited apoptosis and decreased intracellular ROS level in the oxidative stress cell model and significantly reduced mRNA and protein expression of the inflammatory factors in the mouse model of ear inflammation.
    CONCLUSIONS: The fusion protein hPP10-Cu, Zn-SOD capable of penetrating the cell membrane possesses strong antioxidant and anti-inflammatory activities with only minimal cytotoxicity, demonstrating the value of hPP10 as an efficient drug delivery vector and the potential of hPP10-Cu, Zn-SOD in the development of skincare products.
    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
    白血病是一种起源于骨髓的血液系统恶性肿瘤,它提供了启动的基本信号,programming,和白血病复发。然而,如何特异性地将药物递送到骨髓仍然难以捉摸。这里,我们通过用脂质体(HSPC脂质体)输注造血干细胞和祖细胞(HSPC)膜来开发仿生囊泡,通过透明质酸-CD44轴迁移到白血病小鼠的骨髓。此外,仿生囊泡通过细胞间粘附分子-1(ICAM-1)/整联蛋白β2(ITGB2)相互作用对白血病细胞表现出优异的结合亲和力。进一步的实验证实,携带化疗药物阿糖胞苷(Ara-C@HSPC-Lipo)的囊泡明显抑制细胞增殖,诱导白血病细胞凋亡和分化,减少白血病干细胞的数量。机械上,RNA-seq显示Ara-C@HSPC-Lipo处理诱导细胞凋亡和分化并抑制致癌途径。最后,我们证实HSPC脂质体在小鼠体内是安全的。本研究为靶向骨髓和治疗白血病提供了一种方法。
    Leukemia is a kind of hematological malignancy originating from bone marrow, which provides essential signals for initiation, progression, and recurrence of leukemia. However, how to specifically deliver drugs to the bone marrow remains elusive. Here, we develop biomimetic vesicles by infusing hematopoietic stem and progenitor cell (HSPC) membrane with liposomes (HSPC liposomes), which migrate to the bone marrow of leukemic mice via hyaluronic acid-CD44 axis. Moreover, the biomimetic vesicles exhibit superior binding affinity to leukemia cells through intercellular cell adhesion molecule-1 (ICAM-1)/integrin β2 (ITGB2) interaction. Further experiments validate that the vesicles carrying chemotherapy drug cytarabine (Ara-C@HSPC-Lipo) markedly inhibit proliferation, induce apoptosis and differentiation of leukemia cells, and decrease number of leukemia stem cells. Mechanically, RNA-seq reveals that Ara-C@HSPC-Lipo treatment induces apoptosis and differentiation and inhibits the oncogenic pathways. Finally, we verify that HSPC liposomes are safe in mice. This study provides a method for targeting bone marrow and treating leukemia.
    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
    广泛耐药和泛耐药的铜绿假单胞菌日益流行是全球公共卫生的主要问题。因此,开发特异性靶向铜绿假单胞菌及其生物膜的新型抗微生物剂至关重要。在本研究中,我们确定盐酸小檗碱抑制浮游细菌的生长以及阻止生物膜的形成。此外,我们观察到pslA和pelA生物膜相关基因的表达下调。与现有抗生素相比,盐酸小檗碱对铜绿假单胞菌表现出多种作用模式。我们的发现表明,盐酸小檗碱通过破坏细菌细胞膜发挥其抗菌作用,产生活性氧(ROS),降低细胞内三磷酸腺苷(ATP)水平。此外,盐酸小檗碱显示最小的细胞毒性和降低的耐药性。在腹膜炎的小鼠模型中,显著抑制铜绿假单胞菌的生长,表现出强烈的抑菌作用。总之,盐酸小檗碱是一种安全有效的抑制铜绿假单胞菌生长的抗菌剂。
    The increasing prevalence of extensively drug-and pan-drug-resistant Pseudomonas aeruginosa is a major concern for global public health. Therefore, it is crucial to develop novel antimicrobials that specifically target P. aeruginosa and its biofilms. In the present study, we determined that berberine hydrochloride inhibited the growth of planktonic bacteria as well as prevented the formation of biofilms. Moreover, we observed downregulation in the expression of pslA and pelA biofilm-related genes. Compared with existing antibiotics, berberine hydrochloride exhibits multiple modes of action against P. aeruginosa. Our findings suggest that berberine hydrochloride exerts its antimicrobial effects by damaging bacterial cell membranes, generating reactive oxygen species (ROS), and reducing intracellular adenosine triphosphate (ATP) levels. Furthermore, berberine hydrochloride showed minimal cytotoxicity and reduced susceptibility to drug resistance. In a mouse model of peritonitis, it significantly inhibited the growth of P. aeruginosa and exhibited a strong bacteriostatic action. In conclusion, berberine hydrochloride is a safe and effective antibacterial agent that inhibits the growth of P. aeruginosa.
    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
    被称为细胞外囊泡(EV)的天然生成的脂质纳米颗粒作为可工程化的治疗递送载体具有显著的前景。然而,以可用于递送的方式将蛋白质货物主动装载到EV中仍然是一个挑战。这里,我们证明,通过合理设计蛋白质运输到质膜并与脂筏相关联,我们可以增强一组结构多样的跨膜和外周膜蛋白的蛋白质货物装载到EV中。然后我们证明了选择的脂质标签介导增加的EV负载和工程化转录因子的功能性递送以调节靶细胞中的基因表达的能力。我们设想可以利用这项技术来开发新的基于EV的疗法,提供广泛的大分子货物。
    Naturally generated lipid nanoparticles termed extracellular vesicles (EVs) hold significant promise as engineerable therapeutic delivery vehicles. However, active loading of protein cargo into EVs in a manner that is useful for delivery remains a challenge. Here, we demonstrate that by rationally designing proteins to traffic to the plasma membrane and associate with lipid rafts, we can enhance loading of protein cargo into EVs for a set of structurally diverse transmembrane and peripheral membrane proteins. We then demonstrate the capacity of select lipid tags to mediate increased EV loading and functional delivery of an engineered transcription factor to modulate gene expression in target cells. We envision that this technology could be leveraged to develop new EV-based therapeutics that deliver a wide array of macromolecular cargo.
    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
    开发具有增强的生物相容性和靶向能力的T1加权磁共振成像(MRI)造影剂至关重要,因为人们担心当前药物的潜在毒性和次优性能。从“仿生伪装”中汲取灵感,“我们通过挤压方法从人胶质母细胞瘤(T98G)细胞系中分离细胞膜(CMs),以促进同源型胶质瘤的靶向。在六水合氯化铁与没食子酸(GA)的质量比为8:1时,所得的铁(Fe)-GA纳米颗粒(NPs)被证明是有效的T1加权MRI造影剂。T98GCM涂层的Fe-GANP显示出改善的同型胶质瘤靶向,通过普鲁士蓝染色和体外MRI验证。这种仿生伪装策略有望以安全有效的方式开发靶向治疗药物。
    Developing T1-weighted magnetic resonance imaging (MRI) contrast agents with enhanced biocompatibility and targeting capabilities is crucial owing to concerns over current agents\' potential toxicity and suboptimal performance. Drawing inspiration from \"biomimetic camouflage,\" we isolated cell membranes (CMs) from human glioblastoma (T98G) cell lines via the extrusion method to facilitate homotypic glioma targeting. At an 8:1 mass ratio of ferric chloride hexahydrate to gallic acid (GA), the resulting iron (Fe)-GA nanoparticles (NPs) proved effective as a T1-weighted MRI contrast agent. T98G CM-coated Fe-GA NPs demonstrated improved homotypic glioma targeting, validated through Prussian blue staining and in vitro MRI. This biomimetic camouflage strategy holds promise for the development of targeted theranostic agents in a safe and effective manner.
    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
    过氧化物酶体是执行多种代谢功能的多功能细胞器。PEX3,过氧化物酶体的关键调节剂,参与与过氧化物酶体相关的各种生物过程。PEX3是否参与过氧化物酶体相关的氧化还原稳态和心肌再生修复仍然难以捉摸。我们研究了心肌细胞特异性PEX3敲除(Pex3-KO)导致氧化还原稳态失衡,并破坏了不同时间和空间位置的内源性增殖/发育。使用Pex3-KO小鼠和心肌靶向干预方法,探讨了PEX3在生理和病理阶段对心肌再生修复的影响。机械上,脂质代谢组学显示PEX3通过影响缩醛磷脂代谢促进心肌再生修复。Further,我们发现PEX3调节的缩醛磷脂通过ITGB3的质膜定位激活AKT/GSK3β信号通路。我们的研究表明,PEX3可能是损伤后心肌再生修复的新治疗靶标。
    The peroxisome is a versatile organelle that performs diverse metabolic functions. PEX3, a critical regulator of the peroxisome, participates in various biological processes associated with the peroxisome. Whether PEX3 is involved in peroxisome-related redox homeostasis and myocardial regenerative repair remains elusive. We investigate that cardiomyocyte-specific PEX3 knockout (Pex3-KO) results in an imbalance of redox homeostasis and disrupts the endogenous proliferation/development at different times and spatial locations. Using Pex3-KO mice and myocardium-targeted intervention approaches, the effects of PEX3 on myocardial regenerative repair during both physiological and pathological stages are explored. Mechanistically, lipid metabolomics reveals that PEX3 promotes myocardial regenerative repair by affecting plasmalogen metabolism. Further, we find that PEX3-regulated plasmalogen activates the AKT/GSK3β signaling pathway via the plasma membrane localization of ITGB3. Our study indicates that PEX3 may represent a novel therapeutic target for myocardial regenerative repair following injury.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号