首页 > 最新文献

bioRxiv - Cell Biology最新文献

英文 中文
RIPK1 is essential for Herpes Simplex Virus-triggered ZBP1-dependent necroptosis in human cells RIPK1 对人类细胞中由单纯疱疹病毒触发的 ZBP1 依赖性坏死至关重要
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.17.613393
Oluwamuyiwa T. Amusan, Shuqi Wang, Chaoran Yin, Heather S. Koehler, Yixun Li, Tencho Tenev, Rebecca Wilson, Benjamin Bellenie, Ting Zhang, Jian Wang, chang liu, kim seong, Seyedeh L. Poorbaghi, Joseph Yates, Yuchen Shen, Jason W. Upton, Pascal Meier, Siddharth Balachandra, Hongyan Guo
Necroptosis initiated by the host sensor Z-NA Binding Protein-1 (ZBP1) is essential for host defense against a growing number of viruses, including Herpes Simplex Virus-1 (HSV-1). Studies with HSV-1 and other necroptogenic stimuli in murine settings have suggested that ZBP1 triggers necroptosis by directly complexing with the kinase RIPK3. Whether this is also the case in human cells, or whether additional co-factors are needed for ZBP1-mediated necroptosis, is unclear. Here, we show that ZBP1-induced necroptosis in human cells requires RIPK1. We have found that RIPK1 is essential for forming a stable and functional ZBP1-RIPK3 complex in human cells, but is dispensable for the formation of the equivalent murine complex. The RIP Homology Interaction Motif (RHIM) in RIPK3 is responsible for this difference between the two species, because replacing the RHIM in human RIPK3 with the RHIM from murine RIPK3 is sufficient to overcome the requirement for RIPK1 in human cells. These observations describe a critical mechanistic difference between mice and humans in how ZBP1 engages in necroptosis, with important implications for treating human diseases.
由宿主传感器 Z-NA 结合蛋白-1(ZBP1)引发的坏死对于宿主抵御越来越多的病毒(包括单纯疱疹病毒-1(HSV-1))至关重要。在小鼠体内使用 HSV-1 和其他致坏死刺激物进行的研究表明,ZBP1 通过直接与激酶 RIPK3 复合物触发坏死。在人体细胞中是否也是这种情况,或者 ZBP1 介导的坏死是否需要额外的辅助因子,目前还不清楚。在这里,我们发现 ZBP1 诱导的人体细胞坏死需要 RIPK1。我们发现,RIPK1 对于在人体细胞中形成稳定的、功能性的 ZBP1-RIPK3 复合物是必不可少的,但对于形成等效的鼠类复合物则是不可或缺的。RIPK3中的RIP同源相互作用元件(RIP Homology Interaction Motif,RHIM)是造成这两种生物之间差异的原因,因为用鼠RIPK3中的RHIM取代人RIPK3中的RHIM足以克服人体细胞对RIPK1的需要。这些观察结果描述了小鼠和人类在 ZBP1 如何参与坏死过程方面的关键机制差异,对治疗人类疾病具有重要意义。
{"title":"RIPK1 is essential for Herpes Simplex Virus-triggered ZBP1-dependent necroptosis in human cells","authors":"Oluwamuyiwa T. Amusan, Shuqi Wang, Chaoran Yin, Heather S. Koehler, Yixun Li, Tencho Tenev, Rebecca Wilson, Benjamin Bellenie, Ting Zhang, Jian Wang, chang liu, kim seong, Seyedeh L. Poorbaghi, Joseph Yates, Yuchen Shen, Jason W. Upton, Pascal Meier, Siddharth Balachandra, Hongyan Guo","doi":"10.1101/2024.09.17.613393","DOIUrl":"https://doi.org/10.1101/2024.09.17.613393","url":null,"abstract":"Necroptosis initiated by the host sensor Z-NA Binding Protein-1 (ZBP1) is essential for host defense against a growing number of viruses, including Herpes Simplex Virus-1 (HSV-1). Studies with HSV-1 and other necroptogenic stimuli in murine settings have suggested that ZBP1 triggers necroptosis by directly complexing with the kinase RIPK3. Whether this is also the case in human cells, or whether additional co-factors are needed for ZBP1-mediated necroptosis, is unclear. Here, we show that ZBP1-induced necroptosis in human cells requires RIPK1. We have found that RIPK1 is essential for forming a stable and functional ZBP1-RIPK3 complex in human cells, but is dispensable for the formation of the equivalent murine complex. The RIP Homology Interaction Motif (RHIM) in RIPK3 is responsible for this difference between the two species, because replacing the RHIM in human RIPK3 with the RHIM from murine RIPK3 is sufficient to overcome the requirement for RIPK1 in human cells. These observations describe a critical mechanistic difference between mice and humans in how ZBP1 engages in necroptosis, with important implications for treating human diseases.","PeriodicalId":501590,"journal":{"name":"bioRxiv - Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265194","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The binary protein interactome mapping of the Giardia lamblia proteasome lid reveals extra proteasomal functions of GlRpn11 蓝氏贾第鞭毛虫蛋白酶体盖的二元蛋白相互作用组图谱揭示了 GlRpn11 的额外蛋白酶体功能
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.18.613619
Ankita Das, Atrayee Ray, Nibedita Ray Chaudhuri, Soumyajit Mukherjee, Shubhra Ghosh Dastidar, Alok Ghosh, Sandipan Ganguly, Kuladip Jana, Srimonti Sarkar
Giardia lamblia does not encode Rpn12 and Sem1, two proteins crucial for assembling the proteasome lid. To understand how the interactions between the giardial proteasome lid subunits may have changed to compensate for their absence, we used the yeast two-hybrid assay to generate a binary protein interaction map of the Giardia lid subunits. Most interactions within the Giardia proteasome lid are stronger than those within the Saccharomyces cerevisiae lid. These may compensate for the absence of Rpn12 and Sem1. A notable exception was the weaker interaction between GlRpn11 and GlRpn8, compared to the strong interaction between Rpn11-Rpn8 of yeast. The Rpn11-Rpn8 dimer provides a platform for lid assembly and their interaction involves the insertion of a methionine residue of Rpn11 into a hydrophobic pocket of Rpn8. Molecular modeling indicates that GlRpn8s pocket is wider, reconciling the experimental observation of its weak interaction with GlRpn11. This weaker interaction may have evolved to support extra proteasomal functions of GlRpn11, which localizes to multiple subcellular regions where other proteasome subunits have not been detected. One such location is the mitosome. Functional complementation in yeast shows that GlRpn11 can influence mitochondrial function and distribution. This, together with its mitosomal localization, indicates that GlRpn11 functions at the mitosome. Thus, this parasites proteasome lid has a simpler subunit architecture and structural attributes that may support dual functionalities for GlRpn11. Such parasite-specific proteasome features could provide new avenues for controlling the transmission of Giardia.
蓝氏贾第鞭毛虫不编码Rpn12和Sem1,而这两种蛋白质对蛋白酶体盖的组装至关重要。为了了解贾第虫蛋白酶体盖亚基之间的相互作用是如何变化的,以弥补它们的缺失,我们使用酵母双杂交试验生成了贾第虫蛋白酶体盖亚基的二元蛋白质相互作用图谱。贾第虫蛋白酶体盖子内的大多数相互作用比酿酒酵母盖子内的相互作用更强。这可能弥补了 Rpn12 和 Sem1 的缺失。与酵母的 Rpn11-Rpn8 之间的强相互作用相比,GlRpn11 和 GlRpn8 之间的相互作用较弱,这是一个明显的例外。Rpn11-Rpn8 二聚体为盖子的组装提供了一个平台,它们之间的相互作用涉及 Rpn11 的一个蛋氨酸残基插入 Rpn8 的一个疏水口袋。分子建模表明,GlRpn8 的口袋更宽,这与实验观察到的它与 GlRpn11 的微弱相互作用相吻合。这种较弱的相互作用可能是为了支持 GlRpn11 的额外蛋白酶体功能而演变而来的,GlRpn11 定位在多个亚细胞区域,而在这些区域尚未检测到其他蛋白酶体亚基。其中一个位置就是有丝分裂体。酵母中的功能互补显示,GlRpn11 可以影响线粒体的功能和分布。这与其有丝分裂体定位一起表明,GlRpn11 在有丝分裂体中发挥作用。因此,这种寄生虫蛋白酶体盖具有更简单的亚基结构和结构属性,可能支持 GlRpn11 的双重功能。这种寄生虫特异性蛋白酶体特征可为控制贾第虫的传播提供新的途径。
{"title":"The binary protein interactome mapping of the Giardia lamblia proteasome lid reveals extra proteasomal functions of GlRpn11","authors":"Ankita Das, Atrayee Ray, Nibedita Ray Chaudhuri, Soumyajit Mukherjee, Shubhra Ghosh Dastidar, Alok Ghosh, Sandipan Ganguly, Kuladip Jana, Srimonti Sarkar","doi":"10.1101/2024.09.18.613619","DOIUrl":"https://doi.org/10.1101/2024.09.18.613619","url":null,"abstract":"Giardia lamblia does not encode Rpn12 and Sem1, two proteins crucial for assembling the proteasome lid. To understand how the interactions between the giardial proteasome lid subunits may have changed to compensate for their absence, we used the yeast two-hybrid assay to generate a binary protein interaction map of the Giardia lid subunits. Most interactions within the Giardia proteasome lid are stronger than those within the Saccharomyces cerevisiae lid. These may compensate for the absence of Rpn12 and Sem1. A notable exception was the weaker interaction between GlRpn11 and GlRpn8, compared to the strong interaction between Rpn11-Rpn8 of yeast. The Rpn11-Rpn8 dimer provides a platform for lid assembly and their interaction involves the insertion of a methionine residue of Rpn11 into a hydrophobic pocket of Rpn8. Molecular modeling indicates that GlRpn8s pocket is wider, reconciling the experimental observation of its weak interaction with GlRpn11. This weaker interaction may have evolved to support extra proteasomal functions of GlRpn11, which localizes to multiple subcellular regions where other proteasome subunits have not been detected. One such location is the mitosome. Functional complementation in yeast shows that GlRpn11 can influence mitochondrial function and distribution. This, together with its mitosomal localization, indicates that GlRpn11 functions at the mitosome. Thus, this parasites proteasome lid has a simpler subunit architecture and structural attributes that may support dual functionalities for GlRpn11. Such parasite-specific proteasome features could provide new avenues for controlling the transmission of Giardia.","PeriodicalId":501590,"journal":{"name":"bioRxiv - Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265195","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The role of disease state in confined migration 疾病状态在封闭迁移中的作用
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.16.608435
Annika Meid
Cell migration is a fundamental process in both normal and cancerous tissues, playing a crucial role in development, immune responses, and, in the case of cancer cells, metastasis- a leading cause of cancer-related mortality. Understanding the differences between healthy and cancerous cell migration is essential for uncovering potential therapeutic targets. This study aims to elucidate these differences by comparing the migratory behaviors of healthy cells (nHDF cells) and cancer cells (MDA-MB-231 cells). Our findings reveal that cancer cells significantly reduce their stiffness during migration through narrow channels, a phenomenon not observed in healthy cells. Additionally, DNA and membrane repair mechanisms are more active in healthy cells during migration compared to tumor cells. Notably, the use of MDA-MB-231 FUCCI cells demonstrates that the cell cycle profoundly influences cell migration under confined conditions. These insights provide a deeper understanding of the cellular mechanisms driving migration in both healthy and cancerous cells.
细胞迁移是正常组织和癌变组织的基本过程,在发育、免疫反应以及癌细胞转移(癌症相关死亡的主要原因)中发挥着至关重要的作用。了解健康细胞和癌细胞迁移之间的差异对于发现潜在的治疗靶点至关重要。本研究旨在通过比较健康细胞(nHDF 细胞)和癌细胞(MDA-MB-231 细胞)的迁移行为来阐明这些差异。我们的研究结果表明,癌细胞在通过狭窄通道迁移的过程中会明显降低其硬度,而这是在健康细胞中观察不到的现象。此外,与肿瘤细胞相比,健康细胞在迁移过程中的 DNA 和膜修复机制更为活跃。值得注意的是,MDA-MB-231 FUCCI 细胞的使用表明,在封闭条件下,细胞周期对细胞迁移有深刻影响。这些见解加深了人们对驱动健康细胞和癌细胞迁移的细胞机制的理解。
{"title":"The role of disease state in confined migration","authors":"Annika Meid","doi":"10.1101/2024.09.16.608435","DOIUrl":"https://doi.org/10.1101/2024.09.16.608435","url":null,"abstract":"Cell migration is a fundamental process in both normal and cancerous tissues, playing a crucial role in development, immune responses, and, in the case of cancer cells, metastasis- a leading cause of cancer-related mortality. Understanding the differences between healthy and cancerous cell migration is essential for uncovering potential therapeutic targets. This study aims to elucidate these differences by comparing the migratory behaviors of healthy cells (nHDF cells) and cancer cells (MDA-MB-231 cells). Our findings reveal that cancer cells significantly reduce their stiffness during migration through narrow channels, a phenomenon not observed in healthy cells. Additionally, DNA and membrane repair mechanisms are more active in healthy cells during migration compared to tumor cells. Notably, the use of MDA-MB-231 FUCCI cells demonstrates that the cell cycle profoundly influences cell migration under confined conditions. These insights provide a deeper understanding of the cellular mechanisms driving migration in both healthy and cancerous cells.","PeriodicalId":501590,"journal":{"name":"bioRxiv - Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265192","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Differential translocation of bacteriophages across the intestinal barrier in health and Crohn's disease 噬菌体在健康人和克罗恩病患者肠道屏障上的转运差异
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.17.613249
Clara Douadi, Ilias Theodorou, Quentin Lamy-Besnier, Yanis Sbardella, Loic Brot, Paolo Emidio Costantini, Roberto Saporetti, Alberto Danielli, Matteo Calvaresi, Marianne De Paepe, Harry Sokol, Diego Garcia-Weber, Veronique Carriere, Sophie Thenet, Luisa De Sordi
Impaired intestinal barrier function is a major feature of Crohn's disease (CD), leading to exacerbated inflammation in response to the microbiota. In this context, the translocation of intestinal bacteriophages (phages) and their effects on the host have been little investigated. We used phage fluorescence imaging coupled with ex-vivo and in-vitro models that mimic physiological and inflammatory conditions and found that phages can translocate across the intestinal barrier without disrupting its integrity. Although the translocation rate across the intestinal epithelium depended on phage morphology and the condition of the barrier, these factors did not influence the crossing of phage across the vascular endothelium. Virome analysis confirmed that viral sequences shared between blood and stool samples are more abundant in CD patients than healthy subjects, indicating that a barrier defect facilitates phage translocation from the gut to the bloodstream.
肠道屏障功能受损是克罗恩病(CD)的一个主要特征,它会导致炎症加剧,从而对微生物群产生反应。在这种情况下,肠道噬菌体(噬菌体)的转运及其对宿主的影响却鲜有研究。我们使用噬菌体荧光成像技术,结合模拟生理和炎症条件的体内外模型,发现噬菌体可以在不破坏肠道屏障完整性的情况下转运穿过肠道屏障。虽然穿越肠上皮细胞的转运率取决于噬菌体的形态和屏障的状况,但这些因素并不影响噬菌体穿越血管内皮细胞。病毒组分析证实,与健康人相比,CD患者血液和粪便样本中共享的病毒序列更丰富,这表明屏障缺陷促进了噬菌体从肠道向血液的转运。
{"title":"Differential translocation of bacteriophages across the intestinal barrier in health and Crohn's disease","authors":"Clara Douadi, Ilias Theodorou, Quentin Lamy-Besnier, Yanis Sbardella, Loic Brot, Paolo Emidio Costantini, Roberto Saporetti, Alberto Danielli, Matteo Calvaresi, Marianne De Paepe, Harry Sokol, Diego Garcia-Weber, Veronique Carriere, Sophie Thenet, Luisa De Sordi","doi":"10.1101/2024.09.17.613249","DOIUrl":"https://doi.org/10.1101/2024.09.17.613249","url":null,"abstract":"Impaired intestinal barrier function is a major feature of Crohn's disease (CD), leading to exacerbated inflammation in response to the microbiota. In this context, the translocation of intestinal bacteriophages (phages) and their effects on the host have been little investigated. We used phage fluorescence imaging coupled with ex-vivo and in-vitro models that mimic physiological and inflammatory conditions and found that phages can translocate across the intestinal barrier without disrupting its integrity. Although the translocation rate across the intestinal epithelium depended on phage morphology and the condition of the barrier, these factors did not influence the crossing of phage across the vascular endothelium. Virome analysis confirmed that viral sequences shared between blood and stool samples are more abundant in CD patients than healthy subjects, indicating that a barrier defect facilitates phage translocation from the gut to the bloodstream.","PeriodicalId":501590,"journal":{"name":"bioRxiv - Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265189","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Dynamic phosphorylation of Hcm1 promotes fitness in chronic stress Hcm1的动态磷酸化可在慢性压力下促进健康
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.18.613713
Michelle M Conti, Jillian P Bail, Rui Li, Lihua Julie Zhu, Jennifer A Benanti
Cell survival depends upon the ability to adapt to changing environments. Environmental stressors trigger an acute stress response program that rewires cell physiology, downregulates proliferation genes and pauses the cell cycle until the cell adapts. Here, we show that dynamic phosphorylation of the yeast cell cycle-regulatory transcription factor Hcm1 is required to maintain fitness in chronic stress. Hcm1 is activated by cyclin dependent kinase (CDK) and inactivated by the phosphatase calcineurin (CN) in response to stressors that signal through increases in cytosolic Ca2+. Expression of a constitutively active, phosphomimetic Hcm1 mutant reduces fitness in stress, suggesting Hcm1 inactivation is required. However, a comprehensive analysis of Hcm1 phosphomutants revealed that Hcm1 activity is also important to survive stress, demonstrating that Hcm1 activity must be toggled on and off to promote gene expression and fitness. These results suggest that dynamic control of cell cycle regulators is critical for survival in stressful environments.
细胞的存活取决于适应环境变化的能力。环境胁迫会触发急性应激反应程序,重构细胞生理结构、下调增殖基因并暂停细胞周期,直到细胞适应为止。在这里,我们发现酵母细胞周期调控转录因子 Hcm1 的动态磷酸化是在慢性压力下保持健康所必需的。Hcm1被细胞周期蛋白依赖激酶(CDK)激活,并在通过细胞膜Ca2+增加发出信号的应激源作用下被磷酸酶钙调磷酸酶(CN)灭活。表达具有组成型活性的拟磷酸化 Hcm1 突变体会降低应激时的适应性,这表明 Hcm1 失活是必需的。然而,对 Hcm1 磷酸拟态突变体的综合分析表明,Hcm1 的活性对于在应激中存活也很重要,这表明 Hcm1 的活性必须通过开关切换才能促进基因表达和存活。这些结果表明,细胞周期调节因子的动态控制对于在应激环境中生存至关重要。
{"title":"Dynamic phosphorylation of Hcm1 promotes fitness in chronic stress","authors":"Michelle M Conti, Jillian P Bail, Rui Li, Lihua Julie Zhu, Jennifer A Benanti","doi":"10.1101/2024.09.18.613713","DOIUrl":"https://doi.org/10.1101/2024.09.18.613713","url":null,"abstract":"Cell survival depends upon the ability to adapt to changing environments. Environmental stressors trigger an acute stress response program that rewires cell physiology, downregulates proliferation genes and pauses the cell cycle until the cell adapts. Here, we show that dynamic phosphorylation of the yeast cell cycle-regulatory transcription factor Hcm1 is required to maintain fitness in chronic stress. Hcm1 is activated by cyclin dependent kinase (CDK) and inactivated by the phosphatase calcineurin (CN) in response to stressors that signal through increases in cytosolic Ca<sup>2+</sup>. Expression of a constitutively active, phosphomimetic Hcm1 mutant reduces fitness in stress, suggesting Hcm1 inactivation is required. However, a comprehensive analysis of Hcm1 phosphomutants revealed that Hcm1 activity is also important to survive stress, demonstrating that Hcm1 activity must be toggled on and off to promote gene expression and fitness. These results suggest that dynamic control of cell cycle regulators is critical for survival in stressful environments.","PeriodicalId":501590,"journal":{"name":"bioRxiv - Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265190","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Development of a cell-permeable Biotin-HaloTag ligand to explore functional differences between protein variants across cellular generations 开发细胞渗透性生物素-HaloTag 配体,探索不同细胞世代蛋白质变体之间的功能差异
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.18.613519
Anoop Kumar Yadav, Abhijeet S. Jadhav, Pawel Szczepanik, Paolo Fagherazzi, Ivo Kabelka, Robert Vacha, Jakub Svenda, Hana Polasek-Sedlackova
HaloTag technology represents a versatile tool for studying proteins. Fluorescent HaloTag ligands employed in sequential labeling led to the discovery of distinct protein variants for histones, cohesins, and MCM complexes. Nonetheless, an efficient biochemical approach to separate the distinct protein variants to study their biological functions is missing. Principally being a gap in technology, the HaloTag toolbox lacks affinity ligands displaying good cell permeability and efficient affinity capture. Here, we describe the design, synthesis, and validation of a new cell-permeable Biotin-HaloTag ligand, which allows rapid labeling of Halo-tagged proteins in live cells and their efficient separation using streptavidin pull-down. Our work outlines how to use the herein-developed affinity ligand in sequential labeling to biochemically separate distinct protein variants and study their biological properties. The approach holds immense potential for addressing fundamental questions concerning essential cellular processes, including genome duplication and chromatin maintenance.
HaloTag 技术是研究蛋白质的多功能工具。采用荧光 HaloTag 配体进行连续标记,发现了组蛋白、粘合蛋白和 MCM 复合物的不同蛋白质变体。然而,目前还缺少一种有效的生化方法来分离不同的蛋白质变体,以研究它们的生物功能。HaloTag 工具箱缺乏具有良好细胞渗透性和高效亲和捕获能力的亲和配体,这主要是技术上的空白。在这里,我们描述了一种新型细胞渗透性生物素-HaloTag 配体的设计、合成和验证,这种配体可以在活细胞中快速标记 Halo 标记蛋白,并利用链霉亲和素牵引技术高效分离这些蛋白。我们的工作概述了如何使用这种开发的亲和配体进行连续标记,以生物化学方法分离不同的蛋白质变体并研究它们的生物学特性。这种方法在解决包括基因组复制和染色质维护在内的重要细胞过程的基本问题方面具有巨大的潜力。
{"title":"Development of a cell-permeable Biotin-HaloTag ligand to explore functional differences between protein variants across cellular generations","authors":"Anoop Kumar Yadav, Abhijeet S. Jadhav, Pawel Szczepanik, Paolo Fagherazzi, Ivo Kabelka, Robert Vacha, Jakub Svenda, Hana Polasek-Sedlackova","doi":"10.1101/2024.09.18.613519","DOIUrl":"https://doi.org/10.1101/2024.09.18.613519","url":null,"abstract":"HaloTag technology represents a versatile tool for studying proteins. Fluorescent HaloTag ligands employed in sequential labeling led to the discovery of distinct protein variants for histones, cohesins, and MCM complexes. Nonetheless, an efficient biochemical approach to separate the distinct protein variants to study their biological functions is missing. Principally being a gap in technology, the HaloTag toolbox lacks affinity ligands displaying good cell permeability and efficient affinity capture. Here, we describe the design, synthesis, and validation of a new cell-permeable Biotin-HaloTag ligand, which allows rapid labeling of Halo-tagged proteins in live cells and their efficient separation using streptavidin pull-down. Our work outlines how to use the herein-developed affinity ligand in sequential labeling to biochemically separate distinct protein variants and study their biological properties. The approach holds immense potential for addressing fundamental questions concerning essential cellular processes, including genome duplication and chromatin maintenance.","PeriodicalId":501590,"journal":{"name":"bioRxiv - Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265191","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Beta cell extracellular vesicle PD-L1 as a novel regulator of CD8+ T cell activity and biomarker during the evolution of Type 1 Diabetes β细胞胞外囊泡 PD-L1 是 CD8+ T 细胞活性的新型调节因子和 1 型糖尿病演变过程中的生物标记物
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.18.613649
Chaitra Rao, Daniel T Cater, Saptarshi Roy, Jerry Xu, Andre De G Olivera, Carmella Evans-Molina, Decio L Eizirik, Jon D Piganelli, Raghavendra Mirmira, Emily Kristen Sims
Aims/hypothesis: Surviving beta cells in type 1 diabetes respond to inflammation by upregulating programmed death-ligand 1 (PD-L1) to engage immune cell programmed death-1 (PD-1) and limit destruction by self-reactive immune cells. Extracellular vesicles (EVs) and their cargo can serve as biomarkers of beta cell health and contribute to islet intercellular communication. We hypothesized that the inflammatory milieu of type 1 diabetes increases PD-L1 in beta cell EV cargo and that EV PD-L1 may protect beta cells against immune-mediated cell death.Methods: Beta cell lines and human islets were treated with proinflammatory cytokines to model the proinflammatory type 1 diabetes microenvironment. EVs were isolated using ultracentrifugation or size exclusion chromatography and analysed via immunoblot, flow cytometry, and ELISA. EV PD-L1: PD-1 binding was assessed using a competitive binding assay and in vitro functional assays testing the ability of EV PD-L1 to inhibit NOD CD8 T cells. Plasma EV and soluble PD-L1 were assayed in plasma of individuals with islet autoantibody positivity (Ab+) or recent-onset type 1 diabetes and compared to non-diabetic controls.Results: PD-L1 protein colocalized with tetraspanin-associated proteins intracellularly and was detected on the surface of beta cell EVs. 24-h IFN-α or IFN-ɣ treatment induced a two-fold increase in EV PD-L1 cargo without a corresponding increase in number of EVs. IFN exposure predominantly increased PD-L1 expression on the surface of beta cell EVs and beta cell EV PD-L1 showed a dose-dependent capacity to bind PD-1. Functional experiments demonstrated specific effects of beta cell EV PD-L1 to suppress proliferation and cytotoxicity of murine CD8 T cells. Plasma EV PD-L1 levels were increased in islet Ab+ individuals, particularly in those with single Ab+, Additionally, in from individuals with either Ab+ or type 1 diabetes, but not in controls, plasma EV PD-L1 positively correlated with circulating C-peptide, suggesting that higher EV-PD-L1 could be protective for residual beta cell function. Conclusions/interpretation: IFN exposure increases PD-L1 on the beta cell EV surface. Beta cell EV PD-L1 binds PD1 and inhibits CD8 T cell proliferation and cytotoxicity. Circulating EV PD-L1 is higher in islet autoantibody positive patients compared to controls. Circulating EV PD-L1 levels correlate with residual C-peptide at different stages in type 1 diabetes progression. These findings suggest that EV PD-L1 could contribute to heterogeneity in type 1 diabetes progression and residual beta cell function and raise the possibility that EV PD-L1 could be exploited as a means to inhibit immune-mediated beta cell death.
目的/假设:1型糖尿病患者中存活的β细胞会通过上调程序性死亡配体1(PD-L1)来应对炎症,从而与免疫细胞程序性死亡-1(PD-1)结合,限制自我反应免疫细胞的破坏。细胞外囊泡 (EV) 及其载体可作为β细胞健康的生物标志物,并有助于胰岛细胞间的交流。我们假设,1 型糖尿病的炎症环境会增加贝塔细胞 EV 货物中的 PD-L1,而 EV PD-L1 可保护贝塔细胞免受免疫介导的细胞死亡:方法:用促炎细胞因子处理β细胞系和人类胰岛,以模拟促炎的1型糖尿病微环境。使用超速离心法或尺寸排阻色谱法分离 EV,并通过免疫印迹、流式细胞术和 ELISA 进行分析。使用竞争性结合试验和体外功能试验评估了 EV PD-L1:PD-1 的结合情况,测试 EV PD-L1 抑制 NOD CD8 T 细胞的能力。对胰岛自身抗体阳性(Ab+)或近期发病的1型糖尿病患者血浆中的EV和可溶性PD-L1进行了检测,并与非糖尿病对照组进行了比较:结果:PD-L1蛋白在细胞内与四泛素相关蛋白共定位,并在β细胞EV表面被检测到。24小时IFN-α或IFN-ɣ处理诱导的EV PD-L1货物增加了两倍,但EV数量没有相应增加。IFN暴露主要增加了β细胞EV表面的PD-L1表达,β细胞EV PD-L1与PD-1的结合能力呈剂量依赖性。功能实验证明,β细胞 EV PD-L1 有抑制小鼠 CD8 T 细胞增殖和细胞毒性的特异性作用。此外,在胰岛Ab+或1型糖尿病患者(而非对照组)中,血浆EV PD-L1水平与循环C肽呈正相关,这表明较高的EV-PD-L1水平可保护残留的β细胞功能。结论/解释:IFN暴露会增加β细胞EV表面的PD-L1。Beta 细胞 EV PD-L1 与 PD1 结合,抑制 CD8 T 细胞增殖和细胞毒性。与对照组相比,胰岛自身抗体阳性患者的循环EV PD-L1更高。在1型糖尿病进展的不同阶段,循环中的EV PD-L1水平与残留的C肽相关。这些研究结果表明,EV PD-L1 可导致 1 型糖尿病进展和残留β细胞功能的异质性,并提出了利用 EV PD-L1 抑制免疫介导的β细胞死亡的可能性。
{"title":"Beta cell extracellular vesicle PD-L1 as a novel regulator of CD8+ T cell activity and biomarker during the evolution of Type 1 Diabetes","authors":"Chaitra Rao, Daniel T Cater, Saptarshi Roy, Jerry Xu, Andre De G Olivera, Carmella Evans-Molina, Decio L Eizirik, Jon D Piganelli, Raghavendra Mirmira, Emily Kristen Sims","doi":"10.1101/2024.09.18.613649","DOIUrl":"https://doi.org/10.1101/2024.09.18.613649","url":null,"abstract":"Aims/hypothesis: Surviving beta cells in type 1 diabetes respond to inflammation by upregulating programmed death-ligand 1 (PD-L1) to engage immune cell programmed death-1 (PD-1) and limit destruction by self-reactive immune cells. Extracellular vesicles (EVs) and their cargo can serve as biomarkers of beta cell health and contribute to islet intercellular communication. We hypothesized that the inflammatory milieu of type 1 diabetes increases PD-L1 in beta cell EV cargo and that EV PD-L1 may protect beta cells against immune-mediated cell death.\u0000Methods: Beta cell lines and human islets were treated with proinflammatory cytokines to model the proinflammatory type 1 diabetes microenvironment. EVs were isolated using ultracentrifugation or size exclusion chromatography and analysed via immunoblot, flow cytometry, and ELISA. EV PD-L1: PD-1 binding was assessed using a competitive binding assay and in vitro functional assays testing the ability of EV PD-L1 to inhibit NOD CD8 T cells. Plasma EV and soluble PD-L1 were assayed in plasma of individuals with islet autoantibody positivity (Ab+) or recent-onset type 1 diabetes and compared to non-diabetic controls.\u0000Results: PD-L1 protein colocalized with tetraspanin-associated proteins intracellularly and was detected on the surface of beta cell EVs. 24-h IFN-α or IFN-ɣ treatment induced a two-fold increase in EV PD-L1 cargo without a corresponding increase in number of EVs. IFN exposure predominantly increased PD-L1 expression on the surface of beta cell EVs and beta cell EV PD-L1 showed a dose-dependent capacity to bind PD-1. Functional experiments demonstrated specific effects of beta cell EV PD-L1 to suppress proliferation and cytotoxicity of murine CD8 T cells. Plasma EV PD-L1 levels were increased in islet Ab+ individuals, particularly in those with single Ab+, Additionally, in from individuals with either Ab+ or type 1 diabetes, but not in controls, plasma EV PD-L1 positively correlated with circulating C-peptide, suggesting that higher EV-PD-L1 could be protective for residual beta cell function. Conclusions/interpretation: IFN exposure increases PD-L1 on the beta cell EV surface. Beta cell EV PD-L1 binds PD1 and inhibits CD8 T cell proliferation and cytotoxicity. Circulating EV PD-L1 is higher in islet autoantibody positive patients compared to controls. Circulating EV PD-L1 levels correlate with residual C-peptide at different stages in type 1 diabetes progression. These findings suggest that EV PD-L1 could contribute to heterogeneity in type 1 diabetes progression and residual beta cell function and raise the possibility that EV PD-L1 could be exploited as a means to inhibit immune-mediated beta cell death.","PeriodicalId":501590,"journal":{"name":"bioRxiv - Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265188","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Substrate-specific regulation of mTORC1 activity by G protein-coupled receptors G 蛋白偶联受体对 mTORC1 活性的底物特异性调控
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.18.613687
Samuel J. Atkinson, William Vincent Ritchie, Kyle Thompson, Dawn Thompson, James N. Hislop, Riko Hatakeyama
The mammalian/mechanistic Target of Rapamycin Complex 1 (mTORC1) kinase controls cell growth in response to various external stimuli. mTORC1 has a myriad of substrates, and the activation of distinct downstream pathways has important physiological outcomes. Emerging evidence suggests that mTORC1 can respond to upstream signals in a nuanced manner, differentially regulating individual substrates and downstream biological processes. The nature of signals that determine the signaling selectivity of mTORC1 is not fully understood. Here, we studied mTORC1 regulation by G protein-coupled receptors (GPCRs). We found that muscarinic acetylcholine receptor M5, formyl peptide receptor 2, and beta-2 adrenergic receptor, which are coupled to distinct effector G proteins, all trigger substrate-specific activation of mTORC1. Remarkably, phosphorylation of the TFEB transcription factor, a non-canonical mTORC1 substrate that controls lysosome biogenesis, responded to GPCRs differently, sometimes even oppositely, compared to canonical mTORC1 substrates such as S6K1 and 4EBP1. This study highlights the need to re-evaluate the effects of GPCRs on mTORC1 by parallelly monitoring individual substrates, an important consideration to be made when assessing GPCR ligands as therapeutic tools to manipulate the mTORC1 pathway.
哺乳动物/雷帕霉素机械靶标复合体 1(mTORC1)激酶控制细胞生长以应对各种外部刺激。新的证据表明,mTORC1 能以微妙的方式对上游信号做出反应,对单个底物和下游生物过程进行不同的调节。目前还不完全清楚决定 mTORC1 信号选择性的信号的性质。在这里,我们研究了 G 蛋白偶联受体(GPCR)对 mTORC1 的调控。我们发现,毒蕈碱乙酰胆碱受体 M5、甲酰肽受体 2 和 beta-2 肾上腺素能受体与不同的效应 G 蛋白偶联,都会触发 mTORC1 的底物特异性激活。值得注意的是,与 S6K1 和 4EBP1 等标准 mTORC1 底物相比,TFEB 转录因子(一种控制溶酶体生物生成的非标准 mTORC1 底物)对 GPCR 的磷酸化反应不同,有时甚至相反。这项研究强调了通过平行监测单个底物来重新评估 GPCR 对 mTORC1 的影响的必要性,这是在评估 GPCR 配体作为操纵 mTORC1 通路的治疗工具时需要考虑的一个重要因素。
{"title":"Substrate-specific regulation of mTORC1 activity by G protein-coupled receptors","authors":"Samuel J. Atkinson, William Vincent Ritchie, Kyle Thompson, Dawn Thompson, James N. Hislop, Riko Hatakeyama","doi":"10.1101/2024.09.18.613687","DOIUrl":"https://doi.org/10.1101/2024.09.18.613687","url":null,"abstract":"The mammalian/mechanistic Target of Rapamycin Complex 1 (mTORC1) kinase controls cell growth in response to various external stimuli. mTORC1 has a myriad of substrates, and the activation of distinct downstream pathways has important physiological outcomes. Emerging evidence suggests that mTORC1 can respond to upstream signals in a nuanced manner, differentially regulating individual substrates and downstream biological processes. The nature of signals that determine the signaling selectivity of mTORC1 is not fully understood. Here, we studied mTORC1 regulation by G protein-coupled receptors (GPCRs). We found that muscarinic acetylcholine receptor M5, formyl peptide receptor 2, and beta-2 adrenergic receptor, which are coupled to distinct effector G proteins, all trigger substrate-specific activation of mTORC1. Remarkably, phosphorylation of the TFEB transcription factor, a non-canonical mTORC1 substrate that controls lysosome biogenesis, responded to GPCRs differently, sometimes even oppositely, compared to canonical mTORC1 substrates such as S6K1 and 4EBP1. This study highlights the need to re-evaluate the effects of GPCRs on mTORC1 by parallelly monitoring individual substrates, an important consideration to be made when assessing GPCR ligands as therapeutic tools to manipulate the mTORC1 pathway.","PeriodicalId":501590,"journal":{"name":"bioRxiv - Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265193","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Uncovering the mechanistic basis of intracellular Raf inhibitor sensitivity reveals synergistic cotreatment strategies 揭示细胞内 Raf 抑制剂敏感性的机理基础,揭示协同处理策略
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.18.613772
Ethan G Stoddard, Linglan Fang, Yuhao Zhong, Zachary E Potter, Daniel S Brush, Jessica J Simon, Martin Golkowski, Dustin J Maly
Raf kinases are crucial effectors in the Ras-Raf-Mek-Erk signaling pathway, making them important targets for the development of cancer therapeutics. This study investigates the variable potency of DFG-out-stabilizing Raf inhibitors in mutant KRas-expressing cell lines. We demonstrate that inhibitor potency correlates with basal Raf activity, with more active Raf being more susceptible to inhibition. We further show that DFG-out-stabilizing inhibitors disrupt high-affinity Raf-Mek interactions, promoting the formation of inhibited Raf dimers. Furthermore, we identify cobimetinib as a Mek inhibitor that uniquely sensitizes Raf to DFG-out inhibitors by disrupting autoinhibited Raf-Mek complexes. Building on this insight, we developed cobimetinib analogs with enhanced sensitization properties. Our findings provide a mechanistic framework for understanding the cellular determinants of DFG-out-stabilizing inhibitor sensitivity and offer strategies for optimizing synergistic Raf-Mek inhibitor combinations.
Raf 激酶是 Ras-Raf-Mek-Erk 信号通路中的关键效应因子,因此成为开发癌症疗法的重要靶点。本研究调查了DFG-出稳定Raf抑制剂在突变KRas表达细胞系中的不同效力。我们证明,抑制剂的效力与基础 Raf 活性相关,活性越高的 Raf 越容易受到抑制。我们进一步证明,DFG-出稳定抑制剂会破坏高亲和力的 Raf-Mek 相互作用,促进被抑制的 Raf 二聚体的形成。此外,我们发现 cobimetinib 是一种 Mek 抑制剂,它通过破坏自动抑制的 Raf-Mek 复合物,使 Raf 对 DFG-out 抑制剂独特地敏感。基于这一认识,我们开发出了具有更强增敏特性的氯贝替尼类似物。我们的发现为了解细胞对 DFG-out 稳定抑制剂敏感性的决定因素提供了一个机理框架,并为优化 Raf-Mek 抑制剂的协同组合提供了策略。
{"title":"Uncovering the mechanistic basis of intracellular Raf inhibitor sensitivity reveals synergistic cotreatment strategies","authors":"Ethan G Stoddard, Linglan Fang, Yuhao Zhong, Zachary E Potter, Daniel S Brush, Jessica J Simon, Martin Golkowski, Dustin J Maly","doi":"10.1101/2024.09.18.613772","DOIUrl":"https://doi.org/10.1101/2024.09.18.613772","url":null,"abstract":"Raf kinases are crucial effectors in the Ras-Raf-Mek-Erk signaling pathway, making them important targets for the development of cancer therapeutics. This study investigates the variable potency of DFG-out-stabilizing Raf inhibitors in mutant KRas-expressing cell lines. We demonstrate that inhibitor potency correlates with basal Raf activity, with more active Raf being more susceptible to inhibition. We further show that DFG-out-stabilizing inhibitors disrupt high-affinity Raf-Mek interactions, promoting the formation of inhibited Raf dimers. Furthermore, we identify cobimetinib as a Mek inhibitor that uniquely sensitizes Raf to DFG-out inhibitors by disrupting autoinhibited Raf-Mek complexes. Building on this insight, we developed cobimetinib analogs with enhanced sensitization properties. Our findings provide a mechanistic framework for understanding the cellular determinants of DFG-out-stabilizing inhibitor sensitivity and offer strategies for optimizing synergistic Raf-Mek inhibitor combinations.","PeriodicalId":501590,"journal":{"name":"bioRxiv - Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265196","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Red Fluorescent Lifeact Marker to Study Actin Morphology in Podocytes 研究荚膜细胞肌动蛋白形态的红色荧光生命活动标记物
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.17.613449
Eva Wiesner, Julia Binz-Lotter, Simon E. Troeder, David Unnersjoe-Jess, Nelli Rutkowski, Branko Zevnik, Thomas Benzing, Roland Wedlich-Soldner, Matthias J. Hackl
F-actin is a major component of the cellular cytoskeleton, responsible for maintaining cell shape, enabling movement and facilitating intracellular transport. In the kidney, glomerular podocytes are highly dependent on their actin cytoskeleton shaping their unique foot processes. Hereditary mutations in actin-binding proteins cause focal segmental glomerulosclerosis, while other organs remain largely unaffected.So far, actin visualization in podocytes has been limited to electron microscopy or indirect immunofluorescent labeling of actin-binding proteins. However, the short F-actin-binding peptide Lifeact enables researchers to study actin dynamics in vitro and in vivo with minimal interference with actin metabolism.Here we introduce a new mouse model with conditional expression of a Lifeact.mScarlet I fusion protein providing red labeling of actin. Cre recombinase-mediated activity allows cell-specific and mosaic expression in podocytes, enabling selective labeling of individual cells to contrast with non-expressing neighboring cells. Transgenic mice are born healthy and young animals display no kidney-related phenotype. By intravital imaging and super-resolution microscopy, we show subcellular localization of actin to the foot processes in a resolution previously only obtainable by electron microscopy. Our novel mouse line provides the opportunity to study the actin cytoskeleton in podocytes and other cell types by intravital imaging and other conventional light microscopy techniques.
肌动蛋白是细胞细胞骨架的主要组成部分,负责维持细胞形状、促进运动和细胞内运输。在肾脏中,肾小球荚膜细胞高度依赖肌动蛋白细胞骨架塑造其独特的足部过程。肌动蛋白结合蛋白的遗传突变会导致局灶节段性肾小球硬化,而其他器官则基本不受影响。迄今为止,荚膜细胞中肌动蛋白的可视化仅限于电子显微镜或肌动蛋白结合蛋白的间接免疫荧光标记。然而,短的 F-肌动蛋白结合肽 Lifeact 使研究人员能够在体外和体内研究肌动蛋白的动态,而对肌动蛋白代谢的干扰最小。Cre 重组酶介导的活性允许在荚膜细胞中进行细胞特异性和镶嵌表达,使单个细胞的选择性标记与邻近的非表达细胞形成对比。转基因小鼠出生时非常健康,幼鼠也没有表现出与肾脏有关的表型。通过眼内成像和超分辨率显微镜,我们以以前只能通过电子显微镜获得的分辨率显示了肌动蛋白在足突的亚细胞定位。我们的新型小鼠品系为通过体内成像和其他传统光镜技术研究荚膜细胞和其他细胞类型的肌动蛋白细胞骨架提供了机会。
{"title":"A Red Fluorescent Lifeact Marker to Study Actin Morphology in Podocytes","authors":"Eva Wiesner, Julia Binz-Lotter, Simon E. Troeder, David Unnersjoe-Jess, Nelli Rutkowski, Branko Zevnik, Thomas Benzing, Roland Wedlich-Soldner, Matthias J. Hackl","doi":"10.1101/2024.09.17.613449","DOIUrl":"https://doi.org/10.1101/2024.09.17.613449","url":null,"abstract":"F-actin is a major component of the cellular cytoskeleton, responsible for maintaining cell shape, enabling movement and facilitating intracellular transport. In the kidney, glomerular podocytes are highly dependent on their actin cytoskeleton shaping their unique foot processes. Hereditary mutations in actin-binding proteins cause focal segmental glomerulosclerosis, while other organs remain largely unaffected.\u0000So far, actin visualization in podocytes has been limited to electron microscopy or indirect immunofluorescent labeling of actin-binding proteins. However, the short F-actin-binding peptide Lifeact enables researchers to study actin dynamics in vitro and in vivo with minimal interference with actin metabolism.\u0000Here we introduce a new mouse model with conditional expression of a Lifeact.mScarlet I fusion protein providing red labeling of actin. Cre recombinase-mediated activity allows cell-specific and mosaic expression in podocytes, enabling selective labeling of individual cells to contrast with non-expressing neighboring cells. Transgenic mice are born healthy and young animals display no kidney-related phenotype. By intravital imaging and super-resolution microscopy, we show subcellular localization of actin to the foot processes in a resolution previously only obtainable by electron microscopy. Our novel mouse line provides the opportunity to study the actin cytoskeleton in podocytes and other cell types by intravital imaging and other conventional light microscopy techniques.","PeriodicalId":501590,"journal":{"name":"bioRxiv - Cell Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142265200","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
bioRxiv - Cell Biology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1