border cell migration

  • 文章类型: Journal Article
    蛋白质毒性应激驱动许多退行性疾病。细胞最初通过激活未折叠的蛋白质反应(UPR)来适应错误折叠的蛋白质,包括内质网相关蛋白降解(ERAD)。然而,持续的压力触发细胞凋亡。增强ERAD是蛋白质错误折叠疾病的有希望的治疗方法。ER定位的Zn2转运蛋白ZIP7从植物到人类是保守的,并且是肠道自我更新所必需的,陷波信号,细胞运动性,和生存。然而,这些不同表型的统一机制尚不清楚.在研究果蝇边缘细胞迁移时,我们发现ZIP7介导的Zn2+转运通过蛋白酶体盖中的Rpn11Zn2+金属蛋白酶增强蛋白质的强制性去泛素化。在人类细胞中,ZIP7和Zn2+限制了去泛素化。在由错误折叠的视紫红质(Rh1)引起的果蝇神经变性模型中,ZIP7过表达降解错误折叠的Rh1并挽救光感受器活力和飞行视力。因此,ZIP7介导的Zn2+转运是以前未知的,在蛋白质错误折叠疾病中具有治疗潜力的体内ERAD的限速步骤。
    Proteotoxic stress drives numerous degenerative diseases. Cells initially adapt to misfolded proteins by activating the unfolded protein response (UPR), including endoplasmic-reticulum-associated protein degradation (ERAD). However, persistent stress triggers apoptosis. Enhancing ERAD is a promising therapeutic approach for protein misfolding diseases. The ER-localized Zn2+ transporter ZIP7 is conserved from plants to humans and required for intestinal self-renewal, Notch signaling, cell motility, and survival. However, a unifying mechanism underlying these diverse phenotypes was unknown. In studying Drosophila border cell migration, we discovered that ZIP7-mediated Zn2+ transport enhances the obligatory deubiquitination of proteins by the Rpn11 Zn2+ metalloproteinase in the proteasome lid. In human cells, ZIP7 and Zn2+ are limiting for deubiquitination. In a Drosophila model of neurodegeneration caused by misfolded rhodopsin (Rh1), ZIP7 overexpression degrades misfolded Rh1 and rescues photoreceptor viability and fly vision. Thus, ZIP7-mediated Zn2+ transport is a previously unknown, rate-limiting step for ERAD in vivo with therapeutic potential in protein misfolding diseases.
    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
    磷脂酰肌醇(PI)4,5-二磷酸(PIP2)参与许多生物学功能。然而,PIP2在集体细胞迁移中的机制仍然难以捉摸。本研究通过调节PIP2的不对称分布,强调了三磷酸胞苷合酶(CTPsyn)在集体边界细胞迁移中的调节作用。我们证明了含有突变CTPsyn细胞的边界细胞簇抑制了迁移。CTPsyn在果蝇边缘细胞簇的前缘与肌动蛋白共富集,其中PIP2富集,这种富集取决于CTPsyn的活性。在CTPsyn突变体与PI生物合成基因之间发现了边界细胞迁移的遗传相互作用。CTPsyn减少导致胞吞再循环的不对称活性丧失。此外,揭示了外囊复合体和CTPsyn突变体的成分之间的遗传相互作用,表明CTPsyn活性调节PIP2相关的不对称胞吐活性。此外,CTPsyn活性对于边缘细胞簇中的RTK极化分布至关重要。我们提出了一个模型,在该模型中,PIP2的不对称生成需要CTPsyn活性,以通过集体细胞迁移中的内吞再循环来丰富RTK信号传导。
    Phosphatidylinositol (PI) 4,5-bisphosphate (PIP2) is involved in many biological functions. However, the mechanisms of PIP2 in collective cell migration remain elusive. This study highlights the regulatory role of cytidine triphosphate synthase (CTPsyn) in collective border cell migration through regulating the asymmetrical distribution of PIP2. We demonstrated that border cell clusters containing mutant CTPsyn cells suppressed migration. CTPsyn was co-enriched with Actin at the leading edge of the Drosophila border cell cluster where PIP2 was enriched, and this enrichment depended on the CTPsyn activity. Genetic interactions of border cell migration were found between CTPsyn mutant and genes in PI biosynthesis. The CTPsyn reduction resulted in loss of the asymmetric activity of endocytosis recycling. Also, genetic interactions were revealed between components of the exocyst complex and CTPsyn mutant, indicating that CTPsyn activity regulates the PIP2-related asymmetrical exocytosis activity. Furthermore, CTPsyn activity is essential for RTK-polarized distribution in the border cell cluster. We propose a model in which CTPsyn activity is required for the asymmetrical generation of PIP2 to enrich RTK signaling through endocytic recycling in collective cell migration.
    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
    细胞迁移是形态发生所必需的,组织稳态,伤口愈合和免疫反应。它也与疾病有关。特别是,细胞迁移是转移固有的。细胞可以单独或成组迁移。要高效迁移,细胞需要能够组织成通过形成膜延伸而突出的前前缘和收缩的后缘。在集体细胞运动期间,该组织在小组一级扩大。如果一个单元或一组单元不能限制其前缘,从而限制前部突起的形成,定向运动受损或被废除。在这里,我们总结了我们目前对集体细胞迁移中限制突起形成的机制的理解。我们集中在三个体内例子:神经c细胞迁移,果蝇卵室周围卵泡细胞的旋转迁移和卵子发生过程中的边界细胞迁移。
    Cell migration is necessary for morphogenesis, tissue homeostasis, wound healing and immune response. It is also involved in diseases. In particular, cell migration is inherent in metastasis. Cells can migrate individually or in groups. To migrate efficiently, cells need to be able to organize into a leading front that protrudes by forming membrane extensions and a trailing edge that contracts. This organization is scaled up at the group level during collective cell movements. If a cell or a group of cells is unable to limit its leading edge and hence to restrict the formation of protrusions to the front, directional movements are impaired or abrogated. Here we summarize our current understanding of the mechanisms restricting protrusion formation in collective cell migration. We focus on three in vivo examples: the neural crest cell migration, the rotatory migration of follicle cells around the Drosophila egg chamber and the border cell migration during oogenesis.
    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
    细胞迁移组中的不对称性对于在正常发育和病理条件如肿瘤细胞转移中观察到的有效定向运动至关重要。这在极化突起和各种极性蛋白质在集体移动簇中的差异定位的水平上被明显地检测到。多年来,果蝇卵子发生中的边界细胞迁移已成为研究细胞迁移组中极性的出色模型系统。在这里,我们报告了两种方案,采用活细胞成像和组织免疫组织化学来评估迁移边界细胞簇的极性。
    Asymmetry in the migrating group of cells is critical for efficient directed movement observed in normal development and in pathological conditions like tumor cell metastasis. This is conspicuously detected at the level of polarized protrusions and differential localization of various polarity proteins in collectively moving clusters. Over the years, border cell migration in Drosophila oogenesis has emerged as an excellent model system for studying polarity in the migrating group of cells. Here we report two protocols employing live cell imaging and tissue immunohistochemistry to evaluate the polarity in migrating border cell clusters.
    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
    Cell migration is a key component in development, homeostasis, immune function, and pathology. It is important to understand the molecular activity that allows some cells to migrate. Drosophila melanogaster is a useful model system because its genes are largely conserved with humans and it is straightforward to study biologically. The well-conserved transcriptional regulator Signal Transducer and Activator of Transcription (STAT) promotes cell migration, but its signaling is modulated by downstream targets Apontic (APT) and Slow Border Cells (SLBO). Inhibition of STAT activity by APT and cross-repression of APT and SLBO determines whether an epithelial cell in the Drosophila egg chamber becomes motile or remains stationary. Through mathematical modeling and analysis, we examine how the interaction of STAT, APT, and SLBO creates bistability in the Janus Kinase (JAK)/STAT signaling pathway. In this paper, we update and analyze earlier models to represent mechanistically the processes of the JAK/STAT pathway. We utilize parameter, bifurcation, and phase portrait analyses, and make reductions to the system to produce a minimal three-variable quantitative model. We analyze the manifold between migratory and stationary steady states in this minimal model and show that when the initial conditions of our model are near this manifold, cell migration can be delayed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Collective cell migration is involved in various developmental and pathological processes, including the dissemination of various cancer cells. During Drosophila melanogaster oogenesis, a group of cells called border cells migrate collectively toward the oocyte. Herein, we show that members of the Arf family of small GTPases and some of their regulators are required for normal border cell migration. Notably, we found that the ArfGAP Drongo and its GTPase-activating function are essential for the initial detachment of the border cell cluster from the basal lamina. We demonstrate through protein localization and genetic interactions that Drongo controls the localization of the myosin phosphatase in order to regulate myosin II activity at the back of the cluster. Moreover, we show that toward the class III Arf, Drongo acts antagonistically to the guanine exchange factor Steppke. Overall, our work describes a mechanistic pathway that promotes the local actomyosin contractility necessary for border cell detachment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    由于集体细胞迁移与后生动物发育的不同方面密切相关,这一过程背后的分子机制正在各种发展环境中进行探索。果蝇卵子发生过程中的边界细胞(BC)迁移已成为研究集体细胞迁移的出色遗传模型。BCs是上皮起源的,但在作为一组向卵母细胞迁移之前获得部分间充质特征。这里,我们报道了胰岛素信号调节果蝇卵子发生过程中的集体BC运动。支持胰岛素途径的参与,我们证明了BCs中胰岛素样受体(InR)水平的下降,抑制他们的迁移。此外,我们证明了典型的胰岛素信号通路成分参与了这一过程。有趣的是,InR耗尽的BC簇的可视化,使用延时成像,显示BC簇与周围前卵泡细胞的脱离延迟,并改变了突起动力学。最后,基于InR之间的遗传相互作用,极性决定因素,par-1和果蝇肌球蛋白(意大利南瓜)的调节亚基,我们认为胰岛素信号传导可能通过果蝇肌球蛋白影响par-1活性,从而设计边界细胞脱离和随后的运动。
    As collective cell migration is intimately involved in different aspects of metazoan development, molecular mechanisms underlying this process are being explored in a variety of developmental contexts. Border cell (BC) migration during Drosophila oogenesis has emerged as an excellent genetic model for studying collective cell migration. BCs are of epithelial origin but acquire partial mesenchymal characteristics before migrating as a group towards the oocyte. Here, we report that insulin signaling modulates collective BC movement during Drosophila oogenesis. Supporting the involvement of Insulin pathway, we demonstrate that compromising Insulin-like Receptor (InR) levels in BCs, inhibits their migration. Furthermore, we show that canonical Insulin signaling pathway components participate in this process. Interestingly, visualization of InR-depleted BC clusters, using time-lapse imaging, revealed a delay in detachment of BC clusters from the surrounding anterior follicle cells and altered protrusion dynamics. Lastly, based on genetic interactions between InR, the polarity determinant, par-1 and a regulatory subunit of Drosophila Myosin (spaghetti squash), we propose that Insulin signaling likely influences par-1 activity to engineer border cell detachment and subsequent movement via Drosophila Myosin.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    The Suppressor of Cytokine Signaling (SOCS) proteins are critical, highly conserved feedback inhibitors of signal transduction cascades. The family of SOCS proteins is divided into two groups: ancestral and vertebrate-specific SOCS proteins. Vertebrate-specific SOCS proteins have been heavily studied as a result of their strong mutant phenotypes. However, the ancestral clade remains less studied, a potential result of genetic redundancies in mammals. Use of the genetically tractable organism Drosophila melanogaster enables in vivo assessment of signaling components and mechanisms with less concern about the functional redundancy observed in mammals. In this study, we investigated how the SOCS family member Suppressor of Cytokine Signaling at 36E (Socs36E) attenuates Janus Kinase/Signal Transducer and Activator of Transcription (Jak/STAT) activation during specification of motile border cells in Drosophila oogenesis. We found that Socs36E genetically interacts with the Cullin2 (Cul2) scaffolding protein. Like Socs36E, Cul2 is required to limit the number of motile cells in egg chambers. We demonstrated that loss of Cul2 in the follicle cells significantly increased nuclear STAT protein levels, which resulted in additional cells acquiring invasive properties. Further, reduction of Cul2 suppressed border cell migration defects that occur in a Stat92E-sensitized genetic background. Our data incorporated Cul2 into a previously described Jak/STAT-directed genetic regulatory network that is required to generate a discrete boundary between cell fates. We also found that Socs36E is able to attenuate STAT activity in the egg chamber when it does not have a functional SOCS box. Collectively, this work contributes mechanistic insight to a Jak/STAT regulatory genetic circuit, and suggests that Socs36E regulates Jak/STAT signaling via a Cul2-dependent mechanism, as well as by a Cullin-independent manner, in vivo.
    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
    The Hippo pathway is a key signaling cascade in controlling organ size. The core components of this pathway are two kinases, Hippo (Hpo) and Warts (Wts), and a transcriptional coactivator, Yorkie (Yki). Yes-associated protein (YAP, a Yki homolog in mammals) promotes epithelial-mesenchymal transition and cell migration in vitro. Here, we use border cells in the Drosophila ovary as a model to study Hippo pathway functions in cell migration in vivo. During oogenesis, polar cells secrete Unpaired (Upd), which activates JAK/STAT signaling of neighboring cells and specifies them into outer border cells. The outer border cells form a cluster with polar cells and undergo migration. We find that hpo and wts are required for migration of the border cell cluster. In outer border cells, overexpression of hpo disrupts polarization of the actin cytoskeleton and attenuates migration. In polar cells, knockdown of hpo and wts or overexpression of yki impairs border cell induction and disrupts migration. These manipulations in polar cells reduce JAK/STAT activity in outer border cells. Expression of upd-lacZ is increased and decreased in yki and hpo mutant polar cells, respectively. Furthermore, forced expression of upd in polar cells rescues defects of border cell induction and migration caused by wts knockdown. These results suggest that Yki negatively regulates border cell induction by inhibiting JAK/STAT signaling. Together, our data elucidate two distinct mechanisms of the Hippo pathway in controlling border cell migration: (1) in outer border cells, it regulates polarized distribution of the actin cytoskeleton; (2) in polar cells, it regulates upd expression to control border cell induction and migration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Considering the degree of detail available at the genetic and cellular levels, the Drosophila ovary stands out as a powerful system to identify new players in the regulation of key aspects of cancer progression. In this review, we will comment on how the use of the Drosophila ovary has helped to elucidate some of the molecular bases of ovarian malignancies and to identify and characterize critical tumour suppressor genes and oncogenes with an impact in human pathologies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号