首页 > 最新文献

European journal of cell biology最新文献

英文 中文
Exploring optimal protocols for generating and preserving glucose-responsive insulin-secreting progenitor cells derived from human pluripotent stem cells 探索生成和保存源自人类多能干细胞的葡萄糖反应型胰岛素分泌祖细胞的最佳方案
IF 4.5 3区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-10-24 DOI: 10.1016/j.ejcb.2024.151464
Human pluripotent stem cells (hPSCs) represent an unlimited source of β-like cells for both disease modeling and cellular therapy for diabetes. Numerous protocols have been published describing the differentiation of hPSCs into β-like cells that secret insulin in response to a glucose challenge. However, among the most widely used protocols it is not clear which yield the most functional cells with reproducible glucose-stimulated insulin-secretion (GSIS). Moreover, the technical challenges in culturing and differentiating hPSCs is a barrier for many researchers. In this study, we performed a side-by-side functional comparison based on three widely used methods to generate insulin expressing cells and identified optimal stages and conditions for cryopreserving and reconstituting stem cell (SC)-derived islets with a robust GSIS. Despite the fact that each protocol yields SC-islets consisting of insulin and glucagon-expressing cells, the SC-islets obtained from the two more recent revised protocols were more functional as measured by robust and reproducible GSIS. Moreover, we demonstrate that pancreatic progenitors and differentiated endocrine cells that have been cryopreserved for up to 10 months, can be reconstituted into glucose responsive SC-islets. These findings should enable the use of human PSC-derived β-like cells technologies even by groups with minimal PSC culture experience.
人类多能干细胞(hPSCs)是用于疾病建模和糖尿病细胞疗法的β样细胞的无限来源。已发表的大量方案描述了如何将hPSCs分化成β样细胞,在葡萄糖挑战下分泌胰岛素。然而,在最广泛使用的方案中,并不清楚哪种方案能产生功能最强、可重复的葡萄糖刺激胰岛素分泌(GSIS)细胞。此外,培养和分化 hPSCs 的技术难题也是许多研究人员面临的障碍。在这项研究中,我们根据三种广泛使用的生成胰岛素表达细胞的方法进行了并排功能比较,并确定了冷冻保存和重组干细胞(SC)衍生胰岛的最佳阶段和条件,使其具有强大的GSIS。尽管每种方案都能产生由胰岛素和胰高血糖素表达细胞组成的干细胞小体,但从稳健和可重复的GSIS衡量,从最近修订的两种方案中获得的干细胞小体功能更强。此外,我们还证明,冷冻保存长达 10 个月的胰腺祖细胞和分化的内分泌细胞可以重组为葡萄糖反应性 SC-小体。这些研究结果将有助于使用人类胰腺造血干细胞衍生的β样细胞技术,即使是没有多少胰腺造血干细胞培养经验的小组也能使用。
{"title":"Exploring optimal protocols for generating and preserving glucose-responsive insulin-secreting progenitor cells derived from human pluripotent stem cells","authors":"","doi":"10.1016/j.ejcb.2024.151464","DOIUrl":"10.1016/j.ejcb.2024.151464","url":null,"abstract":"<div><div>Human pluripotent stem cells (hPSCs) represent an unlimited source of β-like cells for both disease modeling and cellular therapy for diabetes. Numerous protocols have been published describing the differentiation of hPSCs into β-like cells that secret insulin in response to a glucose challenge. However, among the most widely used protocols it is not clear which yield the most functional cells with reproducible glucose-stimulated insulin-secretion (GSIS). Moreover, the technical challenges in culturing and differentiating hPSCs is a barrier for many researchers. In this study, we performed a side-by-side functional comparison based on three widely used methods to generate insulin expressing cells and identified optimal stages and conditions for cryopreserving and reconstituting stem cell (SC)-derived islets with a robust GSIS. Despite the fact that each protocol yields SC-islets consisting of insulin and glucagon-expressing cells, the SC-islets obtained from the two more recent revised protocols were more functional as measured by robust and reproducible GSIS. Moreover, we demonstrate that pancreatic progenitors and differentiated endocrine cells that have been cryopreserved for up to 10 months, can be reconstituted into glucose responsive SC-islets. These findings should enable the use of human PSC-derived β-like cells technologies even by groups with minimal PSC culture experience.</div></div>","PeriodicalId":12010,"journal":{"name":"European journal of cell biology","volume":null,"pages":null},"PeriodicalIF":4.5,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142561071","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
An unexpected role of IL10 in mesoderm induction and differentiation from pluripotent stem cells: Implications in zebrafish angiogenic sprouting, vascular organoid development, and therapeutic angiogenesis IL10在中胚层诱导和多能干细胞分化中的意外作用:斑马鱼血管萌发、血管器官发育和治疗性血管生成的意义
IF 4.5 3区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-10-24 DOI: 10.1016/j.ejcb.2024.151465
Mesoderm induction is a crucial step for vascular cell specification, vascular development and vasculogenesis. However, the cellular and molecular mechanisms underlying mesoderm induction remain elusive. In the present study, a chemically-defined differentiation protocol was used to induce mesoderm formation and generate functional vascular cells including smooth muscle cells (SMCs) and endothelial cells (ECs) from human induced pluripotent stem cells (hiPSCs). Zebrafish larvae were used to detect an in vivo function of interleukin 10 (IL10) in mesoderm formation and vascular development. A three dimensional approach was used to create hiPSC-derived blood vessel organoid (BVO) and explore a potential impact of IL10 on BVO formation. A murine model hind limb ischemia was applied to investigate a therapeutic potential of hiPSC-derived cells treated with or without IL10 during differentiation. We found that IL10 was significantly and specifically up-regulated during mesoderm stage of vascular differentiation. IL10 addition in mesoderm induction media dramatically increased mesoderm induction and vascular cell generation from hiPSCs, whereas an opposite effect was observed with IL10 inhibition. Mechanistic studies revealed that IL10 promotes mesoderm formation and vascular cell differentiation by activating signal transducer and activator of transcription 3 signal pathway. Functional studies with an in vivo model system confirmed that knockdown of IL10 using morpholino antisense oligonucleotides in zebrafish larvae caused defective mesoderm formation, angiogenic sprouting and vascular development. Additionally, our data also show IL10 promotes blood vessel organoid development and enhances vasculogenesis and angiogenesis. Importantly, we demonstrate that IL10 treatment during mesoderm induction stage enhances blood flow perfusion recovery and increases vasculogenesis and therapeutic angiogenesis after hind limb ischemia. Our data, therefore, demonstrate a regulatory role for IL10 in mesoderm formation from hiPSCs and during zebrafish vascular development, providing novel insights into mesoderm induction and vascular cell specifications.
中胚层诱导是血管细胞规格化、血管发育和血管生成的关键步骤。然而,中胚层诱导的细胞和分子机制仍然难以捉摸。本研究采用化学定义的分化方案诱导中胚层形成,并从人类诱导多能干细胞(hiPSCs)中生成包括平滑肌细胞(SMCs)和内皮细胞(ECs)在内的功能性血管细胞。斑马鱼幼体被用来检测白细胞介素10(IL10)在体内中胚层形成和血管发育中的功能。研究人员采用三维方法创建了源自 hiPSC 的血管器官(BVO),并探索了 IL10 对 BVO 形成的潜在影响。应用小鼠后肢缺血模型研究了分化过程中用或不用IL10处理hiPSC衍生细胞的治疗潜力。我们发现,在血管分化的中胚层阶段,IL10明显且特异性地上调。在中胚层诱导培养基中添加IL10可显著增加中胚层诱导和hiPSCs血管细胞的生成,而抑制IL10则会产生相反的效果。机理研究发现,IL10通过激活信号转导子和转录激活子3信号通路促进中胚层形成和血管细胞分化。体内模型系统的功能研究证实,在斑马鱼幼体中使用吗啉诺反义寡核苷酸敲除IL10会导致中胚层形成、血管萌发和血管发育缺陷。此外,我们的数据还显示,IL10 能促进血管器官发育,增强血管生成和血管形成。重要的是,我们证明了在中胚层诱导阶段处理 IL10 能增强后肢缺血后的血流灌注恢复,增加血管生成和治疗性血管生成。因此,我们的数据证明了IL10在hiPSCs中胚层形成和斑马鱼血管发育过程中的调控作用,为中胚层诱导和血管细胞规格提供了新的见解。
{"title":"An unexpected role of IL10 in mesoderm induction and differentiation from pluripotent stem cells: Implications in zebrafish angiogenic sprouting, vascular organoid development, and therapeutic angiogenesis","authors":"","doi":"10.1016/j.ejcb.2024.151465","DOIUrl":"10.1016/j.ejcb.2024.151465","url":null,"abstract":"<div><div>Mesoderm induction is a crucial step for vascular cell specification, vascular development and vasculogenesis. However, the cellular and molecular mechanisms underlying mesoderm induction remain elusive. In the present study, a chemically-defined differentiation protocol was used to induce mesoderm formation and generate functional vascular cells including smooth muscle cells (SMCs) and endothelial cells (ECs) from human induced pluripotent stem cells (hiPSCs). Zebrafish larvae were used to detect an in vivo function of interleukin 10 (IL10) in mesoderm formation and vascular development. A three dimensional approach was used to create hiPSC-derived blood vessel organoid (BVO) and explore a potential impact of IL10 on BVO formation. A murine model hind limb ischemia was applied to investigate a therapeutic potential of hiPSC-derived cells treated with or without IL10 during differentiation. We found that IL10 was significantly and specifically up-regulated during mesoderm stage of vascular differentiation. IL10 addition in mesoderm induction media dramatically increased mesoderm induction and vascular cell generation from hiPSCs, whereas an opposite effect was observed with IL10 inhibition. Mechanistic studies revealed that IL10 promotes mesoderm formation and vascular cell differentiation by activating signal transducer and activator of transcription 3 signal pathway. Functional studies with an <em>in vivo</em> model system confirmed that knockdown of IL10 using morpholino antisense oligonucleotides in zebrafish larvae caused defective mesoderm formation, angiogenic sprouting and vascular development. Additionally, our data also show IL10 promotes blood vessel organoid development and enhances vasculogenesis and angiogenesis. Importantly, we demonstrate that IL10 treatment during mesoderm induction stage enhances blood flow perfusion recovery and increases vasculogenesis and therapeutic angiogenesis after hind limb ischemia. Our data, therefore, demonstrate a regulatory role for IL10 in mesoderm formation from hiPSCs and during zebrafish vascular development, providing novel insights into mesoderm induction and vascular cell specifications.</div></div>","PeriodicalId":12010,"journal":{"name":"European journal of cell biology","volume":null,"pages":null},"PeriodicalIF":4.5,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142544577","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Editorial overview: EJCB Special Issue - Cell host-pathogen interactions. 编辑综述:EJCB 特刊--细胞宿主与病原体之间的相互作用。
IF 4.5 3区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-10-18 DOI: 10.1016/j.ejcb.2024.151462
Serge Mostowy, Theresia E B Stradal
{"title":"Editorial overview: EJCB Special Issue - Cell host-pathogen interactions.","authors":"Serge Mostowy, Theresia E B Stradal","doi":"10.1016/j.ejcb.2024.151462","DOIUrl":"https://doi.org/10.1016/j.ejcb.2024.151462","url":null,"abstract":"","PeriodicalId":12010,"journal":{"name":"European journal of cell biology","volume":null,"pages":null},"PeriodicalIF":4.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142497505","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
dCas9-HDAC8-EGFP fusion enables epigenetic editing of breast cancer cells by H3K9 deacetylation dCas9-HDAC8-EGFP 融合可通过 H3K9 去乙酰化对乳腺癌细胞进行表观遗传编辑。
IF 4.5 3区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-10-18 DOI: 10.1016/j.ejcb.2024.151463
Epigenetic editing is thriving as a robust tool for manipulating transcriptional regulation and cell fate. Despite its regulatory role in gene downregulation, epigenetic editing with histone deacetylation has been sparsely studied, especially in the context of cancer. In this current study, we have reconstructed a dCas9-HDAC8-EGFP fusion to perform histone deacetylation on the promoter of the ESR1, TERT and CDKN1C genes for the first time in breast cancer cell lines MCF-7 and MDA-MB-231 as well as in HEK293T cells. Our results demonstrated that dCas9-HDAC8-EGFP in combination with appropriate gRNAs were able to downregulate the expression of the ESR1, TERT and CDKN1C genes transcriptionally by specifically depleting the H3K9ac level on the recruitment loci. The addition of histone deacetylase inhibitors was found to neutralize the outcomes of dCas9-HDAC8-EGFP-induced epigenetic editing. Furthermore, we observed a significant downregulation of full length ERα expression in epigenetically edited MCF-7 cells with consequential alteration in cellular response toward estradiol and tamoxifen treatment due to dCas9-HDAC8-EGFP mediated epigenetic editing of the ESR1 gene. Overall, dCas9-HDAC8-EGFP is a novel circuit that enabled downregulation of crucial genes with cellular outcome in breast cancer cells by preferentially inducing H3K9 deacetylation of specific promoter regions.
表观遗传编辑作为操纵转录调控和细胞命运的有力工具正在蓬勃发展。尽管表观遗传编辑在基因下调中发挥着调控作用,但对组蛋白去乙酰化的研究却很少,尤其是在癌症方面。在本研究中,我们首次在乳腺癌细胞系 MCF-7 和 MDA-MB-231 以及 HEK293T 细胞中重建了 dCas9-HDAC8-EGFP 融合体,对 ESR1、TERT 和 CDKN1C 基因的启动子进行组蛋白去乙酰化。我们的研究结果表明,dCas9-HDAC8-EGFP 与适当的 gRNA 结合使用,能够通过特异性地消耗招募位点上的 H3K9ac 水平,转录下调 ESR1、TERT 和 CDKN1C 基因的表达。加入组蛋白去乙酰化酶抑制剂可中和 dCas9-HDAC8-EGFP 诱导的表观遗传编辑结果。此外,由于 dCas9-HDAC8-EGFP 介导了 ESR1 基因的表观遗传编辑,我们观察到表观遗传编辑的 MCF-7 细胞中 ERα 的全长表达明显下调,细胞对雌二醇和他莫昔芬处理的反应也随之改变。总之,dCas9-HDAC8-EGFP 是一种新型回路,它能通过优先诱导特定启动子区域的 H3K9 去乙酰化来下调乳腺癌细胞中影响细胞结果的关键基因。
{"title":"dCas9-HDAC8-EGFP fusion enables epigenetic editing of breast cancer cells by H3K9 deacetylation","authors":"","doi":"10.1016/j.ejcb.2024.151463","DOIUrl":"10.1016/j.ejcb.2024.151463","url":null,"abstract":"<div><div>Epigenetic editing is thriving as a robust tool for manipulating transcriptional regulation and cell fate. Despite its regulatory role in gene downregulation, epigenetic editing with histone deacetylation has been sparsely studied, especially in the context of cancer. In this current study, we have reconstructed a dCas9-HDAC8-EGFP fusion to perform histone deacetylation on the promoter of the <em>ESR1</em>, <em>TERT</em> and <em>CDKN1C</em> genes for the first time in breast cancer cell lines MCF-7 and MDA-MB-231 as well as in HEK293T cells. Our results demonstrated that dCas9-HDAC8-EGFP in combination with appropriate gRNAs were able to downregulate the expression of the <em>ESR1</em>, <em>TERT</em> and <em>CDKN1C</em> genes transcriptionally by specifically depleting the H3K9ac level on the recruitment loci. The addition of histone deacetylase inhibitors was found to neutralize the outcomes of dCas9-HDAC8-EGFP-induced epigenetic editing. Furthermore, we observed a significant downregulation of full length ERα expression in epigenetically edited MCF-7 cells with consequential alteration in cellular response toward estradiol and tamoxifen treatment due to dCas9-HDAC8-EGFP mediated epigenetic editing of the <em>ESR1</em> gene. Overall, dCas9-HDAC8-EGFP is a novel circuit that enabled downregulation of crucial genes with cellular outcome in breast cancer cells by preferentially inducing H3K9 deacetylation of specific promoter regions.</div></div>","PeriodicalId":12010,"journal":{"name":"European journal of cell biology","volume":null,"pages":null},"PeriodicalIF":4.5,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142497504","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The double deficiency of the SNARE proteins vti1a and vti1b affects neurite outgrowth and signaling in N1E-115 neuroblastoma cells. SNARE 蛋白 vti1a 和 vti1b 的双重缺乏会影响 N1E-115 神经母细胞瘤细胞的神经元生长和信号传导。
IF 4.5 3区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-10-09 DOI: 10.1016/j.ejcb.2024.151461
During intracellular trafficking N-ethylmaleimide-sensitive-factor attachment receptor (SNARE) proteins catalyze the membrane fusion by assembling into a four-helix complex. In the mouse model, loss of the endosomal SNAREs vti1a and vti1b results in a perinatal lethal phenotype and neuronal defects including decreased neurite outgrowth in cultured primary neurons.
We used a CRISPR/Cas9 system to generate a Vti1a Vti1b double knockout (DKO) in the neuroblastoma cell line N1E-115. Three different DKO cell lines were obtained and examined at genome and protein level. The double deficiency impaired proper differentiation based on lower levels of synaptic proteins as well as reduced neurite formation and elongation compared to wild type cells in differentiation medium. Neurite elongation can be induced by a variety of extracellular signals via different signaling cascades. Treatment with the Rho kinase inhibitor Y27632, which stimulates enlargeosome exocytosis, or with neurotrophic factors (BDNF, NGF and NT3) resulted in reduced stimulation of all DKO clones and in significantly shorter neurites compared to wild type cells. The loss of vti1a and vti1b disrupted Akt signaling during enlargeosome-mediated and Erk signaling during BDNF-induced neurite outgrowth.
在细胞内转运过程中,N-乙基马来酰亚胺敏感因子附着受体(SNARE)蛋白通过组装成一个四螺旋复合物来催化膜融合。我们利用 CRISPR/Cas9 系统在神经母细胞瘤细胞系 N1E-115 中产生了 Vti1a Vti1b 双基因敲除(DKO)。我们获得了三种不同的 DKO 细胞系,并对它们进行了基因组和蛋白质水平的检测。与分化培养基中的野生型细胞相比,双重基因缺失会降低突触蛋白的水平,并减少神经元的形成和伸长,从而影响细胞的正常分化。神经元的伸长可由多种细胞外信号通过不同的信号级联诱导。Rho激酶抑制剂Y27632可刺激 enlargeosome外渗,而神经营养因子(BDNF、NGF和NT3)则可减少对所有DKO克隆的刺激,与野生型细胞相比,神经元的长度也明显缩短。vti1a 和 vti1b 的缺失破坏了增大体介导的 Akt 信号转导和 BDNF 诱导的神经元生长过程中的 Erk 信号转导。
{"title":"The double deficiency of the SNARE proteins vti1a and vti1b affects neurite outgrowth and signaling in N1E-115 neuroblastoma cells.","authors":"","doi":"10.1016/j.ejcb.2024.151461","DOIUrl":"10.1016/j.ejcb.2024.151461","url":null,"abstract":"<div><div>During intracellular trafficking N-ethylmaleimide-sensitive-factor attachment receptor (SNARE) proteins catalyze the membrane fusion by assembling into a four-helix complex. In the mouse model, loss of the endosomal SNAREs vti1a and vti1b results in a perinatal lethal phenotype and neuronal defects including decreased neurite outgrowth in cultured primary neurons.</div><div>We used a CRISPR/Cas9 system to generate a <em>Vti1a Vti1b</em> double knockout (DKO) in the neuroblastoma cell line N1E-115. Three different DKO cell lines were obtained and examined at genome and protein level. The double deficiency impaired proper differentiation based on lower levels of synaptic proteins as well as reduced neurite formation and elongation compared to wild type cells in differentiation medium. Neurite elongation can be induced by a variety of extracellular signals via different signaling cascades. Treatment with the Rho kinase inhibitor Y27632, which stimulates enlargeosome exocytosis, or with neurotrophic factors (BDNF, NGF and NT3) resulted in reduced stimulation of all DKO clones and in significantly shorter neurites compared to wild type cells. The loss of vti1a and vti1b disrupted Akt signaling during enlargeosome-mediated and Erk signaling during BDNF-induced neurite outgrowth.</div></div>","PeriodicalId":12010,"journal":{"name":"European journal of cell biology","volume":null,"pages":null},"PeriodicalIF":4.5,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142425283","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The expression level of VEGFR2 regulates mechanotransduction, tumor growth and metastasis of high grade serous ovarian cancer cells VEGFR2 的表达水平调控着高级别浆液性卵巢癌细胞的机械传导、肿瘤生长和转移。
IF 4.5 3区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 DOI: 10.1016/j.ejcb.2024.151459
Recent data shows that alterations in the expression and/or activation of the vascular endothelial growth factor receptor 2 (VEGFR2) in high grade serous ovarian cancer (HGSOC) modulate tumor progression. However, controversial results have been obtained, showing that in some cases VEGFR2 inhibition can promote tumorigenesis and metastasis. Thus, it is urgent to better define the role of the VEGF/VEGFR2 system to understand/predict the effects of its inhibitors administered as anti-angiogenic in HGSOC. Here, we modulated the expression levels of VEGFR2 and analyzed the effects in two cellular models of HGSOC. VEGFR2 silencing (or its pharmacological inhibition) promote the growth and invasive potential of OVCAR3 cells in vitro and in vivo. Consistent with this, the low levels of VEGFR2 in OV7 cells are associated with more pronounced proliferative and motile phenotypes when compared to OVCAR3 cells, and VEGFR2 overexpression in OV7 cells inhibits cell growth. In vitro data confirmed that VEGFR2 silencing in OVCAR3 cells favors the acquisition of an invasive phenotype by loosening cell-ECM contacts, reducing the size and the signaling of focal adhesion contacts (FAs). This is translated into a reduced FAK activity at FAs, ECM-dependent alterations of mechanical forces through FAs and YAP nuclear translocation. Together, the data show that low expression, silencing or inhibition of VEGFR2 in HGSOC cells alter mechanotransduction and lead to the acquisition of a pro-proliferative/invasive phenotype which explains the need for a more cautious use of anti-VEGFR2 drugs in ovarian cancer.
最近的数据显示,高级别浆液性卵巢癌(HGSOC)中血管内皮生长因子受体 2(VEGFR2)的表达和/或激活的改变可调节肿瘤的进展。然而,有争议的结果表明,在某些情况下,抑制血管内皮生长因子受体 2 可促进肿瘤发生和转移。因此,当务之急是更好地界定 VEGF/VEGFR2 系统的作用,以了解/预测其抑制剂作为抗血管生成药物在 HGSOC 中的作用。在这里,我们调节了 VEGFR2 的表达水平,并分析了两种 HGSOC 细胞模型的效果。VEGFR2 沉默(或其药物抑制)促进了 OVCAR3 细胞在体外和体内的生长和侵袭潜力。与此相一致的是,与 OVCAR3 细胞相比,OV7 细胞中低水平的 VEGFR2 与更明显的增殖和运动表型相关,而 OV7 细胞中 VEGFR2 的过表达会抑制细胞生长。体外数据证实,OVCAR3 细胞中的 VEGFR2 沉默通过松动细胞-ECM 接触、减小病灶粘附接触(FA)的大小和信号传导,有利于获得侵袭表型。这将转化为 FAs 上 FAK 活性的降低、通过 FAs 机械力的 ECM 依赖性改变以及 YAP 核转位。这些数据共同表明,HGSOC 细胞中 VEGFR2 的低表达、沉默或抑制改变了机械传导,并导致获得一种促增殖/侵袭性表型,这解释了在卵巢癌中更谨慎使用抗 VEGFR2 药物的必要性。
{"title":"The expression level of VEGFR2 regulates mechanotransduction, tumor growth and metastasis of high grade serous ovarian cancer cells","authors":"","doi":"10.1016/j.ejcb.2024.151459","DOIUrl":"10.1016/j.ejcb.2024.151459","url":null,"abstract":"<div><div>Recent data shows that alterations in the expression and/or activation of the vascular endothelial growth factor receptor 2 (VEGFR2) in high grade serous ovarian cancer (HGSOC) modulate tumor progression. However, controversial results have been obtained, showing that in some cases VEGFR2 inhibition can promote tumorigenesis and metastasis. Thus, it is urgent to better define the role of the VEGF/VEGFR2 system to understand/predict the effects of its inhibitors administered as anti-angiogenic in HGSOC. Here, we modulated the expression levels of VEGFR2 and analyzed the effects in two cellular models of HGSOC. VEGFR2 silencing (or its pharmacological inhibition) promote the growth and invasive potential of OVCAR3 cells <em>in vitro</em> and <em>in vivo</em>. Consistent with this, the low levels of VEGFR2 in OV7 cells are associated with more pronounced proliferative and motile phenotypes when compared to OVCAR3 cells, and VEGFR2 overexpression in OV7 cells inhibits cell growth. <em>In vitro</em> data confirmed that VEGFR2 silencing in OVCAR3 cells favors the acquisition of an invasive phenotype by loosening cell-ECM contacts, reducing the size and the signaling of focal adhesion contacts (FAs). This is translated into a reduced FAK activity at FAs, ECM-dependent alterations of mechanical forces through FAs and YAP nuclear translocation. Together, the data show that low expression, silencing or inhibition of VEGFR2 in HGSOC cells alter mechanotransduction and lead to the acquisition of a pro-proliferative/invasive phenotype which explains the need for a more cautious use of anti-VEGFR2 drugs in ovarian cancer.</div></div>","PeriodicalId":12010,"journal":{"name":"European journal of cell biology","volume":null,"pages":null},"PeriodicalIF":4.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142389093","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cryo-EM structures of cardiac muscle α-actin mutants M305L and A331P give insights into the structural mechanisms of hypertrophic cardiomyopathy 心肌α-肌动蛋白突变体M305L和A331P的低温电子显微镜结构揭示了肥厚型心肌病的结构机制。
IF 4.5 3区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 DOI: 10.1016/j.ejcb.2024.151460
Cardiac muscle α-actin is a key protein of the thin filament in the muscle sarcomere that, together with myosin thick filaments, produce force and contraction important for normal heart function. Missense mutations in cardiac muscle α-actin can cause hypertrophic cardiomyopathy, a complex disorder of the heart characterized by hypercontractility at the molecular scale that leads to diverse clinical phenotypes. While the clinical aspects of hypertrophic cardiomyopathy have been extensively studied, the molecular mechanisms of missense mutations in cardiac muscle α-actin that cause the disease remain largely elusive. Here we used cryo-electron microscopy to reveal the structures of hypertrophic cardiomyopathy-associated actin mutations M305L and A331P in the filamentous state. We show that the mutations have subtle impacts on the overall architecture of the actin filament with mutation-specific changes in the nucleotide binding cleft active site, interprotomer interfaces, and local changes around the mutation site. This suggests that structural changes induced by M305L and A331P have implications for the positioning of the thin filament protein tropomyosin and the interaction with myosin motors. Overall, this study supports a structural model whereby altered interactions between thick and thin filament proteins contribute to disease mechanisms in hypertrophic cardiomyopathy.
心肌α-肌动蛋白是肌肉肌节中细丝的一种关键蛋白质,它与肌球蛋白粗丝一起产生对正常心脏功能很重要的力量和收缩。心肌α-肌动蛋白的错义突变可导致肥厚型心肌病,这是一种复杂的心脏疾病,其特点是分子尺度上的过度收缩,导致不同的临床表型。虽然肥厚型心肌病的临床表现已被广泛研究,但心肌α-肌动蛋白的错义突变导致该病的分子机制在很大程度上仍然难以捉摸。在这里,我们利用低温电子显微镜揭示了肥厚型心肌病相关肌动蛋白突变 M305L 和 A331P 在丝状状态下的结构。我们发现,这两种突变对肌动蛋白丝的整体结构有着微妙的影响,核苷酸结合裂隙活性位点、原体间界面以及突变位点周围的局部都发生了突变特异性变化。这表明,M305L 和 A331P 诱导的结构变化对细丝蛋白肌球蛋白的定位以及与肌球蛋白马达的相互作用有影响。总之,这项研究支持一种结构模型,即粗丝蛋白和细丝蛋白之间相互作用的改变有助于肥厚型心肌病的发病机制。
{"title":"Cryo-EM structures of cardiac muscle α-actin mutants M305L and A331P give insights into the structural mechanisms of hypertrophic cardiomyopathy","authors":"","doi":"10.1016/j.ejcb.2024.151460","DOIUrl":"10.1016/j.ejcb.2024.151460","url":null,"abstract":"<div><div>Cardiac muscle α-actin is a key protein of the thin filament in the muscle sarcomere that, together with myosin thick filaments, produce force and contraction important for normal heart function. Missense mutations in cardiac muscle α-actin can cause hypertrophic cardiomyopathy, a complex disorder of the heart characterized by hypercontractility at the molecular scale that leads to diverse clinical phenotypes. While the clinical aspects of hypertrophic cardiomyopathy have been extensively studied, the molecular mechanisms of missense mutations in cardiac muscle α-actin that cause the disease remain largely elusive. Here we used cryo-electron microscopy to reveal the structures of hypertrophic cardiomyopathy-associated actin mutations M305L and A331P in the filamentous state. We show that the mutations have subtle impacts on the overall architecture of the actin filament with mutation-specific changes in the nucleotide binding cleft active site, interprotomer interfaces, and local changes around the mutation site. This suggests that structural changes induced by M305L and A331P have implications for the positioning of the thin filament protein tropomyosin and the interaction with myosin motors. Overall, this study supports a structural model whereby altered interactions between thick and thin filament proteins contribute to disease mechanisms in hypertrophic cardiomyopathy.</div></div>","PeriodicalId":12010,"journal":{"name":"European journal of cell biology","volume":null,"pages":null},"PeriodicalIF":4.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142406264","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Paving the way to a neural fate – RNA signatures in naive and trans-differentiating mesenchymal stem cells 为神经命运铺平道路--原始间充质干细胞和经分化间充质干细胞的 RNA 特征
IF 4.5 3区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-09-26 DOI: 10.1016/j.ejcb.2024.151458
Mesenchymal Stem Cells (MSCs) derived from the embryonic mesoderm persist as a viable source of multipotent cells in adults and have a crucial role in tissue repair. One of the most promising aspects of MSCs is their ability to trans-differentiate into cell types outside of the mesodermal lineage, such as neurons. This characteristic positions MSCs as potential therapeutic tools for neurological disorders. However, the definition of a clear MSC signature is an ongoing topic of debate. Likewise, there is still a significant knowledge gap about functional alterations of MSCs during their transition to a neural fate. In this study, our focus is on the dynamic expression of RNA in MSCs as they undergo trans-differentiation compared to undifferentiated MSCs. To track and correlate changes in cellular signaling, we conducted high-throughput RNA expression profiling during the early time-course of human MSC neurogenic trans-differentiation. The expression of synapse maturation markers, including NLGN2 and NPTX1, increased during the first 24 h. The expression of neuron differentiation markers, such as GAP43 strongly increased during 48 h of trans-differentiation. Neural stem cell marker NES and neuron differentiation marker, including TUBB3 and ENO1, were highly expressed in mesenchymal stem cells and remained so during trans-differentiation. Pathways analyses revealed early changes in MSCs signaling that can be linked to the acquisition of neuronal features. Furthermore, we identified microRNAs (miRNAs) as potential drivers of the cellular trans-differentiation process. We also determined potential risk factors related to the neural trans-differentiation process. These factors include the persistence of stemness features and the expression of factors involved in neurofunctional abnormalities and tumorigenic processes. In conclusion, our findings contribute valuable insights into the intricate landscape of MSCs during neural trans-differentiation. These insights can pave the way for the development of safer treatments of neurological disorders.
来源于胚胎中胚层的间充质干细胞(MSCs)是成人多能细胞的一个可行来源,在组织修复中起着至关重要的作用。间充质干细胞最有前途的方面之一是它们能够向中胚层以外的细胞类型(如神经元)进行转分化。这一特性使间叶干细胞成为治疗神经系统疾病的潜在工具。然而,如何定义明确的间充质干细胞特征仍是一个争论不休的话题。同样,关于间充质干细胞在向神经命运转变过程中的功能改变,目前仍存在很大的知识空白。在这项研究中,我们的重点是间充质干细胞与未分化间充质干细胞相比,在发生转分化过程中RNA的动态表达。为了跟踪和关联细胞信号的变化,我们在人间叶干细胞神经源性转分化的早期时间过程中进行了高通量 RNA 表达谱分析。在最初的 24 小时内,突触成熟标志物(包括 NLGN2 和 NPTX1)的表达增加。神经元分化标志物(如 GAP43)的表达在转分化的 48 小时内强烈增加。神经干细胞标志物NES和神经元分化标志物(包括TUBB3和ENO1)在间充质干细胞中高度表达,并在转分化过程中保持不变。通路分析揭示了间充质干细胞信号传导的早期变化,这些变化可能与神经元特征的获得有关。此外,我们还发现微RNA(miRNA)是细胞跨分化过程的潜在驱动因素。我们还确定了与神经跨分化过程相关的潜在风险因素。这些因素包括干性特征的持续存在以及涉及神经功能异常和肿瘤发生过程的因子的表达。总之,我们的研究结果为了解神经跨分化过程中间叶干细胞的复杂情况提供了宝贵的见解。这些见解可为开发更安全的神经系统疾病治疗方法铺平道路。
{"title":"Paving the way to a neural fate – RNA signatures in naive and trans-differentiating mesenchymal stem cells","authors":"","doi":"10.1016/j.ejcb.2024.151458","DOIUrl":"10.1016/j.ejcb.2024.151458","url":null,"abstract":"<div><div>Mesenchymal Stem Cells (MSCs) derived from the embryonic mesoderm persist as a viable source of multipotent cells in adults and have a crucial role in tissue repair. One of the most promising aspects of MSCs is their ability to trans-differentiate into cell types outside of the mesodermal lineage, such as neurons. This characteristic positions MSCs as potential therapeutic tools for neurological disorders. However, the definition of a clear MSC signature is an ongoing topic of debate. Likewise, there is still a significant knowledge gap about functional alterations of MSCs during their transition to a neural fate. In this study, our focus is on the dynamic expression of RNA in MSCs as they undergo trans-differentiation compared to undifferentiated MSCs. To track and correlate changes in cellular signaling, we conducted high-throughput RNA expression profiling during the early time-course of human MSC neurogenic trans-differentiation. The expression of synapse maturation markers, including <em>NLGN2</em> and <em>NPTX1</em>, increased during the first 24 h. The expression of neuron differentiation markers, such as <em>GAP43</em> strongly increased during 48 h of trans-differentiation. Neural stem cell marker <em>NES</em> and neuron differentiation marker, including <em>TUBB3</em> and <em>ENO1</em>, were highly expressed in mesenchymal stem cells and remained so during trans-differentiation. Pathways analyses revealed early changes in MSCs signaling that can be linked to the acquisition of neuronal features. Furthermore, we identified microRNAs (miRNAs) as potential drivers of the cellular trans-differentiation process. We also determined potential risk factors related to the neural trans-differentiation process. These factors include the persistence of stemness features and the expression of factors involved in neurofunctional abnormalities and tumorigenic processes. In conclusion, our findings contribute valuable insights into the intricate landscape of MSCs during neural trans-differentiation. These insights can pave the way for the development of safer treatments of neurological disorders.</div></div>","PeriodicalId":12010,"journal":{"name":"European journal of cell biology","volume":null,"pages":null},"PeriodicalIF":4.5,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142327003","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Insulin receptor substrate 1 is a novel member of EGFR signaling in pancreatic cells 胰岛素受体底物 1 是胰腺细胞中表皮生长因子受体信号转导的新成员
IF 4.5 3区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-09-18 DOI: 10.1016/j.ejcb.2024.151457
Pancreatic ductal adenocarcinoma is an extremely incurable cancer type characterized by cells with highly proliferative capacity and resistance against the current therapeutic options. Our study reveals that IRS1 acts as a bridging molecule between EGFR and IGFR/InsR signalization providing a potential mechanism for the interplay between signaling pathways and bypassing EGFR-targeted or IGFR/InsR-targeted therapies. The analysis of IRS1 phosphorylation status in four pancreatic cell lines identified the impact of EGFR signaling on IRS1 activation in comparison with InsR/IGFR signaling. Significantly reduced viability was observed in IRS1-silenced cells even upon EGF stimulation showing the critical role of IRS1 in the EGFR signaling network in both malignant and normal pancreatic cells. This study also demonstrated that EGFR binds directly to IRS1 and at least on two tyrosine sites, Y612 and Y896, IRS1 becomes phosphorylated in response to EGF stimulation. Mechanistically, the EGFR-mediated phosphorylation of IRS1 can further activate the MAPK signaling pathway with the recruitment of GRB2 protein. Collectively, in this study, IRS1 was identified as a crucial regulator in the EGFR signaling suggesting IRS1 as a potential target for more durable responses to targeted PDAC therapy.
胰腺导管腺癌是一种极难治愈的癌症类型,其特点是细胞具有高度增殖能力和对现有治疗方案的抗药性。我们的研究发现,IRS1 是表皮生长因子受体(EGFR)和表皮生长因子受体(IGFR)/表皮生长因子受体(InsR)信号转导之间的桥梁分子,为信号通路之间的相互作用提供了潜在机制,并可绕过 EGFR 靶向疗法或 IGFR/InsR 靶向疗法。通过分析四种胰腺细胞系的IRS1磷酸化状态,确定了与InsR/IGFR信号传导相比,表皮生长因子受体信号传导对IRS1激活的影响。即使在表皮生长因子受刺激的情况下,IRS1沉默细胞的存活率也会显著降低,这表明IRS1在恶性和正常胰腺细胞的表皮生长因子受体信号转导网络中起着关键作用。这项研究还证明,表皮生长因子受体直接与 IRS1 结合,至少在两个酪氨酸位点 Y612 和 Y896 上,IRS1 会在表皮生长因子受体的刺激下发生磷酸化。从机制上讲,表皮生长因子受体介导的 IRS1 磷酸化可通过招募 GRB2 蛋白进一步激活 MAPK 信号通路。总之,本研究发现 IRS1 是表皮生长因子受体信号传导过程中的一个关键调节因子,这表明 IRS1 是 PDAC 靶向治疗产生更持久反应的潜在靶点。
{"title":"Insulin receptor substrate 1 is a novel member of EGFR signaling in pancreatic cells","authors":"","doi":"10.1016/j.ejcb.2024.151457","DOIUrl":"10.1016/j.ejcb.2024.151457","url":null,"abstract":"<div><div>Pancreatic ductal adenocarcinoma is an extremely incurable cancer type characterized by cells with highly proliferative capacity and resistance against the current therapeutic options. Our study reveals that IRS1 acts as a bridging molecule between EGFR and IGFR/InsR signalization providing a potential mechanism for the interplay between signaling pathways and bypassing EGFR-targeted or IGFR/InsR-targeted therapies. The analysis of IRS1 phosphorylation status in four pancreatic cell lines identified the impact of EGFR signaling on IRS1 activation in comparison with InsR/IGFR signaling. Significantly reduced viability was observed in IRS1-silenced cells even upon EGF stimulation showing the critical role of IRS1 in the EGFR signaling network in both malignant and normal pancreatic cells. This study also demonstrated that EGFR binds directly to IRS1 and at least on two tyrosine sites, Y612 and Y896, IRS1 becomes phosphorylated in response to EGF stimulation. Mechanistically, the EGFR-mediated phosphorylation of IRS1 can further activate the MAPK signaling pathway with the recruitment of GRB2 protein. Collectively, in this study, IRS1 was identified as a crucial regulator in the EGFR signaling suggesting IRS1 as a potential target for more durable responses to targeted PDAC therapy.</div></div>","PeriodicalId":12010,"journal":{"name":"European journal of cell biology","volume":null,"pages":null},"PeriodicalIF":4.5,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142319543","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Androgen receptor activation inhibits endothelial cell migration in vitro and angiogenesis in vivo 雄激素受体激活抑制体外内皮细胞迁移和体内血管生成
IF 4.5 3区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-09-11 DOI: 10.1016/j.ejcb.2024.151456

Our previous research revealed that androgen receptor (AR) activation reduces endothelial cell proliferation via non-genomic pathways. We hypothesized that AR activation might also affect endothelial cell migration, a critical step in angiogenesis. Our data demonstrates that treatment of human umbilical vein endothelial cells (HUVECs) with AR agonists, metribolone (R1881) or dihydrotestosterone (DHT), results in a dose-dependent reduction in migration, which can be reversed by AR antagonists or AR knockdown. Mechanistically, R1881 inhibits HUVEC migration by suppressing RhoA activity through the cSrc/FAK/paxillin pathway and promoting RhoA degradation via RhoA-p27 complex formation, ultimately resulting in RhoA ubiquitination. Transfection with constitutively active RhoA-V14 rescues the inhibitory effect of R1881 on HUVEC migration. Furthermore, R1881 elevates intracellular vascular endothelial growth factor (VEGF) and connective tissue growth factor (CTGF) levels but reduces VEGF secretion from HUVECs. This reduction is attributed to the formation of VEGF-CTGF complexes in the cytosol induced by R1881. Transfection with RhoA-V14 reduces CTGF levels and VEGF-CTGF complex formation, leading to enhanced VEGF secretion. Pre-treatment with WP631, a CTGF inhibitor, mitigates the R1881-induced reduction in VEGF secretion and HUVECs migration. In vivo assessments using zebrafish angiogenesis and mouse matrigel plug assays validate the anti-angiogenic effects of R1881. These findings provide insight into the molecular mechanisms through which AR activation modulates endothelial cell migration and angiogenesis.

我们之前的研究发现,雄激素受体(AR)激活会通过非基因组途径减少内皮细胞增殖。我们假设,AR 的激活也可能影响内皮细胞的迁移,而迁移是血管生成的关键步骤。我们的数据表明,用AR激动剂美曲勃龙(R1881)或双氢睾酮(DHT)处理人脐静脉内皮细胞(HUVECs),会导致迁移量呈剂量依赖性减少,而AR拮抗剂或AR敲除可逆转这种减少。从机理上讲,R1881通过cSrc/FAK/paxillin途径抑制RhoA活性,并通过RhoA-p27复合物的形成促进RhoA降解,最终导致RhoA泛素化,从而抑制HUVEC迁移。转染组成型活性 RhoA-V14 可挽救 R1881 对 HUVEC 迁移的抑制作用。此外,R1881 还能提高细胞内血管内皮生长因子(VEGF)和结缔组织生长因子(CTGF)的水平,但却能减少 HUVEC 的 VEGF 分泌。这种减少归因于 R1881 诱导细胞质中 VEGF-CTGF 复合物的形成。转染 RhoA-V14 可降低 CTGF 水平和 VEGF-CTGF 复合物的形成,从而增强 VEGF 的分泌。用 CTGF 抑制剂 WP631 进行预处理可减轻 R1881 诱导的 VEGF 分泌减少和 HUVECs 迁移。使用斑马鱼血管生成和小鼠 matrigel 塞实验进行的体内评估验证了 R1881 的抗血管生成作用。这些发现有助于深入了解 AR 激活调节内皮细胞迁移和血管生成的分子机制。
{"title":"Androgen receptor activation inhibits endothelial cell migration in vitro and angiogenesis in vivo","authors":"","doi":"10.1016/j.ejcb.2024.151456","DOIUrl":"10.1016/j.ejcb.2024.151456","url":null,"abstract":"<div><p>Our previous research revealed that androgen receptor (AR) activation reduces endothelial cell proliferation via non-genomic pathways. We hypothesized that AR activation might also affect endothelial cell migration, a critical step in angiogenesis. Our data demonstrates that treatment of human umbilical vein endothelial cells (HUVECs) with AR agonists, metribolone (R1881) or dihydrotestosterone (DHT), results in a dose-dependent reduction in migration, which can be reversed by AR antagonists or AR knockdown. Mechanistically, R1881 inhibits HUVEC migration by suppressing RhoA activity through the cSrc/FAK/paxillin pathway and promoting RhoA degradation via RhoA-p27 complex formation, ultimately resulting in RhoA ubiquitination. Transfection with constitutively active RhoA-V14 rescues the inhibitory effect of R1881 on HUVEC migration. Furthermore, R1881 elevates intracellular vascular endothelial growth factor (VEGF) and connective tissue growth factor (CTGF) levels but reduces VEGF secretion from HUVECs. This reduction is attributed to the formation of VEGF-CTGF complexes in the cytosol induced by R1881. Transfection with RhoA-V14 reduces CTGF levels and VEGF-CTGF complex formation, leading to enhanced VEGF secretion. Pre-treatment with WP631, a CTGF inhibitor, mitigates the R1881-induced reduction in VEGF secretion and HUVECs migration. In vivo assessments using zebrafish angiogenesis and mouse matrigel plug assays validate the anti-angiogenic effects of R1881. These findings provide insight into the molecular mechanisms through which AR activation modulates endothelial cell migration and angiogenesis.</p></div>","PeriodicalId":12010,"journal":{"name":"European journal of cell biology","volume":null,"pages":null},"PeriodicalIF":4.5,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0171933524000736/pdfft?md5=d810f7b09670b347a00ba56406f43b8d&pid=1-s2.0-S0171933524000736-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142242280","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
European journal of 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