首页 > 最新文献

Cellular signalling最新文献

英文 中文
TRPC6 regulates necroptosis in myocardial ischemia/reperfusion injury via Ca2+/CaMKII signaling pathway TRPC6通过Ca2+/CaMKII信号通路调节心肌缺血再灌注损伤中的坏死。
IF 4.4 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-08-10 DOI: 10.1016/j.cellsig.2024.111344

Myocardial ischemia-reperfusion injury (MIRI) frequently complicates postoperative cardiovascular disease treatment. Necroptosis, a cell death mechanism similar to apoptosis, is regulated by specific signaling pathways and plays an important role in MIRI. Receptor-interacting protein 3 (RIP3), a key protein regulating necroptosis during MIRI, directly phosphorylates calmodulin-dependent protein kinase II (CaMKII). Leading to mitochondrial permeablity transition pore (mPTP) opening and inducing necroptosis. Transient receptor potential canonical channel 6 (TRPC6) regulats Ca2+ entry, is linked to CaMKII as an important upstream effector. However, the connection between TRPC6 and MIRI necroptosis remains unclear. The study aimed to investigate the relationship between TRPC6 and MIRI necroptosis, with a specific focus on elucidating the role of TRPC6 in regulating CaMKII phosphorylation during cardiac necroptosis via Ca2+ modulation.

Methods and results

The experiment used wild-type (WT) and TRPC6 knockout (TRPC6−/−) mice for I/R model construction, and H9c2 myocardial cell line for H/R model. After ischemia-reperfusion (I/R), TRPC6 protein levels in mice significantly increased, exacerbating myocardial injury, infarct size (IS), and cardiac function in WT mice. In contrast, TRPC6 knockout attenuated myocardial injury, IS, and improved cardiac function. The results showed a significant correlation between changes in CaMKII and TRPC6. TRPC6 knockout led to decreased intracellular calcium levels, CaMKII phosphorylation, reactive oxygen species levels, mPTP opening, and improve mitochondrial structure.

Conclusion

I/R upregulates TRPC6, which mediates Ca2+ entry and CaMKII phosphorylation, exacerbates oxidative stress, and induces necroptosis. These findings suggest a potential therapeutic avenue for mitigating MIRI by targeting TRPC6.

心肌缺血再灌注损伤(MIRI)经常是心血管疾病术后治疗的并发症。坏死是一种类似于细胞凋亡的细胞死亡机制,受特定信号通路调控,在心肌缺血再灌注损伤中发挥着重要作用。受体相互作用蛋白 3(RIP3)是 MIRI 期间调控坏死凋亡的关键蛋白,它直接使钙调素依赖性蛋白激酶 II(CaMKII)磷酸化。导致线粒体通透性转换孔(mPTP)开放并诱导坏死。瞬时受体电位典型通道 6(TRPC6)调节 Ca2+ 进入,与 CaMKII 相关联,是重要的上游效应器。然而,TRPC6 与 MIRI 坏死之间的联系仍不清楚。本研究旨在探讨 TRPC6 与 MIRI 坏死之间的关系,重点是阐明 TRPC6 在心脏坏死过程中通过 Ca2+ 调节 CaMKII 磷酸化的作用。方法和结果:实验使用野生型(WT)和 TRPC6 基因敲除(TRPC6-/-)小鼠构建 I/R 模型,使用 H9c2 心肌细胞系构建 H/R 模型。缺血再灌注(I/R)后,小鼠体内的TRPC6蛋白水平显著升高,加剧了WT小鼠的心肌损伤、梗死面积(IS)和心功能。相反,TRPC6 基因敲除可减轻心肌损伤、IS 并改善心功能。结果表明,CaMKII 和 TRPC6 的变化之间存在明显的相关性。TRPC6 基因敲除可降低细胞内钙水平、CaMKII 磷酸化、活性氧水平、mPTP 开放,并改善线粒体结构。结论:I/R 会上调 TRPC6,而 TRPC6 会介导 Ca2+ 进入和 CaMKII 磷酸化,加剧氧化应激并诱导坏死。这些发现为通过靶向 TRPC6 缓解 MIRI 提供了潜在的治疗途径。
{"title":"TRPC6 regulates necroptosis in myocardial ischemia/reperfusion injury via Ca2+/CaMKII signaling pathway","authors":"","doi":"10.1016/j.cellsig.2024.111344","DOIUrl":"10.1016/j.cellsig.2024.111344","url":null,"abstract":"<div><p>Myocardial ischemia-reperfusion injury (MIRI) frequently complicates postoperative cardiovascular disease treatment. Necroptosis, a cell death mechanism similar to apoptosis, is regulated by specific signaling pathways and plays an important role in MIRI. Receptor-interacting protein 3 (RIP3), a key protein regulating necroptosis during MIRI, directly phosphorylates calmodulin-dependent protein kinase II (CaMKII). Leading to mitochondrial permeablity transition pore (mPTP) opening and inducing necroptosis. Transient receptor potential canonical channel 6 (TRPC6) regulats Ca<sup>2+</sup> entry, is linked to CaMKII as an important upstream effector. However, the connection between TRPC6 and MIRI necroptosis remains unclear. The study aimed to investigate the relationship between TRPC6 and MIRI necroptosis, with a specific focus on elucidating the role of TRPC6 in regulating CaMKII phosphorylation during cardiac necroptosis via Ca<sup>2+</sup> modulation.</p></div><div><h3>Methods and results</h3><p>The experiment used wild-type (WT) and TRPC6 knockout (<em>TRPC6</em><sup><em>−/−</em></sup>) mice for I/R model construction, and H9c2 myocardial cell line for H/R model. After ischemia-reperfusion (I/R), TRPC6 protein levels in mice significantly increased, exacerbating myocardial injury, infarct size (IS), and cardiac function in WT mice. In contrast, TRPC6 knockout attenuated myocardial injury, IS, and improved cardiac function. The results showed a significant correlation between changes in CaMKII and TRPC6. TRPC6 knockout led to decreased intracellular calcium levels, CaMKII phosphorylation, reactive oxygen species levels, mPTP opening, and improve mitochondrial structure.</p></div><div><h3>Conclusion</h3><p>I/R upregulates TRPC6, which mediates Ca<sup>2+</sup> entry and CaMKII phosphorylation, exacerbates oxidative stress, and induces necroptosis. These findings suggest a potential therapeutic avenue for mitigating MIRI by targeting TRPC6.</p></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-08-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141970732","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
The potential roles of HIF-1α in epithelial-mesenchymal transition and ferroptosis in tumor cells HIF-1α在肿瘤细胞上皮-间质转化和铁变态过程中的潜在作用。
IF 4.4 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-08-10 DOI: 10.1016/j.cellsig.2024.111345

In tumors, the rapid proliferation of cells and the imperfect blood supply system lead to hypoxia, which can regulate the adaptation of tumor cells to the hypoxic environment through hypoxia-inducible factor-1α (HIF-1α) and promote tumor development in multiple ways. Recent studies have found that epithelial-mesenchymal transition (EMT) and ferroptosis play important roles in the progression of tumor cells. The activation of HIF-1α is considered a key factor in inducing EMT in tumor cells. When HIF-1α is activated, it can regulate EMT-related genes, causing tumor cells to gradually lose their epithelial characteristics and acquire more invasive mesenchymal traits. The occurrence of EMT allows tumor cells to better adapt to changes in the surrounding tissue, enhancing their migratory and invasive capabilities, thus promoting tumor progression. At the same time, HIF-1α also plays a crucial regulatory role in ferroptosis in tumor cells. In a hypoxic environment, HIF-1α may affect processes such as iron metabolism and oxidative stress responses, inducing ferroptosis in tumor cells. This article briefly reviews the dual role of HIF-1α in EMT and ferroptosis in tumor cells, helping to gain a deeper understanding of the regulatory pathways of HIF-1α in the development of tumor cells, providing a new perspective for understanding the pathogenesis of tumors. The regulation of HIF-1α may become an important strategy for future tumor therapy.

在肿瘤中,细胞的快速增殖和供血系统的不完善导致缺氧,缺氧可通过缺氧诱导因子-1α(HIF-1α)调控肿瘤细胞对缺氧环境的适应,并通过多种途径促进肿瘤的发展。最近的研究发现,上皮-间质转化(EMT)和铁凋亡在肿瘤细胞的发展过程中起着重要作用。HIF-1α 的激活被认为是诱导肿瘤细胞 EMT 的关键因素。当 HIF-1α 被激活时,它可以调控 EMT 相关基因,使肿瘤细胞逐渐失去上皮特征,获得更具侵袭性的间质特征。EMT 的发生可使肿瘤细胞更好地适应周围组织的变化,增强其迁移和侵袭能力,从而促进肿瘤的进展。与此同时,HIF-1α 在肿瘤细胞的铁突变中也发挥着重要的调节作用。在缺氧环境中,HIF-1α可能会影响铁代谢和氧化应激反应等过程,诱导肿瘤细胞的铁突变。本文简要综述了HIF-1α在肿瘤细胞EMT和铁凋亡中的双重作用,有助于深入了解HIF-1α在肿瘤细胞发育过程中的调控途径,为理解肿瘤的发病机制提供一个新的视角。对HIF-1α的调控可能成为未来肿瘤治疗的重要策略。
{"title":"The potential roles of HIF-1α in epithelial-mesenchymal transition and ferroptosis in tumor cells","authors":"","doi":"10.1016/j.cellsig.2024.111345","DOIUrl":"10.1016/j.cellsig.2024.111345","url":null,"abstract":"<div><p>In tumors, the rapid proliferation of cells and the imperfect blood supply system lead to hypoxia, which can regulate the adaptation of tumor cells to the hypoxic environment through hypoxia-inducible factor-1α (HIF-1α) and promote tumor development in multiple ways. Recent studies have found that epithelial-mesenchymal transition (EMT) and ferroptosis play important roles in the progression of tumor cells. The activation of HIF-1α is considered a key factor in inducing EMT in tumor cells. When HIF-1α is activated, it can regulate EMT-related genes, causing tumor cells to gradually lose their epithelial characteristics and acquire more invasive mesenchymal traits. The occurrence of EMT allows tumor cells to better adapt to changes in the surrounding tissue, enhancing their migratory and invasive capabilities, thus promoting tumor progression. At the same time, HIF-1α also plays a crucial regulatory role in ferroptosis in tumor cells. In a hypoxic environment, HIF-1α may affect processes such as iron metabolism and oxidative stress responses, inducing ferroptosis in tumor cells. This article briefly reviews the dual role of HIF-1α in EMT and ferroptosis in tumor cells, helping to gain a deeper understanding of the regulatory pathways of HIF-1α in the development of tumor cells, providing a new perspective for understanding the pathogenesis of tumors. The regulation of HIF-1α may become an important strategy for future tumor therapy.</p></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-08-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0898656824003139/pdfft?md5=c034434acae9c24757b4486abd3a5757&pid=1-s2.0-S0898656824003139-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141970731","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Glaucoma and the ocular renin-angiotensin-aldosterone system: Update on molecular signalling and treatment perspectives 青光眼与眼部肾素-血管紧张素-醛固酮系统:分子信号和治疗前景的最新进展。
IF 4.4 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1016/j.cellsig.2024.111343

Glaucoma, a leading cause of blindness worldwide, encompasses a group of pathological conditions affecting the optic nerve and is characterized by progressive retinal ganglion cell loss, cupping of the optic nerve head, and distinct visual field defects. While elevated intraocular pressure (IOP) is the main risk factor for glaucoma, many patients do not have elevated IOP. Consequently, other risk factors, such as ocular blood flow abnormalities and immunological factors, have been implicated in its pathophysiology. Traditional therapeutic strategies primarily aim to reduce IOP, but there is growing interest in developing novel treatment approaches to improve disease management and reduce the high rates of severe visual impairment. In this context, targeting the ocular renin-angiotensin-aldosterone system (RAAS) has been found as a potential curative strategy. The RAAS contributes to glaucoma development through key effectors such as prorenin, angiotensin II, and aldosterone. Recent evidence has highlighted the potential of using RAAS modulators to combat glaucoma, yielding encouraging results. Our study aims to explore the molecular pathways linking the ocular RAAS and glaucoma, summarizing recent advances that elucidate the role of the RAAS in triggering oxidative stress, inflammation, and remodelling in the pathogenesis of glaucoma. Additionally, we will present emerging therapeutic approaches that utilize RAAS modulators and antioxidants to slow the progression of glaucoma.

青光眼是全球致盲的主要原因之一,它是一组影响视神经的病理状态,其特征是视网膜神经节细胞进行性丧失、视神经头凹陷和明显的视野缺损。虽然眼压升高是青光眼的主要风险因素,但许多患者并没有眼压升高。因此,其他风险因素,如眼部血流异常和免疫因素,也与青光眼的病理生理学有关。传统治疗策略的主要目的是降低眼压,但人们对开发新型治疗方法以改善疾病管理并降低严重视力损伤的高发率越来越感兴趣。在这种情况下,针对眼部肾素-血管紧张素-醛固酮系统(RAAS)的治疗被认为是一种潜在的治疗策略。RAAS 通过肾素、血管紧张素 II 和醛固酮等关键效应因子促进青光眼的发展。最近有证据表明,使用 RAAS 调节剂治疗青光眼具有潜力,并取得了令人鼓舞的成果。我们的研究旨在探索连接眼部 RAAS 和青光眼的分子途径,总结最新进展,阐明 RAAS 在青光眼发病机制中引发氧化应激、炎症和重塑的作用。此外,我们还将介绍利用 RAAS 调节剂和抗氧化剂减缓青光眼进展的新兴治疗方法。
{"title":"Glaucoma and the ocular renin-angiotensin-aldosterone system: Update on molecular signalling and treatment perspectives","authors":"","doi":"10.1016/j.cellsig.2024.111343","DOIUrl":"10.1016/j.cellsig.2024.111343","url":null,"abstract":"<div><p>Glaucoma, a leading cause of blindness worldwide, encompasses a group of pathological conditions affecting the optic nerve and is characterized by progressive retinal ganglion cell loss, cupping of the optic nerve head, and distinct visual field defects. While elevated intraocular pressure (IOP) is the main risk factor for glaucoma, many patients do not have elevated IOP. Consequently, other risk factors, such as ocular blood flow abnormalities and immunological factors, have been implicated in its pathophysiology. Traditional therapeutic strategies primarily aim to reduce IOP, but there is growing interest in developing novel treatment approaches to improve disease management and reduce the high rates of severe visual impairment. In this context, targeting the ocular renin-angiotensin-aldosterone system (RAAS) has been found as a potential curative strategy. The RAAS contributes to glaucoma development through key effectors such as prorenin, angiotensin II, and aldosterone. Recent evidence has highlighted the potential of using RAAS modulators to combat glaucoma, yielding encouraging results. Our study aims to explore the molecular pathways linking the ocular RAAS and glaucoma, summarizing recent advances that elucidate the role of the RAAS in triggering oxidative stress, inflammation, and remodelling in the pathogenesis of glaucoma. Additionally, we will present emerging therapeutic approaches that utilize RAAS modulators and antioxidants to slow the progression of glaucoma.</p></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0898656824003115/pdfft?md5=d57d7006e0a72d33f99cba44176bd233&pid=1-s2.0-S0898656824003115-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141912022","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
LOC730101 transmitted by exosomes facilitates laryngeal squamous cell carcinoma tumorigenesis via regulation of p38 MAPK gamma 由外泌体传播的 LOC730101 通过调节 p38 MAPK gamma 促进喉鳞状细胞癌的发生。
IF 4.4 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1016/j.cellsig.2024.111336

Laryngeal squamous cell carcinoma (LSCC) is a prevalent human cancer with a complex pathogenesis that remains incompletely understood. Here, we unveil a long non-coding RNA (lncRNA) associated with LSCC tumorigenesis and progression. LOC730101 exhibits significant overexpression in human LSCC tissues, and elevated LOC730101 levels correlate with malignant clinicopathological characteristics. Moreover, we demonstrate that LOC730101 is encapsulated into exosomes in an hnRNPA2B1-dependent manner, serving as a promising plasma biomarker for discriminating LSCC patients from healthy individuals (AUC = 0.92 with 89.36% sensitivity and 86.36% specificity). Exosomes derived from LSCC cells enhance the viability, DNA synthesis rate, and invasiveness of normal nasopharynx epithelial cells, with pronounced effects observed upon LOC730101 overexpression. Additionally, exosomal LOC730101 promotes tumor growth in vivo. Mechanistically, exosomal LOC730101 internalization by normal nasopharynx epithelial cells leads to increased H3K4me3 levels on the p38 MAPK gamma (p38γ) promoter via direct interaction with hnRNPA2B1. This interaction activates p38γ transcription, ultimately driving LSCC tumorigenesis. Collectively, our findings uncover a novel exosomal lncRNA that mediates communication between normal and LSCC cells during LSCC carcinogenesis, suggesting that targeting LOC730101 may represent a promising therapeutic strategy for LSCC treatment.

喉鳞状细胞癌(LSCC)是一种发病率很高的人类癌症,其发病机制复杂,至今仍未完全明了。在这里,我们揭示了一种与 LSCC 肿瘤发生和发展相关的长非编码 RNA(lncRNA)。LOC730101 在人类 LSCC 组织中表现出显著的过表达,LOC730101 水平的升高与恶性临床病理特征相关。此外,我们还证明了 LOC730101 会以 hnRNPA2B1 依赖性的方式被包裹到外泌体中,从而成为区分 LSCC 患者和健康人的一种很有前景的血浆生物标记物(AUC = 0.92,灵敏度为 89.36%,特异度为 86.36%)。从LSCC细胞中提取的外泌体可增强正常鼻咽上皮细胞的活力、DNA合成率和侵袭性,在LOC730101过表达时可观察到明显的效果。此外,外泌体 LOC730101 还能促进体内肿瘤的生长。从机理上讲,外泌体 LOC730101 被正常鼻咽上皮细胞内化后,通过与 hnRNPA2B1 直接相互作用,导致 p38 MAPK gamma(p38γ)启动子上的 H3K4me3 水平升高。这种相互作用激活了 p38γ 的转录,最终推动了 LSCC 的肿瘤发生。总之,我们的研究结果发现了一种新型外泌体lncRNA,它在LSCC癌变过程中介导正常细胞和LSCC细胞之间的交流,这表明靶向LOC730101可能是治疗LSCC的一种有前景的治疗策略。
{"title":"LOC730101 transmitted by exosomes facilitates laryngeal squamous cell carcinoma tumorigenesis via regulation of p38 MAPK gamma","authors":"","doi":"10.1016/j.cellsig.2024.111336","DOIUrl":"10.1016/j.cellsig.2024.111336","url":null,"abstract":"<div><p>Laryngeal squamous cell carcinoma (LSCC) is a prevalent human cancer with a complex pathogenesis that remains incompletely understood. Here, we unveil a long non-coding RNA (lncRNA) associated with LSCC tumorigenesis and progression. LOC730101 exhibits significant overexpression in human LSCC tissues, and elevated LOC730101 levels correlate with malignant clinicopathological characteristics. Moreover, we demonstrate that LOC730101 is encapsulated into exosomes in an hnRNPA2B1-dependent manner, serving as a promising plasma biomarker for discriminating LSCC patients from healthy individuals (AUC = 0.92 with 89.36% sensitivity and 86.36% specificity). Exosomes derived from LSCC cells enhance the viability, DNA synthesis rate, and invasiveness of normal nasopharynx epithelial cells, with pronounced effects observed upon LOC730101 overexpression. Additionally, exosomal LOC730101 promotes tumor growth in vivo. Mechanistically, exosomal LOC730101 internalization by normal nasopharynx epithelial cells leads to increased H3K4me3 levels on the p38 MAPK gamma (p38γ) promoter via direct interaction with hnRNPA2B1. This interaction activates p38γ transcription, ultimately driving LSCC tumorigenesis. Collectively, our findings uncover a novel exosomal lncRNA that mediates communication between normal and LSCC cells during LSCC carcinogenesis, suggesting that targeting LOC730101 may represent a promising therapeutic strategy for LSCC treatment.</p></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141912023","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Ndufa8 promotes white fat Browning by improving mitochondrial respiratory chain complex I function to ameliorate obesity by in vitro and in vivo Ndufa8 通过改善线粒体呼吸链复合物 I 的功能促进白色脂肪棕色化,从而在体外和体内改善肥胖。
IF 4.4 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1016/j.cellsig.2024.111340

Obesity and its complications have become a global health problem that needs to be addressed urgently. White adipose tissue (WAT) browning contributes to consuming excess energy in WAT, which is important for improving obesity and maintaining a healthy energy homeostasis. Mitochondria, as the energy metabolism center of cells, are extensively involved in many metabolic processes, including the browning of WAT. NADH: Ubiquinone oxidoreductase subunit A8 (NDUFA8) is a constituent subunit of respiratory chain complex I (CI), which has been found to participate in a wide range of physiological processes by affecting the activity of respiratory CI. However, the regulatory effect of Ndufa8 on the browning of WAT has not been reported. Here, we used β3-adrenergic agonis CL316, 243 to construct WAT browning models in vivo and in vitro to investigate the role and mechanism of Ndufa8 in the regulation of WAT browning. Briefly, Ndufa8 significantly increased CI activity and suppressed mitochondrial ROS levels in vitro, thereby improving mitochondrial function. Ndufa8 also increased the transcriptional levels and protein levels of UCP1 in vitro and in vivo, which promoted WAT browning. Our findings provide a new molecular approach for the research of browning of WAT in animals, as well as a new target for animal metabolism improvement and obesity treatments.

肥胖症及其并发症已成为亟待解决的全球性健康问题。白脂肪组织(WAT)褐变有助于消耗WAT中过剩的能量,这对改善肥胖和维持健康的能量平衡非常重要。线粒体作为细胞的能量代谢中心,广泛参与了许多代谢过程,包括白脂肪组织的褐变。NADH:泛醌氧化还原酶亚基 A8(NDUFA8)是呼吸链复合体 I(CI)的组成亚基,研究发现它通过影响呼吸链复合体 I 的活性参与多种生理过程。然而,Ndufa8 对 WAT 褐变的调控作用尚未见报道。在此,我们利用β3-肾上腺素能激动剂CL316、243构建体内和体外WAT褐变模型,研究Ndufa8在调控WAT褐变中的作用和机制。简而言之,Ndufa8能显著提高体外CI活性并抑制线粒体ROS水平,从而改善线粒体功能。Ndufa8 还能提高体外和体内 UCP1 的转录水平和蛋白水平,从而促进脂肪褐变。我们的发现为研究动物脂肪褐变提供了一种新的分子方法,也为改善动物代谢和治疗肥胖症提供了一个新的靶点。
{"title":"Ndufa8 promotes white fat Browning by improving mitochondrial respiratory chain complex I function to ameliorate obesity by in vitro and in vivo","authors":"","doi":"10.1016/j.cellsig.2024.111340","DOIUrl":"10.1016/j.cellsig.2024.111340","url":null,"abstract":"<div><p>Obesity and its complications have become a global health problem that needs to be addressed urgently. White adipose tissue (WAT) browning contributes to consuming excess energy in WAT, which is important for improving obesity and maintaining a healthy energy homeostasis. Mitochondria, as the energy metabolism center of cells, are extensively involved in many metabolic processes, including the browning of WAT. NADH: Ubiquinone oxidoreductase subunit A8 (NDUFA8) is a constituent subunit of respiratory chain complex I (CI), which has been found to participate in a wide range of physiological processes by affecting the activity of respiratory CI. However, the regulatory effect of <em>Ndufa8</em> on the browning of WAT has not been reported. Here, we used β3-adrenergic agonis CL316, 243 to construct WAT browning models in vivo and in vitro to investigate the role and mechanism of <em>Ndufa8</em> in the regulation of WAT browning. Briefly, <em>Ndufa8</em> significantly increased CI activity and suppressed mitochondrial ROS levels in vitro, thereby improving mitochondrial function. <em>Ndufa8</em> also increased the transcriptional levels and protein levels of UCP1 in vitro and in vivo, which promoted WAT browning. Our findings provide a new molecular approach for the research of browning of WAT in animals, as well as a new target for animal metabolism improvement and obesity treatments.</p></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141912025","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
PPM1G promotes autophagy and progression of pancreatic cancer via upregulating HMGB1 PPM1G 通过上调 HMGB1 促进自噬和胰腺癌的进展。
IF 4.4 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1016/j.cellsig.2024.111342

Pancreatic cancer remains one of the most aggressive and lethal malignancies worldwide, with a dismal 5-year relative survival rates of only 12%. Therefore, it is urgent to discover the key molecular markers to improve the therapeutic outcomes in pancreatic cancer. Herein, we first demonstrated that PPM1G is upregulated in pancreatic cancer and that PPM1G depletion decreases pancreatic cancer cell growth in vitro and in vivo. High PPM1G expression was linked to short overall survival of pancreatic cancer patients, which was further validated in the TCGA database. Moreover, by detecting Beclin 1, LC3-II, and SQSTM1/p62 expressions and observing autolysosome under transmission electron microscope, we discovered that PPM1G is a novel positive regulator of macroautophagy/autophagy. Furthermore, by using immunoprecipitation-mass spectrometry (IP-MS) analysis and following systemic molecular biology experiment, we demonstrated PPM1G promotes the autophagy and proliferation of pancreatic cancer by directly upregulating HMGB1. Additionally, patients with both high PPM1G and high HMGB1 exhibited poorer prognosis in our cohort. This study preliminarily investigated the possibility of PPM1G as a potential therapeutic target and prognostic biomarker in pancreatic cancer patients.

胰腺癌仍然是全球最具侵袭性和致命性的恶性肿瘤之一,5 年相对生存率仅为 12%,令人沮丧。因此,发现关键分子标记物以改善胰腺癌的治疗效果迫在眉睫。在本文中,我们首次证明了 PPM1G 在胰腺癌中的上调,并且 PPM1G 的缺失会降低胰腺癌细胞在体外和体内的生长。PPM1G的高表达与胰腺癌患者总生存期短有关,这一点在TCGA数据库中得到了进一步验证。此外,通过检测 Beclin 1、LC3-II 和 SQSTM1/p62 的表达以及在透射电子显微镜下观察自溶体,我们发现 PPM1G 是一种新型的大自噬/自噬正调控因子。此外,通过免疫沉淀-质谱(IP-MS)分析和系统分子生物学实验,我们证明了PPM1G通过直接上调HMGB1促进胰腺癌的自噬和增殖。此外,在我们的队列中,高 PPM1G 和高 HMGB1 患者的预后较差。本研究初步探讨了 PPM1G 作为胰腺癌患者潜在治疗靶点和预后生物标志物的可能性。
{"title":"PPM1G promotes autophagy and progression of pancreatic cancer via upregulating HMGB1","authors":"","doi":"10.1016/j.cellsig.2024.111342","DOIUrl":"10.1016/j.cellsig.2024.111342","url":null,"abstract":"<div><p>Pancreatic cancer remains one of the most aggressive and lethal malignancies worldwide, with a dismal 5-year relative survival rates of only 12%. Therefore, it is urgent to discover the key molecular markers to improve the therapeutic outcomes in pancreatic cancer. Herein, we first demonstrated that PPM1G is upregulated in pancreatic cancer and that PPM1G depletion decreases pancreatic cancer cell growth in vitro and in vivo. High PPM1G expression was linked to short overall survival of pancreatic cancer patients, which was further validated in the TCGA database. Moreover, by detecting Beclin 1, LC3-II, and SQSTM1/p62 expressions and observing autolysosome under transmission electron microscope, we discovered that PPM1G is a novel positive regulator of macroautophagy/autophagy. Furthermore, by using immunoprecipitation-mass spectrometry (IP-MS) analysis and following systemic molecular biology experiment, we demonstrated PPM1G promotes the autophagy and proliferation of pancreatic cancer by directly upregulating HMGB1. Additionally, patients with both high PPM1G and high HMGB1 exhibited poorer prognosis in our cohort. This study preliminarily investigated the possibility of PPM1G as a potential therapeutic target and prognostic biomarker in pancreatic cancer patients.</p></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141912026","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
CircATXN7 regulates the proliferation and invasion of esophageal cancer cells through miR-4319/NLRC5 CircATXN7通过miR-4319/NLRC5调节食管癌细胞的增殖和侵袭。
IF 4.4 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1016/j.cellsig.2024.111341

Background

This study aimed to explore the molecular mechanism through which circular RNA of ataxin 7 (circATXN7) regulates the proliferation and invasion of esophageal cancer (EC) cells via microRNA (miR)-4319/NLR family CARD domain containing 5 (NLRC5).

Methods

The localization of circATXN7 in EC cells was determined by RNA fluorescent in situ hybridization (RNA-FISH). The mRNA levels of circATXN7, miR-4319, and NLRC5 were quantified by reverse transcription-polymerase chain reactions. The binding activity of circATXN7 to miR-4319 was assessed using RNA-binding protein immunoprecipitation. Whether circATXN7 regulates the proliferation of EC cells via miR-4319 was explored using dual-luciferase reporter gene colony formation assays. Protein levels were quantified by western blot. The effect of NLRC5 on the proliferation and invasion of EC cells was examined using colony formation and Transwell assays. A subcutaneous transplanted tumor nude mouse model was established to observe the effect of circATXN7 on the proliferation of EC cells in vivo.

Results

circATXN7 localized mainly to the cytoplasm. Overexpression or inhibition of miR-4319 significantly regulated the proliferation of EC cells, while circATXN7 competitively inhibited miR-4319 expression. Overexpression of miR-4319 significantly inhibited NLRC5 expression, indicating NLRC5 is a downstream regulatory target of miR-4319. circATXN7 influenced NLRC5 expression via miR-4319. In vivo tumor formation experiments in nude mice revealed that knocking down circATXN7 regulated NLRC5 expression via miR-4319 and significantly inhibited the proliferation of EC cells.

Conclusions

In vitro cell and in vivo animal experiments showed that circATXN7 regulates the proliferation, invasion, and migration of EC cells through the miR-4319/NLRC5 signaling pathway.

研究背景本研究旨在探讨阿塔克星7的环状RNA(circATXN7)通过microRNA(miR)-4319/NLR家族CARD域包含5(NLRC5)调控食管癌(EC)细胞增殖和侵袭的分子机制:方法:通过RNA荧光原位杂交(RNA-FISH)测定circATXN7在食管癌细胞中的定位。反转录聚合酶链反应定量检测了 circATXN7、miR-4319 和 NLRC5 的 mRNA 水平。利用 RNA 结合蛋白免疫沉淀法评估了 circATXN7 与 miR-4319 的结合活性。使用双荧光素酶报告基因集落形成试验探讨了 circATXN7 是否通过 miR-4319 调节 EC 细胞的增殖。蛋白水平通过蛋白印迹进行量化。使用集落形成和 Transwell 试验检测了 NLRC5 对 EC 细胞增殖和侵袭的影响。结果:circATXN7主要定位于细胞质。过表达或抑制miR-4319可显著调节EC细胞的增殖,而circATXN7可竞争性抑制miR-4319的表达。过表达 miR-4319 能明显抑制 NLRC5 的表达,表明 NLRC5 是 miR-4319 的下游调控靶标。裸鼠体内肿瘤形成实验表明,敲除circATXN7可通过miR-4319调控NLRC5的表达,并显著抑制EC细胞的增殖:体外细胞和体内动物实验表明,circATXN7通过miR-4319/NLRC5信号通路调控EC细胞的增殖、侵袭和迁移。
{"title":"CircATXN7 regulates the proliferation and invasion of esophageal cancer cells through miR-4319/NLRC5","authors":"","doi":"10.1016/j.cellsig.2024.111341","DOIUrl":"10.1016/j.cellsig.2024.111341","url":null,"abstract":"<div><h3>Background</h3><p>This study aimed to explore the molecular mechanism through which circular RNA of ataxin 7 (circATXN7) regulates the proliferation and invasion of esophageal cancer (EC) cells via microRNA (miR)-4319/NLR family CARD domain containing 5 (NLRC5).</p></div><div><h3>Methods</h3><p>The localization of circATXN7 in EC cells was determined by RNA fluorescent in situ hybridization (RNA-FISH). The mRNA levels of circATXN7, miR-4319, and NLRC5 were quantified by reverse transcription-polymerase chain reactions. The binding activity of circATXN7 to miR-4319 was assessed using RNA-binding protein immunoprecipitation. Whether circATXN7 regulates the proliferation of EC cells via miR-4319 was explored using dual-luciferase reporter gene colony formation assays. Protein levels were quantified by western blot. The effect of NLRC5 on the proliferation and invasion of EC cells was examined using colony formation and Transwell assays. A subcutaneous transplanted tumor nude mouse model was established to observe the effect of circATXN7 on the proliferation of EC cells in vivo.</p></div><div><h3>Results</h3><p>circATXN7 localized mainly to the cytoplasm. Overexpression or inhibition of miR-4319 significantly regulated the proliferation of EC cells, while circATXN7 competitively inhibited miR-4319 expression. Overexpression of miR-4319 significantly inhibited NLRC5 expression, indicating NLRC5 is a downstream regulatory target of miR-4319. circATXN7 influenced NLRC5 expression via miR-4319. In vivo tumor formation experiments in nude mice revealed that knocking down circATXN7 regulated NLRC5 expression via miR-4319 and significantly inhibited the proliferation of EC cells.</p></div><div><h3>Conclusions</h3><p>In vitro cell and in vivo animal experiments showed that circATXN7 regulates the proliferation, invasion, and migration of EC cells through the miR-4319/NLRC5 signaling pathway.</p></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141912021","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
The MEF2A/SNHG16/miR-425-5p/NOTCH2 axis induces gemcitabine resistance by inhibiting ferroptosis in the starving bladder tumor microenvironment MEF2A/SNHG16/miR-425-5p/NOTCH2轴通过抑制饥饿膀胱肿瘤微环境中的铁突变,诱导吉西他滨耐药。
IF 4.4 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1016/j.cellsig.2024.111337

Gemcitabine resistance is one of the leading causes of bladder cancer (BCa) recurrence and progression. The dysregulation of ferroptosis is involved in this process; however, the underlying mechanisms remain unclear. In the current study, we found a prominent increase in long non-coding RNA (lncRNA) small nucleolar RNA host gene 16 (SNHG16) in tumor samples, which was related to advanced tumor grade and poor prognosis. SNHG16 is overexpressed in the starving tumor microenvironment (STME) and induces gemcitabine resistance by inhibiting ferroptosis in BCa. SNHG16 knockdown promotes ferroptosis and increases chemosensitivity to gemcitabine. Mechanistically, the transcription factor MEF2A was markedly upregulated in the STME, facilitating SNHG16 expression. SNHG16 acts as a competing endogenous RNA that sponges miR-425-5p and promotes NOTCH2 expression. SNHG16/miR-425-5p/NOTCH2 is demonstrated, for the first time, to suppress ferroptosis by inducing SLC7A11 and GPX4 expression in vitro and in vivo. Upregulation of miR-425-5p reverses NOTCH2-mediated inhibition of ferroptosis, thereby mitigating gemcitabine resistance. In conclusion, these findings reveal that the STME-activated MEF2A/SNHG16/miR-425-5p/NOTCH2 axis induces gemcitabine resistance by inhibiting ferroptosis and implicate SNHG16 as a potential therapeutic target for chemoresistance.

吉西他滨耐药是膀胱癌(BCa)复发和进展的主要原因之一。这一过程涉及铁蛋白沉积失调,但其潜在机制仍不清楚。在目前的研究中,我们发现肿瘤样本中长非编码 RNA(lncRNA)小核仁 RNA 宿主基因 16(SNHG16)显著增加,这与肿瘤晚期分级和预后不良有关。SNHG16在饥饿肿瘤微环境(STME)中过表达,并通过抑制BCa中的铁变态反应诱导吉西他滨耐药。敲除 SNHG16 可促进铁突变,增加对吉西他滨的化疗敏感性。从机制上讲,转录因子MEF2A在STME中明显上调,促进了SNHG16的表达。SNHG16是一种竞争性内源性RNA,它能疏导miR-425-5p并促进NOTCH2的表达。SNHG16/miR-425-5p/NOTCH2 通过诱导体外和体内 SLC7A11 和 GPX4 的表达,首次被证实能抑制铁变态反应。上调 miR-425-5p 可逆转 NOTCH2 介导的铁凋亡抑制,从而减轻吉西他滨耐药性。总之,这些发现揭示了 STME 激活的 MEF2A/SNHG16/miR-425-5p/NOTCH2 轴通过抑制铁突变诱导吉西他滨耐药,并将 SNHG16 作为化疗耐药的潜在治疗靶点。
{"title":"The MEF2A/SNHG16/miR-425-5p/NOTCH2 axis induces gemcitabine resistance by inhibiting ferroptosis in the starving bladder tumor microenvironment","authors":"","doi":"10.1016/j.cellsig.2024.111337","DOIUrl":"10.1016/j.cellsig.2024.111337","url":null,"abstract":"<div><p>Gemcitabine resistance is one of the leading causes of bladder cancer (BCa) recurrence and progression. The dysregulation of ferroptosis is involved in this process; however, the underlying mechanisms remain unclear. In the current study, we found a prominent increase in long non-coding RNA (lncRNA) small nucleolar RNA host gene 16 (SNHG16) in tumor samples, which was related to advanced tumor grade and poor prognosis. SNHG16 is overexpressed in the starving tumor microenvironment (STME) and induces gemcitabine resistance by inhibiting ferroptosis in BCa. SNHG16 knockdown promotes ferroptosis and increases chemosensitivity to gemcitabine. Mechanistically, the transcription factor MEF2A was markedly upregulated in the STME, facilitating SNHG16 expression. SNHG16 acts as a competing endogenous RNA that sponges miR-425-5p and promotes NOTCH2 expression. SNHG16/miR-425-5p/NOTCH2 is demonstrated, for the first time, to suppress ferroptosis by inducing SLC7A11 and GPX4 expression in vitro and in vivo. Upregulation of miR-425-5p reverses NOTCH2-mediated inhibition of ferroptosis, thereby mitigating gemcitabine resistance. In conclusion, these findings reveal that the STME-activated MEF2A/SNHG16/miR-425-5p/NOTCH2 axis induces gemcitabine resistance by inhibiting ferroptosis and implicate SNHG16 as a potential therapeutic target for chemoresistance.</p></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141912027","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
miR-135b-5p promotes gastric carcinogenesis by targeting CLIP4-mediated JAK2/STAT3 signal pathway miR-135b-5p 通过靶向 CLIP4 介导的 JAK2/STAT3 信号通路促进胃癌发生。
IF 4.4 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-08-07 DOI: 10.1016/j.cellsig.2024.111339

Background

Gastric cancer (GC) is a common cancer worldwide; however, its molecular and pathogenic mechanisms remain unclear. MicroRNAs (miRNAs), which target key genes in GC, are associated with tumor promotion or suppression. Therefore, identifying new miRNA mechanisms could improve the novel diagnostic and therapeutic strategies for patients with GC.

Methods

To explore the biological functions of miR-135b-5p in GC, bioinformatic analysis and in vitro functional assays, including colony formation, wound healing, Transwell, and EdU assays, were used to assess the proliferative, invasive, and migratory capacities of GC cells. Target genes were predicted using RNA-seq and online databases. Dual-luciferase reporter assay, fluorescence in situ hybridization and western blotting were used to confirm the regulatory relationship between miR-135b-5p and CLIP4. The role of CLIP4 in tumor progression was assessed using clinical samples and both in vitro and in vivo assays. The tumor-suppressive mechanism of CLIP4 in GC was elucidated using rescue assays.

Results

Our study identified that miR-135b-5p as one of the top three over-expressed miRNAs in GC tissues, with RT-qPCR confirming its upregulation. Functional analysis showed that upregulated miR-135b-5p promoted malignant phenotypes in GC cells. Mechanistic research indicated that miR-135b-5p acts as a cancer promoter by targeting CLIP4. Moreover, our study suggested that CLIP4 exerts its tumor-suppressive function by inhibiting the JAK2/STAT3 signaling pathway.

Conclusion

This study reveals a novel mechanism by which miR-135b-5p exerts its tumor-promoting functions by targeting CLIP4. The tumor-suppressive function of CLIP4 by inactivating the JAK2/STAT3 pathway is also elucidated. Regulatory mechanism of CLIP4 by miR-135b-5p provides a promising novel therapeutic strategy for GC patients.

背景:胃癌(GC)是全球常见的癌症,但其分子和致病机制仍不清楚。微RNA(miRNA)靶向胃癌中的关键基因,与肿瘤的促进或抑制有关。因此,确定新的 miRNA 机制可改善 GC 患者的新型诊断和治疗策略:为了探索 miR-135b-5p 在 GC 中的生物学功能,研究人员利用生物信息学分析和体外功能测试(包括菌落形成、伤口愈合、Transwell 和 EdU 试验)来评估 GC 细胞的增殖、侵袭和迁移能力。利用 RNA-seq 和在线数据库预测了靶基因。利用双荧光素酶报告实验、荧光原位杂交和免疫印迹法证实了 miR-135b-5p 和 CLIP4 之间的调控关系。利用临床样本和体内外试验评估了CLIP4在肿瘤进展中的作用。利用挽救实验阐明了CLIP4在GC中的抑瘤机制:我们的研究发现,miR-135b-5p 是 GC 组织中表达量最高的三个 miRNA 之一,RT-qPCR 证实了它的上调。功能分析显示,上调的 miR-135b-5p 促进了 GC 细胞的恶性表型。机理研究表明,miR-135b-5p 通过靶向 CLIP4 起到促癌作用。此外,我们的研究还表明,CLIP4通过抑制JAK2/STAT3信号通路发挥其抑肿瘤功能:本研究揭示了 miR-135b-5p 通过靶向 CLIP4 发挥肿瘤促进功能的新机制。本研究揭示了 miR-135b-5p 通过靶向 CLIP4 发挥肿瘤促进功能的新机制,同时还阐明了 CLIP4 通过使 JAK2/STAT3 通路失活而发挥肿瘤抑制功能。miR-135b-5p 对 CLIP4 的调控机制为 GC 患者提供了一种前景广阔的新型治疗策略。
{"title":"miR-135b-5p promotes gastric carcinogenesis by targeting CLIP4-mediated JAK2/STAT3 signal pathway","authors":"","doi":"10.1016/j.cellsig.2024.111339","DOIUrl":"10.1016/j.cellsig.2024.111339","url":null,"abstract":"<div><h3>Background</h3><p>Gastric cancer (GC) is a common cancer worldwide; however, its molecular and pathogenic mechanisms remain unclear. MicroRNAs (miRNAs), which target key genes in GC, are associated with tumor promotion or suppression. Therefore, identifying new miRNA mechanisms could improve the novel diagnostic and therapeutic strategies for patients with GC.</p></div><div><h3>Methods</h3><p>To explore the biological functions of miR-135b-5p in GC, bioinformatic analysis and <em>in vitro</em> functional assays, including colony formation, wound healing, Transwell, and EdU assays, were used to assess the proliferative, invasive, and migratory capacities of GC cells. Target genes were predicted using RNA-seq and online databases. Dual-luciferase reporter assay, fluorescence <em>in situ</em> hybridization and western blotting were used to confirm the regulatory relationship between miR-135b-5p and CLIP4. The role of CLIP4 in tumor progression was assessed using clinical samples and both <em>in vitro</em> and <em>in vivo</em> assays. The tumor-suppressive mechanism of CLIP4 in GC was elucidated using rescue assays.</p></div><div><h3>Results</h3><p>Our study identified that miR-135b-5p as one of the top three over-expressed miRNAs in GC tissues, with RT-qPCR confirming its upregulation. Functional analysis showed that upregulated miR-135b-5p promoted malignant phenotypes in GC cells. Mechanistic research indicated that miR-135b-5p acts as a cancer promoter by targeting CLIP4. Moreover, our study suggested that CLIP4 exerts its tumor-suppressive function by inhibiting the JAK2/STAT3 signaling pathway.</p></div><div><h3>Conclusion</h3><p>This study reveals a novel mechanism by which miR-135b-5p exerts its tumor-promoting functions by targeting CLIP4. The tumor-suppressive function of CLIP4 by inactivating the JAK2/STAT3 pathway is also elucidated. Regulatory mechanism of CLIP4 by miR-135b-5p provides a promising novel therapeutic strategy for GC patients.</p></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141912024","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Sodium-glucose cotransporter 2 inhibitors and renin-angiotensin-aldosterone system, possible cellular interactions and benefits 钠-葡萄糖共转运体 2 抑制剂和肾素-血管紧张素-醛固酮系统,可能的细胞相互作用和益处。
IF 4.4 2区 生物学 Q2 CELL BIOLOGY Pub Date : 2024-08-06 DOI: 10.1016/j.cellsig.2024.111335

Sodium glucose cotransporter 2 inhibitors (SGLT2is) are a newly developed class of anti-diabetics which exert potent hypoglycemic effects in the diabetic milieu. However, the evidence suggests that they also have extra-glycemic effects. The renin-angiotensin-aldosterone system (RAAS) is a hormonal system widely distributed in the body that is important for water and electrolyte homeostasis as well as renal and cardiovascular function. Therefore, modulating RAAS activity is a main goal in patients, notably diabetic patients, which are at higher risk of complications involving these organ systems. Some studies have suggested that SGLT2is have modulatory effects on RAAS activity in addition to their hypoglycemic effects and, thus, these drugs can be considered as promising therapeutic agents for renal and cardiovascular disorders. However, the exact molecular interactions between SGLT2 inhibition and RAAS activity are not clearly understood. Therefore, in the current study we surveyed the literature for possible molecular mechanisms by which SGLT2is modulate RAAS activity.

葡萄糖钠共转运体 2 抑制剂(SGLT2is)是一类新开发的抗糖尿病药物,可在糖尿病环境中发挥强效降糖作用。然而,有证据表明,它们还具有降糖以外的作用。肾素-血管紧张素-醛固酮系统(RAAS)是一种广泛分布于体内的激素系统,对水和电解质平衡以及肾脏和心血管功能非常重要。因此,调节 RAAS 活性是糖尿病患者的一个主要目标,因为糖尿病患者发生这些器官系统并发症的风险较高。一些研究表明,SGLT2 除了具有降糖作用外,还具有调节 RAAS 活性的作用,因此,这些药物可被视为治疗肾脏和心血管疾病的有前途的药物。然而,SGLT2 抑制剂与 RAAS 活性之间的确切分子相互作用尚不清楚。因此,在本研究中,我们对文献进行了调查,以了解 SGLT2 调节 RAAS 活性的可能分子机制。
{"title":"Sodium-glucose cotransporter 2 inhibitors and renin-angiotensin-aldosterone system, possible cellular interactions and benefits","authors":"","doi":"10.1016/j.cellsig.2024.111335","DOIUrl":"10.1016/j.cellsig.2024.111335","url":null,"abstract":"<div><p>Sodium glucose cotransporter 2 inhibitors (SGLT2is) are a newly developed class of anti-diabetics which exert potent hypoglycemic effects in the diabetic milieu. However, the evidence suggests that they also have extra-glycemic effects. The renin-angiotensin-aldosterone system (RAAS) is a hormonal system widely distributed in the body that is important for water and electrolyte homeostasis as well as renal and cardiovascular function. Therefore, modulating RAAS activity is a main goal in patients, notably diabetic patients, which are at higher risk of complications involving these organ systems. Some studies have suggested that SGLT2is have modulatory effects on RAAS activity in addition to their hypoglycemic effects and, thus, these drugs can be considered as promising therapeutic agents for renal and cardiovascular disorders. However, the exact molecular interactions between SGLT2 inhibition and RAAS activity are not clearly understood. Therefore, in the current study we surveyed the literature for possible molecular mechanisms by which SGLT2is modulate RAAS activity.</p></div>","PeriodicalId":9902,"journal":{"name":"Cellular signalling","volume":null,"pages":null},"PeriodicalIF":4.4,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906106","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
期刊
Cellular signalling
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