首页 > 最新文献

Cell Communication and Signaling最新文献

英文 中文
Inhibition of SGLT2 protects podocytes in diabetic kidney disease by rebalancing mitochondria-associated endoplasmic reticulum membranes. 通过重新平衡线粒体相关内质网膜,抑制 SGLT2 可保护糖尿病肾病患者的荚膜细胞。
IF 5.4 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-07 DOI: 10.1186/s12964-024-01914-1
Xuehong Li, Qiong Li, Xinying Jiang, Shicong Song, Wei Zou, Qinglan Yang, Sirui Liu, Shuangqin Chen, Cheng Wang

Background: Sodium-glucose cotransporter 2 (SGLT2) inhibitors have changed the therapeutic landscape for diabetic kidney disease (DKD) patients, but their underlying mechanisms are complicated and not fully understood. Mitochondria-associated endoplasmic reticulum membranes (MAMs), the dynamic contact sites between mitochondria and the endoplasmic reticulum (ER), serve as intracellular platforms important for regulating cellular fate and function. This study explored the roles and mechanisms of SGLT2 inhibitors in regulating MAMs formation in diabetic podocytes.

Methods: We assessed MAMs formation in podocytes from DKD patients' renal biopsy samples and induced an increase in MAMs formation in cultured human podocytes by transfecting OMM-ER linker plasmid to investigate the effects of MAMs imbalance on podocyte injury. Empagliflozin-treated diabetic mice and podocyte-specific SGLT2 knockout diabetic mice (diabetic states were induced by streptozotocin and a high-fat diet), empagliflozin-treated podocytes, SGLT2-downregulated podocytes, and SGLT2-overexpressing podocytes were used to investigate the effects and mechanisms of SGLT2 inhibitors on MAMs formation in diabetic podocytes.

Results: MAMs were increased in podocytes and were associated with renal dysfunction in DKD patients. Increased MAMs aggravated HG-induced podocyte injury. The expression of SGLT2 was increased in diabetic podocytes. In addition, empagliflozin-treatment and podocyte-specific SGLT2 knockout attenuated MAMs formation and podocyte injury in diabetic mice. Empagliflozin treatment and SGLT2 knockdown decreased podocyte MAMs formation by activating the AMP-activated protein kinase (AMPK) pathway, while SGLT2 overexpression had the opposite effect.

Conclusions: Inhibition of SGLT2 attenuates MAMs imbalance in diabetic podocytes by activating the AMPK pathway. This study expands our knowledge of the roles of SGLT2 inhibitors in improving DKD podocyte injury and provides new insights into DKD treatment.

背景:钠-葡萄糖共转运体2(SGLT2)抑制剂改变了糖尿病肾病(DKD)患者的治疗格局,但其潜在机制却十分复杂,尚未完全明了。线粒体相关内质网膜(MAMs)是线粒体和内质网(ER)之间的动态接触点,是调节细胞命运和功能的重要细胞内平台。本研究探讨了 SGLT2 抑制剂在调节糖尿病荚膜细胞 MAMs 形成中的作用和机制:我们评估了来自 DKD 患者肾活检样本的荚膜细胞中 MAMs 的形成,并通过转染 OMM-ER 连接质粒诱导培养的人荚膜细胞中 MAMs 形成的增加,以研究 MAMs 失衡对荚膜细胞损伤的影响。利用恩格列净处理的糖尿病小鼠和荚膜特异性SGLT2基因敲除糖尿病小鼠(通过链脲佐菌素和高脂饮食诱导糖尿病状态)、恩格列净处理的荚膜、SGLT2下调的荚膜和SGLT2表达的荚膜,研究SGLT2抑制剂对糖尿病荚膜MAMs形成的影响和机制:结果:MAMs在荚膜细胞中增加,并与DKD患者的肾功能障碍有关。MAMs 的增加加重了 HG 诱导的荚膜损伤。糖尿病荚膜细胞中 SGLT2 的表达增加。此外,恩格列净治疗和荚膜特异性 SGLT2 基因敲除可减轻糖尿病小鼠 MAMs 的形成和荚膜损伤。Empagliflozin 治疗和 SGLT2 基因敲除通过激活 AMP 激活蛋白激酶(AMPK)通路减少了荚膜 MAMs 的形成,而 SGLT2 基因过表达则产生了相反的效果:结论:通过激活 AMPK 通路,抑制 SGLT2 可减轻糖尿病荚膜细胞中 MAMs 的失衡。这项研究拓展了我们对 SGLT2 抑制剂在改善糖尿病荚膜细胞损伤中的作用的认识,并为糖尿病荚膜细胞损伤的治疗提供了新的见解。
{"title":"Inhibition of SGLT2 protects podocytes in diabetic kidney disease by rebalancing mitochondria-associated endoplasmic reticulum membranes.","authors":"Xuehong Li, Qiong Li, Xinying Jiang, Shicong Song, Wei Zou, Qinglan Yang, Sirui Liu, Shuangqin Chen, Cheng Wang","doi":"10.1186/s12964-024-01914-1","DOIUrl":"10.1186/s12964-024-01914-1","url":null,"abstract":"<p><strong>Background: </strong>Sodium-glucose cotransporter 2 (SGLT2) inhibitors have changed the therapeutic landscape for diabetic kidney disease (DKD) patients, but their underlying mechanisms are complicated and not fully understood. Mitochondria-associated endoplasmic reticulum membranes (MAMs), the dynamic contact sites between mitochondria and the endoplasmic reticulum (ER), serve as intracellular platforms important for regulating cellular fate and function. This study explored the roles and mechanisms of SGLT2 inhibitors in regulating MAMs formation in diabetic podocytes.</p><p><strong>Methods: </strong>We assessed MAMs formation in podocytes from DKD patients' renal biopsy samples and induced an increase in MAMs formation in cultured human podocytes by transfecting OMM-ER linker plasmid to investigate the effects of MAMs imbalance on podocyte injury. Empagliflozin-treated diabetic mice and podocyte-specific SGLT2 knockout diabetic mice (diabetic states were induced by streptozotocin and a high-fat diet), empagliflozin-treated podocytes, SGLT2-downregulated podocytes, and SGLT2-overexpressing podocytes were used to investigate the effects and mechanisms of SGLT2 inhibitors on MAMs formation in diabetic podocytes.</p><p><strong>Results: </strong>MAMs were increased in podocytes and were associated with renal dysfunction in DKD patients. Increased MAMs aggravated HG-induced podocyte injury. The expression of SGLT2 was increased in diabetic podocytes. In addition, empagliflozin-treatment and podocyte-specific SGLT2 knockout attenuated MAMs formation and podocyte injury in diabetic mice. Empagliflozin treatment and SGLT2 knockdown decreased podocyte MAMs formation by activating the AMP-activated protein kinase (AMPK) pathway, while SGLT2 overexpression had the opposite effect.</p><p><strong>Conclusions: </strong>Inhibition of SGLT2 attenuates MAMs imbalance in diabetic podocytes by activating the AMPK pathway. This study expands our knowledge of the roles of SGLT2 inhibitors in improving DKD podocyte injury and provides new insights into DKD treatment.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"534"},"PeriodicalIF":5.4,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11542362/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142607526","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
RNF13 protects neurons against ischemia-reperfusion injury via stabilizing p62-mediated Nrf2/HO-1 signaling pathway. RNF13 通过稳定 p62 介导的 Nrf2/HO-1 信号通路,保护神经元免受缺血再灌注损伤。
IF 5.4 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-07 DOI: 10.1186/s12964-024-01905-2
Qiangping Wang, Shuang Li, Wenjie Wu, Wenke Zhou, Kaixuan Yan, Zhen Liu, Lanlan Yan, Baoping Zheng, Fangcheng Zhang, Xiaobing Jiang, Youfan Ye, Haijun Wang

Background: Cerebral ischemia/reperfusion injury (CIRI), a common, universal clinical problem that costs a large proportion of the economic and disease burden. Identifying the key regulators of cerebral I/R injury could provide potential strategies for clinically improving the prognosis of stroke. Ring finger protein 13 (RNF13) has been proven to be involved in the inflammatory response. Here, we aimed to identify the role of RNF13 in cerebral I/R injury and further reveal its immanent mechanisms.

Methods: The CRISPR/Cas9 based knockout mice, RNA sequencing, mass spectrometry, co-immunoprecipitation, GST-pull down, immunofluorescent staining, western blot, RT-PCR were used to investigate biodistribution, function and mechanism of RNF13 during cerebral I/R injury.

Results: In the present study, we found that RNF13 was significantly up-regulated in patients, mice and primary neurons after I/R injury. Deficiency of RNF13 aggravated I/R-induced neurological impairment, inflammatory response and apoptosis while overexpression of RNF13 inhibited I/R injury. Mechanistically, this inhibitory effect of RNF13 during I/R injury was confirmed to be dependent on the blocking of TRIM21-mediated autophagy-dependent degradation of p62 and the stabilization of the p62-mediated Nrf2/HO-1 signaling pathway.

Conclusion: RNF13 is a crucial regulator of cerebral I/R injury that plays its role in inhibiting cell apoptosis and inflammatory response by preventing the autophagy-medicated degradation of the p62/Nrf2/HO-1 signaling pathway via blocking the interaction of TRIM21-p62 complex. Therefore, RNF13 represents a potential pharmacological target in acute ischemia stroke therapy.

背景:脑缺血/再灌注损伤(CIRI)是一种常见的、普遍的临床问题,在经济和疾病负担中占很大比例。确定脑缺血再灌注损伤的关键调节因子可为临床改善中风预后提供潜在策略。环指蛋白 13(RNF13)已被证实参与了炎症反应。在此,我们旨在确定 RNF13 在脑 I/R 损伤中的作用,并进一步揭示其内在机制:方法:采用基于CRISPR/Cas9的基因敲除小鼠、RNA测序、质谱分析、共免疫沉淀、GST-pull down、免疫荧光染色、Western blot、RT-PCR等方法研究RNF13在脑I/R损伤中的生物分布、功能和机制:结果:本研究发现,RNF13在患者、小鼠和原代神经元I/R损伤后显著上调。缺乏 RNF13 会加重 I/R 引起的神经损伤、炎症反应和细胞凋亡,而过表达 RNF13 则会抑制 I/R 损伤。从机理上讲,RNF13在I/R损伤期间的这种抑制作用被证实依赖于阻断TRIM21介导的依赖于自噬的p62降解和稳定p62介导的Nrf2/HO-1信号通路:RNF13是脑I/R损伤的重要调节因子,它通过阻断TRIM21-p62复合物的相互作用,防止p62/Nrf2/HO-1信号通路的自噬降解,从而在抑制细胞凋亡和炎症反应中发挥作用。因此,RNF13 是治疗急性缺血性脑卒中的潜在药物靶点。
{"title":"RNF13 protects neurons against ischemia-reperfusion injury via stabilizing p62-mediated Nrf2/HO-1 signaling pathway.","authors":"Qiangping Wang, Shuang Li, Wenjie Wu, Wenke Zhou, Kaixuan Yan, Zhen Liu, Lanlan Yan, Baoping Zheng, Fangcheng Zhang, Xiaobing Jiang, Youfan Ye, Haijun Wang","doi":"10.1186/s12964-024-01905-2","DOIUrl":"10.1186/s12964-024-01905-2","url":null,"abstract":"<p><strong>Background: </strong>Cerebral ischemia/reperfusion injury (CIRI), a common, universal clinical problem that costs a large proportion of the economic and disease burden. Identifying the key regulators of cerebral I/R injury could provide potential strategies for clinically improving the prognosis of stroke. Ring finger protein 13 (RNF13) has been proven to be involved in the inflammatory response. Here, we aimed to identify the role of RNF13 in cerebral I/R injury and further reveal its immanent mechanisms.</p><p><strong>Methods: </strong>The CRISPR/Cas9 based knockout mice, RNA sequencing, mass spectrometry, co-immunoprecipitation, GST-pull down, immunofluorescent staining, western blot, RT-PCR were used to investigate biodistribution, function and mechanism of RNF13 during cerebral I/R injury.</p><p><strong>Results: </strong>In the present study, we found that RNF13 was significantly up-regulated in patients, mice and primary neurons after I/R injury. Deficiency of RNF13 aggravated I/R-induced neurological impairment, inflammatory response and apoptosis while overexpression of RNF13 inhibited I/R injury. Mechanistically, this inhibitory effect of RNF13 during I/R injury was confirmed to be dependent on the blocking of TRIM21-mediated autophagy-dependent degradation of p62 and the stabilization of the p62-mediated Nrf2/HO-1 signaling pathway.</p><p><strong>Conclusion: </strong>RNF13 is a crucial regulator of cerebral I/R injury that plays its role in inhibiting cell apoptosis and inflammatory response by preventing the autophagy-medicated degradation of the p62/Nrf2/HO-1 signaling pathway via blocking the interaction of TRIM21-p62 complex. Therefore, RNF13 represents a potential pharmacological target in acute ischemia stroke therapy.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"535"},"PeriodicalIF":5.4,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11542339/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142607527","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
Breast cancer cells utilize T3 to trigger proliferation through cellular Ca2+ modulation. 乳腺癌细胞利用 T3 通过细胞 Ca2+ 调节引发增殖。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-06 DOI: 10.1186/s12964-024-01917-y
Ines Tawfik, Katharina Schlick, Julian Ostaku, Doruntina Bresilla, Sonja Gabrijelčič, Benjamin Gottschalk, Alwin Sokolowski, Ernst Malle, Katarina Kalinova, Martin Hirtl, Corina T Madreiter-Sokolowski

High levels of thyroid hormones are linked to increased risk and advanced stages of breast cancer. Our previous work demonstrated that the biologically active triiodothyronine (T3) facilitates mitochondrial ATP production by upregulating Ca2+ handling proteins, thereby boosting mitochondrial Ca2+ uptake and Krebs cycle activity. In this study, different cell types were utilized to investigate whether T3 activates a Ca2+-induced signaling pathway to boost cancer cell proliferation. Using live-cell imaging, biochemical assays, and molecular profiling, differences in intracellular signaling among MCF7 and MDA-MB-468 breast cancer cells, non-cancerous breast cells hTERT-HME1, and PC3 prostate carcinoma cells, previously found to be insensitive to thyroid hormones in terms of proliferation, were investigated. Our findings revealed that T3 upregulates 1,4,5-trisphosphate receptor 3 via thyroid hormone receptor α. This boosts mitochondrial Ca2+ uptake, reduction equivalent yield, and mitochondrial ATP production, supporting the viability and proliferation of breast cancer cells without affecting non-cancerous hTERT-HME1 or PC3 prostate carcinoma cells. Understanding the interplay between T3 signaling, organellar interaction, and breast cancer metabolism could lead to targeted therapies that exploit cancer cell vulnerabilities. Our findings highlight T3 as a crucial regulator of cancer metabolism, reinforcing its potential as a therapeutic target in breast cancer.

高水平的甲状腺激素与乳腺癌的风险增加和晚期阶段有关。我们之前的研究表明,具有生物活性的三碘甲状腺原氨酸(T3)通过上调 Ca2+ 处理蛋白促进线粒体 ATP 的产生,从而提高线粒体 Ca2+ 摄取和克雷布斯循环的活性。本研究利用不同类型的细胞来研究 T3 是否会激活 Ca2+ 诱导的信号通路,从而促进癌细胞增殖。通过活细胞成像、生化检测和分子谱分析,研究了MCF7和MDA-MB-468乳腺癌细胞、非癌乳腺细胞hTERT-HME1以及PC3前列腺癌细胞细胞内信号传导的差异。我们的研究结果表明,T3通过甲状腺激素受体α上调1,4,5-三磷酸受体3,从而促进线粒体Ca2+摄取、还原当量产量和线粒体ATP生成,支持乳腺癌细胞的活力和增殖,而不影响非癌细胞hTERT-HME1或PC3前列腺癌细胞。了解 T3 信号、细胞器相互作用和乳腺癌新陈代谢之间的相互作用,可以开发出利用癌细胞弱点的靶向疗法。我们的研究结果突显了 T3 是癌症代谢的关键调节因子,增强了其作为乳腺癌治疗靶点的潜力。
{"title":"Breast cancer cells utilize T3 to trigger proliferation through cellular Ca<sup>2+</sup> modulation.","authors":"Ines Tawfik, Katharina Schlick, Julian Ostaku, Doruntina Bresilla, Sonja Gabrijelčič, Benjamin Gottschalk, Alwin Sokolowski, Ernst Malle, Katarina Kalinova, Martin Hirtl, Corina T Madreiter-Sokolowski","doi":"10.1186/s12964-024-01917-y","DOIUrl":"10.1186/s12964-024-01917-y","url":null,"abstract":"<p><p>High levels of thyroid hormones are linked to increased risk and advanced stages of breast cancer. Our previous work demonstrated that the biologically active triiodothyronine (T3) facilitates mitochondrial ATP production by upregulating Ca<sup>2+</sup> handling proteins, thereby boosting mitochondrial Ca<sup>2+</sup> uptake and Krebs cycle activity. In this study, different cell types were utilized to investigate whether T3 activates a Ca<sup>2+</sup>-induced signaling pathway to boost cancer cell proliferation. Using live-cell imaging, biochemical assays, and molecular profiling, differences in intracellular signaling among MCF7 and MDA-MB-468 breast cancer cells, non-cancerous breast cells hTERT-HME1, and PC3 prostate carcinoma cells, previously found to be insensitive to thyroid hormones in terms of proliferation, were investigated. Our findings revealed that T3 upregulates 1,4,5-trisphosphate receptor 3 via thyroid hormone receptor α. This boosts mitochondrial Ca<sup>2+</sup> uptake, reduction equivalent yield, and mitochondrial ATP production, supporting the viability and proliferation of breast cancer cells without affecting non-cancerous hTERT-HME1 or PC3 prostate carcinoma cells. Understanding the interplay between T3 signaling, organellar interaction, and breast cancer metabolism could lead to targeted therapies that exploit cancer cell vulnerabilities. Our findings highlight T3 as a crucial regulator of cancer metabolism, reinforcing its potential as a therapeutic target in breast cancer.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"533"},"PeriodicalIF":8.2,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11539297/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142591070","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
Atomoxetine suppresses radioresistance in glioblastoma via circATIC/miR-520d-5p/Notch2-Hey1 axis. 阿托莫西汀通过circATIC/miR-520d-5p/Notch2-Hey1轴抑制胶质母细胞瘤的放射抗性
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-05 DOI: 10.1186/s12964-024-01915-0
Hyun Jeong Seok, Jae Yeon Choi, Dong Hyeon Lee, Incheol Shin, In Hwa Bae

Background: Resistance acquired after radiotherapy is directly related to the failure of various cancer treatments, including GBM. Because the mechanism for overcoming radioresistance has not yet been clearly identified, the development of diagnostic and therapeutic markers to treat radioresistance is necessary. Since increased expression of stemness- and EMT-related markers are reported to be closely correlated with radioresistance, research is underway to develop new drugs targeting these factors.

Methods: To develop an anticancer drug that overcomes radioresistance, a library of drugs already approved by the FDA was used. After treating radioresistant GBM cells with each drug, the expression of stemness- and EMT-related markers was confirmed by qRT-PCR, and as a result, Atomoxetine (ATX) was selected. It was confirmed that radioresistance-induced cell migratory, invasive, sphere formation abilities, and tumor growth using a xenograft mouse model were suppressed upon ATX treatment. Using a miRNA prediction tool, we discovered miR-520d-5p, which targets Notch2 and Hey1, key factors in radioresistance, and discovered circATIC targeting this miRNA, revealing its relationship with ATX. We demonstrated the expression regulation mechanism and signaling mechanism between circATIC, miR-520d-5p, Notch2, and Hey1 factors using a luciferase reporter assay. In addition, the results at the cellular level were clinically verified by confirming the correlation between radiation, miR-520d-5p, and circATIC using patient plasma by qRT-PCR.

Results: ATX showed potential as a treatment for radioresistance by suppressing the malignant phenotype by regulating the circATIC/miR-520d-5p/Notch2-Hey1 signaling mechanism in vitro and in vivo using radioresistant GBM cells.

Conclusions: This study revealed that ATX suppresses radioresistance through the circATIC/miR-520d-5p/Notch2-Hey1 signaling pathway. These results showed the potential of ATX as a new drug that can overcome radioresistance, a major challenge in cancer treatment, and the signaling factors identified in this mechanism suggest the possibility of use as potential targets for the diagnosis and treatment of radioresistance.

背景:放疗后产生的抗药性与各种癌症治疗的失败直接相关,包括肿瘤坏死因子转移抑制剂(GBM)。由于克服放射抗性的机制尚未明确,因此有必要开发治疗放射抗性的诊断和治疗标记物。据报道,干性和 EMT 相关标记物的表达增加与放射抗性密切相关,因此目前正在研究开发针对这些因素的新药:方法:为了开发克服放射抗药性的抗癌药物,我们使用了一个已获美国食品及药物管理局批准的药物库。用每种药物处理具有放射抗性的GBM细胞后,通过qRT-PCR证实了干性和EMT相关标记物的表达,结果选择了阿托莫西汀(ATX)。实验证实,ATX治疗后,放射抗性诱导的细胞迁移、侵袭、球形成能力以及异种移植小鼠模型的肿瘤生长均受到抑制。利用 miRNA 预测工具,我们发现了靶向放射抗性关键因子 Notch2 和 Hey1 的 miR-520d-5p,并发现了靶向该 miRNA 的 circATIC,揭示了其与 ATX 的关系。我们利用荧光素酶报告实验证明了circATIC、miR-520d-5p、Notch2和Hey1因子之间的表达调控机制和信号转导机制。此外,通过使用患者血浆进行 qRT-PCR,证实了辐射、miR-520d-5p 和 circATIC 之间的相关性,从而在临床上验证了细胞水平的结果:结果:ATX通过调节circATIC/miR-520d-5p/Notch2-Hey1信号机制,在体外和体内使用耐放射的GBM细胞抑制恶性表型,显示出治疗放射耐药性的潜力:本研究揭示了ATX通过circATIC/miR-520d-5p/Notch2-Hey1信号通路抑制放射抗性。这些结果表明,ATX有可能成为克服癌症治疗中的一大挑战--放射抗性的新药,而在这一机制中发现的信号传导因子表明,ATX有可能成为诊断和治疗放射抗性的潜在靶点。
{"title":"Atomoxetine suppresses radioresistance in glioblastoma via circATIC/miR-520d-5p/Notch2-Hey1 axis.","authors":"Hyun Jeong Seok, Jae Yeon Choi, Dong Hyeon Lee, Incheol Shin, In Hwa Bae","doi":"10.1186/s12964-024-01915-0","DOIUrl":"10.1186/s12964-024-01915-0","url":null,"abstract":"<p><strong>Background: </strong>Resistance acquired after radiotherapy is directly related to the failure of various cancer treatments, including GBM. Because the mechanism for overcoming radioresistance has not yet been clearly identified, the development of diagnostic and therapeutic markers to treat radioresistance is necessary. Since increased expression of stemness- and EMT-related markers are reported to be closely correlated with radioresistance, research is underway to develop new drugs targeting these factors.</p><p><strong>Methods: </strong>To develop an anticancer drug that overcomes radioresistance, a library of drugs already approved by the FDA was used. After treating radioresistant GBM cells with each drug, the expression of stemness- and EMT-related markers was confirmed by qRT-PCR, and as a result, Atomoxetine (ATX) was selected. It was confirmed that radioresistance-induced cell migratory, invasive, sphere formation abilities, and tumor growth using a xenograft mouse model were suppressed upon ATX treatment. Using a miRNA prediction tool, we discovered miR-520d-5p, which targets Notch2 and Hey1, key factors in radioresistance, and discovered circATIC targeting this miRNA, revealing its relationship with ATX. We demonstrated the expression regulation mechanism and signaling mechanism between circATIC, miR-520d-5p, Notch2, and Hey1 factors using a luciferase reporter assay. In addition, the results at the cellular level were clinically verified by confirming the correlation between radiation, miR-520d-5p, and circATIC using patient plasma by qRT-PCR.</p><p><strong>Results: </strong>ATX showed potential as a treatment for radioresistance by suppressing the malignant phenotype by regulating the circATIC/miR-520d-5p/Notch2-Hey1 signaling mechanism in vitro and in vivo using radioresistant GBM cells.</p><p><strong>Conclusions: </strong>This study revealed that ATX suppresses radioresistance through the circATIC/miR-520d-5p/Notch2-Hey1 signaling pathway. These results showed the potential of ATX as a new drug that can overcome radioresistance, a major challenge in cancer treatment, and the signaling factors identified in this mechanism suggest the possibility of use as potential targets for the diagnosis and treatment of radioresistance.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"532"},"PeriodicalIF":8.2,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11536942/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142584950","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
Regulation of ADAM10 activity through microdomain-dependent intracellular calcium changes. 通过微域依赖性细胞内钙变化调节 ADAM10 的活性。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-04 DOI: 10.1186/s12964-024-01891-5
Federico Guillermo Gharzia, Ahmad Aljohmani, Andreas Beck, Stephan E Philipp, Daniela Yildiz

A disintegrin and metalloproteinases (ADAMs) are transmembrane proteases that cleave other proteins close to the surface in a process called shedding. The prominent member ADAM10 has been linked to several pathologies such as Alzheimer's disease, bacterial infection, cancer development and metastasis. Although the regulation of the ADAM10 activity by calcium influx and calmodulin inhibition has been reported, the spatiotemporal regulation of Ca2+-dependent ADAM10 activation and the required source of Ca2+ ions have not been thoroughly studied. In the present study, we observed the rapid Ca2+-dependent activation of ADAM10 in A549 lung carcinoma cells upon stimulation with ionomycin. The calmodulin-inhibitors trifluoperazine and ophiobolin A mediated delayed activation of ADAM10, which apparently did not depend on intracellular Ca2+ in the case of trifluoperazine. Furthermore, the surface translocation and release of ADAM10 in extracellular vesicles exhibited different kinetics and were only partially linked to catalytic activation. Finally, ADAM10 activation was observed after the entry of Ca2+ through certain channels, such as canonical members of transient receptor potential (TRP) channels. Therefore, the opening of particular channels for Ca2+ entry points and subsequent Ca2+ flux as well as the temporal aspects of the consequent increase in Ca2+ levels, must be considered for future therapeutic options involving the increasing or decreasing ADAM10 activity.

崩解酶和金属蛋白酶(ADAMs)是一种跨膜蛋白酶,能在一种称为脱落的过程中裂解靠近表面的其他蛋白质。其主要成员 ADAM10 与阿尔茨海默病、细菌感染、癌症发展和转移等多种病症有关。虽然钙离子流入和钙调素抑制对 ADAM10 活性的调控已有报道,但对 Ca2+ 依赖性 ADAM10 激活的时空调控和所需的 Ca2+ 离子源尚未进行深入研究。在本研究中,我们观察到 A549 肺癌细胞在受到离子霉素刺激后,ADAM10 会迅速被 Ca2+ 依赖性激活。钙调蛋白抑制剂三氟拉嗪和奥菲波林 A 可延缓 ADAM10 的活化,其中三氟拉嗪的活化显然不依赖于细胞内 Ca2+。此外,ADAM10 在细胞外囊泡中的表面转运和释放表现出不同的动力学,并且只与催化活化有部分联系。最后,通过某些通道(如瞬时受体电位(TRP)通道的典型成员)进入 Ca2+ 后,也能观察到 ADAM10 的活化。因此,在未来涉及增加或减少 ADAM10 活性的治疗方案中,必须考虑 Ca2+ 进入点特定通道的开放、随后的 Ca2+ 通量以及 Ca2+ 水平随之增加的时间方面。
{"title":"Regulation of ADAM10 activity through microdomain-dependent intracellular calcium changes.","authors":"Federico Guillermo Gharzia, Ahmad Aljohmani, Andreas Beck, Stephan E Philipp, Daniela Yildiz","doi":"10.1186/s12964-024-01891-5","DOIUrl":"10.1186/s12964-024-01891-5","url":null,"abstract":"<p><p>A disintegrin and metalloproteinases (ADAMs) are transmembrane proteases that cleave other proteins close to the surface in a process called shedding. The prominent member ADAM10 has been linked to several pathologies such as Alzheimer's disease, bacterial infection, cancer development and metastasis. Although the regulation of the ADAM10 activity by calcium influx and calmodulin inhibition has been reported, the spatiotemporal regulation of Ca<sup>2+</sup>-dependent ADAM10 activation and the required source of Ca<sup>2+</sup> ions have not been thoroughly studied. In the present study, we observed the rapid Ca<sup>2+</sup>-dependent activation of ADAM10 in A549 lung carcinoma cells upon stimulation with ionomycin. The calmodulin-inhibitors trifluoperazine and ophiobolin A mediated delayed activation of ADAM10, which apparently did not depend on intracellular Ca<sup>2+</sup> in the case of trifluoperazine. Furthermore, the surface translocation and release of ADAM10 in extracellular vesicles exhibited different kinetics and were only partially linked to catalytic activation. Finally, ADAM10 activation was observed after the entry of Ca<sup>2+</sup> through certain channels, such as canonical members of transient receptor potential (TRP) channels. Therefore, the opening of particular channels for Ca<sup>2+</sup> entry points and subsequent Ca<sup>2+</sup> flux as well as the temporal aspects of the consequent increase in Ca<sup>2+</sup> levels, must be considered for future therapeutic options involving the increasing or decreasing ADAM10 activity.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"531"},"PeriodicalIF":8.2,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11533308/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142577158","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
Blue light-driven cell cycle arrest in thyroid cancer via Retinal-OPN3 complex. 蓝光通过视网膜-OPN3复合物驱动甲状腺癌细胞周期停滞。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-01 DOI: 10.1186/s12964-024-01908-z
Changrui Zhao, Jiaqiang Bo, Tianyu Li, Jiameng Tian, Tian Long, Yingying He, Siyu Chen, Chang Liu

Background: Papillary thyroid carcinoma (PTC) is the most common type of thyroid malignancy, with a rising incidence. Traditional treatments, such as thyroidectomy and radiotherapy, often lead to significant side effects, including impaired thyroid function. Therefore, there is an urgent need for non-invasive therapeutic approaches. This study aims to explore the potential of photobiomodulation therapy (PBMT), a non-invasive treatment using specific wavelengths of light, in the management of PTC.

Methods: We investigated the effects of blue light PBMT on PTC cells, focusing on the Retinal-OPSIN 3 (OPN3) complex's role in mediating cellular responses. Blue light exposure was applied to PTC cells, and subsequent changes in cellular proliferation, cell cycle progression, and protein expression were analyzed. Statistical tests, including one-way ANOVA and t-tests, were used to evaluate the significance of the findings.

Results: Blue light exposure led to the dissociation of 11-cis-retinal from OPN3, resulting in the accumulation of all-trans retinal. This accumulation disrupted cellular proliferation pathways and induced G0/G1 cell cycle arrest in PTC cells. The Retinal-OPN3 complex was found to be a key mediator in these processes, demonstrating that thyroid cells can respond to specific light wavelengths and utilize their photoreceptive potential for therapeutic purposes.

Conclusions: Our findings suggest that PBMT, through the modulation of the Retinal-OPN3 complex, offers a promising non-invasive approach for treating PTC. This study highlights the therapeutic potential of light signal transduction in non-ocular tissues and opens new avenues for non-invasive cancer therapies.

背景:甲状腺乳头状癌(PTC甲状腺乳头状癌(PTC)是最常见的甲状腺恶性肿瘤,发病率呈上升趋势。传统的治疗方法,如甲状腺切除术和放射治疗,往往会导致严重的副作用,包括甲状腺功能受损。因此,迫切需要非侵入性的治疗方法。本研究旨在探索光生物调节疗法(PBMT)在治疗 PTC 方面的潜力:我们研究了蓝光 PBMT 对 PTC 细胞的影响,重点是视网膜-OPSIN 3(OPN3)复合物在介导细胞反应中的作用。对 PTC 细胞进行蓝光照射,分析细胞增殖、细胞周期进展和蛋白质表达的后续变化。统计检验包括单因素方差分析和 t 检验,以评估研究结果的显著性:结果:蓝光照射导致 11-顺式视网膜与 OPN3 分离,造成全反式视网膜的积累。这种积累破坏了细胞增殖途径,并诱导 PTC 细胞的 G0/G1 细胞周期停滞。研究发现,视网膜-OPN3复合物是这些过程中的关键介质,这表明甲状腺细胞能对特定波长的光做出反应,并利用其感光潜能达到治疗目的:我们的研究结果表明,PBMT 通过调节视网膜-OPN3 复合物,为治疗 PTC 提供了一种前景广阔的非侵入性方法。这项研究凸显了非眼组织中光信号转导的治疗潜力,为非侵入性癌症疗法开辟了新途径。
{"title":"Blue light-driven cell cycle arrest in thyroid cancer via Retinal-OPN3 complex.","authors":"Changrui Zhao, Jiaqiang Bo, Tianyu Li, Jiameng Tian, Tian Long, Yingying He, Siyu Chen, Chang Liu","doi":"10.1186/s12964-024-01908-z","DOIUrl":"10.1186/s12964-024-01908-z","url":null,"abstract":"<p><strong>Background: </strong>Papillary thyroid carcinoma (PTC) is the most common type of thyroid malignancy, with a rising incidence. Traditional treatments, such as thyroidectomy and radiotherapy, often lead to significant side effects, including impaired thyroid function. Therefore, there is an urgent need for non-invasive therapeutic approaches. This study aims to explore the potential of photobiomodulation therapy (PBMT), a non-invasive treatment using specific wavelengths of light, in the management of PTC.</p><p><strong>Methods: </strong>We investigated the effects of blue light PBMT on PTC cells, focusing on the Retinal-OPSIN 3 (OPN3) complex's role in mediating cellular responses. Blue light exposure was applied to PTC cells, and subsequent changes in cellular proliferation, cell cycle progression, and protein expression were analyzed. Statistical tests, including one-way ANOVA and t-tests, were used to evaluate the significance of the findings.</p><p><strong>Results: </strong>Blue light exposure led to the dissociation of 11-cis-retinal from OPN3, resulting in the accumulation of all-trans retinal. This accumulation disrupted cellular proliferation pathways and induced G0/G1 cell cycle arrest in PTC cells. The Retinal-OPN3 complex was found to be a key mediator in these processes, demonstrating that thyroid cells can respond to specific light wavelengths and utilize their photoreceptive potential for therapeutic purposes.</p><p><strong>Conclusions: </strong>Our findings suggest that PBMT, through the modulation of the Retinal-OPN3 complex, offers a promising non-invasive approach for treating PTC. This study highlights the therapeutic potential of light signal transduction in non-ocular tissues and opens new avenues for non-invasive cancer therapies.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"530"},"PeriodicalIF":8.2,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11531186/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142565096","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
PIM1 kinase and its diverse substrate in solid tumors. PIM1 激酶及其在实体瘤中的多种底物。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-01 DOI: 10.1186/s12964-024-01898-y
Rituparna Choudhury, Chandan Kumar Bahadi, Ipsa Pratibimbita Ray, Pragyanshree Dash, Isha Pattanaik, Suman Mishra, Soumya R Mohapatra, Srinivas Patnaik, Kumar Nikhil

The PIM kinase family, consisting of PIM1, PIM2, and PIM3, is a group of serine/threonine protein kinases crucial for cellular growth, immunoregulation, and oncogenesis. PIM1 kinase is often overexpressed in solid and hematopoietic malignancies, promoting cell survival, proliferation, migration, and senescence by activating key genes. In vitro and in vivo studies have established the oncogenic potential of PIM1 kinases. These kinases have been implicated in tumor progression, metastasis, and resistance to chemotherapy, underscoring their potential as a therapeutic target for cancer therapy. This review delves into the intricate molecular mechanisms through which PIM1 interacts with specific substrates in different tumor tissues, leading to diverse outcomes in various human cancers. Over the past decade, the inhibition of PIM1 in cancers has garnered significant attention as a potential standalone treatment. Various in vitro, in vivo, and early clinical trial data have provided support for this approach to varying extents. Novel compounds that inhibit PIM1 kinase have shown effectiveness and a favorable toxicity profile in preclinical studies. Several of these substances are now being studied in clinical trials due to their promising outcomes. This article provides a thorough examination of the PIM1 kinase pathways and the recent advancements in producing PIM1 kinase inhibitors for the treatment of cancer.

由 PIM1、PIM2 和 PIM3 组成的 PIM 激酶家族是一组丝氨酸/苏氨酸蛋白激酶,对细胞生长、免疫调节和肿瘤发生至关重要。PIM1 激酶通常在实体瘤和造血恶性肿瘤中过度表达,通过激活关键基因促进细胞存活、增殖、迁移和衰老。体外和体内研究证实了 PIM1 激酶的致癌潜力。这些激酶与肿瘤的进展、转移和对化疗的耐受性有关,凸显了它们作为癌症治疗靶点的潜力。本综述深入探讨了 PIM1 与不同肿瘤组织中特定底物相互作用的复杂分子机制,这些机制导致了各种人类癌症的不同结果。在过去十年中,抑制癌症中的 PIM1 作为一种潜在的独立治疗方法引起了广泛关注。各种体外、体内和早期临床试验数据为这种方法提供了不同程度的支持。在临床前研究中,抑制 PIM1 激酶的新型化合物已显示出有效性和良好的毒性特征。其中几种物质由于效果良好,目前正在进行临床试验研究。本文深入探讨了 PIM1 激酶通路以及生产用于治疗癌症的 PIM1 激酶抑制剂的最新进展。
{"title":"PIM1 kinase and its diverse substrate in solid tumors.","authors":"Rituparna Choudhury, Chandan Kumar Bahadi, Ipsa Pratibimbita Ray, Pragyanshree Dash, Isha Pattanaik, Suman Mishra, Soumya R Mohapatra, Srinivas Patnaik, Kumar Nikhil","doi":"10.1186/s12964-024-01898-y","DOIUrl":"10.1186/s12964-024-01898-y","url":null,"abstract":"<p><p>The PIM kinase family, consisting of PIM1, PIM2, and PIM3, is a group of serine/threonine protein kinases crucial for cellular growth, immunoregulation, and oncogenesis. PIM1 kinase is often overexpressed in solid and hematopoietic malignancies, promoting cell survival, proliferation, migration, and senescence by activating key genes. In vitro and in vivo studies have established the oncogenic potential of PIM1 kinases. These kinases have been implicated in tumor progression, metastasis, and resistance to chemotherapy, underscoring their potential as a therapeutic target for cancer therapy. This review delves into the intricate molecular mechanisms through which PIM1 interacts with specific substrates in different tumor tissues, leading to diverse outcomes in various human cancers. Over the past decade, the inhibition of PIM1 in cancers has garnered significant attention as a potential standalone treatment. Various in vitro, in vivo, and early clinical trial data have provided support for this approach to varying extents. Novel compounds that inhibit PIM1 kinase have shown effectiveness and a favorable toxicity profile in preclinical studies. Several of these substances are now being studied in clinical trials due to their promising outcomes. This article provides a thorough examination of the PIM1 kinase pathways and the recent advancements in producing PIM1 kinase inhibitors for the treatment of cancer.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"529"},"PeriodicalIF":8.2,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11531143/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142565098","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
Exosome-based immunotherapy as an innovative therapeutic approach in melanoma. 基于外泌体的免疫疗法是治疗黑色素瘤的一种创新方法。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-31 DOI: 10.1186/s12964-024-01906-1
Shabnam Babaei, Manouchehr Fadaee, Hajar Abbasi-Kenarsari, Dariush Shanehbandi, Tohid Kazemi

The malignant form of melanoma is one of the deadliest human cancers that accounts for almost all of the skin tumor-related fatalities in its later stages. Achieving an exhaustive understanding of reliable cancer-specific markers and molecular pathways can provide numerous practical techniques and direct the way toward the development of rational curative medicines to increase the lifespan of patients. Immunotherapy has significantly enhanced the treatment of metastatic and late-stage melanoma, resulting in an incredible increase in positive responses to therapy. Despite the increasing occurrence of melanoma, the median survival rate for patients with advanced, inoperable terminal disease has increased from around six months to almost six years. The current knowledge of the tumor microenvironment (TME) and its interaction with the immune system has resulted in the swift growth of innovative immunotherapy treatments. Exosomes are small extracellular vesicles (EVs), ranging from 30 to 150 nm in size, that the majority of cells released them. Exosomes possess natural advantages such as high compatibility with living organisms and low potential for causing immune reactions, making them practical for delivering therapeutic agents like chemotherapy drugs, nucleic acids, and proteins. This review highlights recent advancements in using exosomes as an approach to providing medications for the treatment of melanoma.

恶性黑色素瘤是人类最致命的癌症之一,几乎造成所有皮肤肿瘤晚期患者死亡。全面了解可靠的癌症特异性标志物和分子通路可提供大量实用技术,并为开发合理的治疗药物、延长患者寿命指明方向。免疫疗法大大提高了转移性黑色素瘤和晚期黑色素瘤的治疗效果,对治疗产生积极反应的患者人数也因此大幅增加。尽管黑色素瘤的发病率越来越高,但无法手术的晚期绝症患者的中位生存率已从六个月左右提高到近六年。目前对肿瘤微环境(TME)及其与免疫系统相互作用的了解促使创新性免疫疗法迅速发展。外泌体是一种小的细胞外囊泡 (EV),大小从 30 纳米到 150 纳米不等,大多数细胞都会释放它们。外泌体具有天然优势,如与生物体的兼容性高、引起免疫反应的可能性低,因此可用于递送化疗药物、核酸和蛋白质等治疗药物。本综述将重点介绍利用外泌体提供治疗黑色素瘤药物的最新进展。
{"title":"Exosome-based immunotherapy as an innovative therapeutic approach in melanoma.","authors":"Shabnam Babaei, Manouchehr Fadaee, Hajar Abbasi-Kenarsari, Dariush Shanehbandi, Tohid Kazemi","doi":"10.1186/s12964-024-01906-1","DOIUrl":"10.1186/s12964-024-01906-1","url":null,"abstract":"<p><p>The malignant form of melanoma is one of the deadliest human cancers that accounts for almost all of the skin tumor-related fatalities in its later stages. Achieving an exhaustive understanding of reliable cancer-specific markers and molecular pathways can provide numerous practical techniques and direct the way toward the development of rational curative medicines to increase the lifespan of patients. Immunotherapy has significantly enhanced the treatment of metastatic and late-stage melanoma, resulting in an incredible increase in positive responses to therapy. Despite the increasing occurrence of melanoma, the median survival rate for patients with advanced, inoperable terminal disease has increased from around six months to almost six years. The current knowledge of the tumor microenvironment (TME) and its interaction with the immune system has resulted in the swift growth of innovative immunotherapy treatments. Exosomes are small extracellular vesicles (EVs), ranging from 30 to 150 nm in size, that the majority of cells released them. Exosomes possess natural advantages such as high compatibility with living organisms and low potential for causing immune reactions, making them practical for delivering therapeutic agents like chemotherapy drugs, nucleic acids, and proteins. This review highlights recent advancements in using exosomes as an approach to providing medications for the treatment of melanoma.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"527"},"PeriodicalIF":8.2,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11526674/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142559519","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
Refractory testicular germ cell tumors are highly sensitive to the targeting of polycomb pathway demethylases KDM6A and KDM6B. 难治性睾丸生殖细胞肿瘤对靶向多聚酶途径去甲基化酶KDM6A和KDM6B高度敏感。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-31 DOI: 10.1186/s12964-024-01912-3
Doha Shokry, Mehwish W Khan, Christine Powell, Samantha Johnson, Brayden C Rennels, Raya I Boyd, Zhengyang Sun, Zeeshan Fazal, Sarah J Freemantle, Maryanna H Parker, Miranda D Vieson, Jonathan P Samuelson, Michael J Spinella, Ratnakar Singh

Testicular germ cell tumors (TGCTs) can be treated with cisplatin-based therapy. However, a clinically significant number of cisplatin-resistant patients die from progressive disease as no effective alternatives exist. Curative cisplatin therapy results in acute and life-long toxicities in the young TGCT patient population providing a rationale to decrease cisplatin exposure. In contrast to genetic alterations, recent evidence suggests that epigenetics is a major driving factor for TGCT formation, progression, and response to chemotherapy. Hence, targeting epigenetic pathways with "epidrugs" is one potential relatively unexplored strategy to advance TGCT treatment beyond cisplatin. In this report, we demonstrate for the first time that targeting polycomb demethylases KDM6A and KDM6B with epidrug GSK-J4 can treat both cisplatin-sensitive and -resistant TGCTs. While GSK-J4 had minimal effects alone on TGCT tumor growth in vivo, it dramatically sensitized cisplatin-sensitive and -resistant TGCTs to cisplatin. We validated KDM6A/KDM6B as the target of GSK-J4 since KDM6A/KDM6B genetic depletion had a similar effect to GSK-J4 on cisplatin-mediated anti-tumor activity and transcriptome alterations. Pharmacologic and genetic targeting of KDM6A/KDM6B potentiated or primed the p53-dominant transcriptional response to cisplatin, with also evidence for basal activation of p53. Further, several chromatin modifier genes, including BRD4, lysine demethylases, chromodomain helicase DNA binding proteins, and lysine methyltransferases, were repressed with cisplatin only in KDM6A/KDM6B-targeted cells, implying that KDM6A/KDM6B inhibition sets the stage for extensive chromatin remodeling of TGCT cells upon cisplatin treatment. Our findings demonstrate that targeting polycomb demethylases is a new potent pharmacologic strategy for treating cisplatin resistant TGCTs that warrants clinical development.

睾丸生殖细胞瘤(TGCT)可采用顺铂疗法治疗。然而,由于没有有效的替代疗法,临床上大量对顺铂耐药的患者死于疾病进展。治愈性顺铂疗法会对年轻的 TGCT 患者造成急性和终身毒性,这就为减少顺铂暴露提供了理由。与基因改变不同,最近的证据表明,表观遗传学是 TGCT 形成、发展和对化疗反应的主要驱动因素。因此,用 "表观药物 "靶向表观遗传途径是一种潜在的、相对尚未开发的策略,可推动顺铂以外的 TGCT 治疗。在本报告中,我们首次证明了用表观药物 GSK-J4 靶向多聚酶去甲基化酶 KDM6A 和 KDM6B 可以治疗顺铂敏感和耐药的 TGCT。虽然 GSK-J4 本身对 TGCT 肿瘤的体内生长影响甚微,但它却能使顺铂敏感和耐药的 TGCT 对顺铂显著增敏。我们验证了 KDM6A/KDM6B 是 GSK-J4 的靶点,因为 KDM6A/KDM6B 基因缺失对顺铂介导的抗肿瘤活性和转录组改变的影响与 GSK-J4 相似。药物和基因靶向 KDM6A/KDM6B 可增强或启动 p53 主导的顺铂转录反应,也有证据表明 p53 的基础激活。此外,一些染色质修饰基因,包括BRD4、赖氨酸去甲基化酶、染色体组螺旋酶DNA结合蛋白和赖氨酸甲基转移酶,只有在靶向KDM6A/KDM6B的细胞中才会受到顺铂的抑制,这意味着KDM6A/KDM6B的抑制为TGCT细胞在顺铂处理后进行广泛的染色质重塑创造了条件。我们的研究结果表明,靶向多聚酶去甲基化酶是治疗顺铂耐药 TGCT 的一种新的有效药物策略,值得进行临床开发。
{"title":"Refractory testicular germ cell tumors are highly sensitive to the targeting of polycomb pathway demethylases KDM6A and KDM6B.","authors":"Doha Shokry, Mehwish W Khan, Christine Powell, Samantha Johnson, Brayden C Rennels, Raya I Boyd, Zhengyang Sun, Zeeshan Fazal, Sarah J Freemantle, Maryanna H Parker, Miranda D Vieson, Jonathan P Samuelson, Michael J Spinella, Ratnakar Singh","doi":"10.1186/s12964-024-01912-3","DOIUrl":"10.1186/s12964-024-01912-3","url":null,"abstract":"<p><p>Testicular germ cell tumors (TGCTs) can be treated with cisplatin-based therapy. However, a clinically significant number of cisplatin-resistant patients die from progressive disease as no effective alternatives exist. Curative cisplatin therapy results in acute and life-long toxicities in the young TGCT patient population providing a rationale to decrease cisplatin exposure. In contrast to genetic alterations, recent evidence suggests that epigenetics is a major driving factor for TGCT formation, progression, and response to chemotherapy. Hence, targeting epigenetic pathways with \"epidrugs\" is one potential relatively unexplored strategy to advance TGCT treatment beyond cisplatin. In this report, we demonstrate for the first time that targeting polycomb demethylases KDM6A and KDM6B with epidrug GSK-J4 can treat both cisplatin-sensitive and -resistant TGCTs. While GSK-J4 had minimal effects alone on TGCT tumor growth in vivo, it dramatically sensitized cisplatin-sensitive and -resistant TGCTs to cisplatin. We validated KDM6A/KDM6B as the target of GSK-J4 since KDM6A/KDM6B genetic depletion had a similar effect to GSK-J4 on cisplatin-mediated anti-tumor activity and transcriptome alterations. Pharmacologic and genetic targeting of KDM6A/KDM6B potentiated or primed the p53-dominant transcriptional response to cisplatin, with also evidence for basal activation of p53. Further, several chromatin modifier genes, including BRD4, lysine demethylases, chromodomain helicase DNA binding proteins, and lysine methyltransferases, were repressed with cisplatin only in KDM6A/KDM6B-targeted cells, implying that KDM6A/KDM6B inhibition sets the stage for extensive chromatin remodeling of TGCT cells upon cisplatin treatment. Our findings demonstrate that targeting polycomb demethylases is a new potent pharmacologic strategy for treating cisplatin resistant TGCTs that warrants clinical development.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"528"},"PeriodicalIF":8.2,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11529429/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142559520","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
A non-catalytic role of IPMK is required for PLCγ1 activation in T cell receptor signaling by stabilizing the PLCγ1-Sam68 complex. 通过稳定 PLCγ1-Sam68 复合物,IPMK 在 T 细胞受体信号传导过程中的非催化作用是 PLCγ1 激活所必需的。
IF 8.2 2区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-10-30 DOI: 10.1186/s12964-024-01907-0
Sehoon Hong, Kyurae Kim, Young-Ri Shim, Jiyeon Park, Sung Eun Choi, Hyungyu Min, Seulgi Lee, Ji-Joon Song, Suk-Jo Kang, Won-Il Jeong, Rho Hyun Seong, Seyun Kim

Background: Phospholipase C gamma 1 (PLCγ1) is an important mediator of the T cell receptor (TCR) and growth factor signaling. PLCγ1 is activated by Src family kinases (SFKs) and produces inositol 1,4,5-triphosphate (InsP3) from phosphatidylinositol 4,5-bisphosphate (PIP2). Inositol polyphosphate multikinase (IPMK) is a pleiotropic enzyme with broad substrate specificity and non-catalytic activities that mediate various functional protein-protein interactions. Therefore, IPMK plays critical functions in key biological events such as cell growth. However, the contribution of IPMK to the activation of PLCγ1 in TCR signaling remains mostly unelucidated. The current study aimed to elucidate the functions of IPMK in TCR signaling and to uncover the mode of IPMK-mediated signaling action in PLCγ1 activation.

Methods: Concanavalin A (ConA)-induced acute hepatitis model was established in CD4+ T cell-specific IPMK knockout mice (IPMKΔCD4). Histological analysis was performed to assess hepatic injury. Primary cultures of naïve CD4+ T cells were used to uncover the role of mechanisms of IPMK in vitro. Western blot analysis, quantitative real-time PCR, and flow cytometry were performed to analyze the TCR-stimulation-induced PLCγ1 activation and the downstream signaling pathway in naïve CD4+ T cells. Yeast two-hybrid screening and co-immunoprecipitation were conducted to identify the IPMK-binding proteins and protein complexes.

Results: IPMKΔCD4 mice showed alleviated ConA-induced acute hepatitis. CD4+ helper T cells in these mice showed reduced PLCγ1 Y783 phosphorylation, which subsequently dampens calcium signaling and IL-2 production. IPMK was found to contribute to PLCγ1 activation via the direct binding of IPMK to Src-associated substrate during mitosis of 68 kDa (Sam68). Mechanistically, IPMK stabilizes the interaction between Sam68 and to PLCγ1, thereby promoting PLCγ1 phosphorylation. Interfering this IPMK-Sam68 binding interaction with IPMK dominant-negative peptides impaired PLCγ1 phosphorylation.

Conclusions: Our results demonstrate that IPMK non-catalytically promotes PLCγ1 phosphorylation by stabilizing the PLCγ1-Sam68 complex. Targeting IPMK in CD4+ T cells may be a promising strategy for managing immune diseases caused by excessive stimulation of TCR.

背景:磷脂酶Cγ1(PLCγ1)是T细胞受体(TCR)和生长因子信号转导的重要介质。PLCγ1由Src家族激酶(SFKs)激活,并从磷脂酰肌醇4,5-二磷酸(PIP2)中产生肌醇1,4,5-三磷酸酯(InsP3)。多磷酸肌醇多激酶(IPMK)是一种多效酶,具有广泛的底物特异性和非催化活性,可介导各种功能性蛋白质-蛋白质相互作用。因此,IPMK 在细胞生长等关键生物事件中发挥着重要功能。然而,IPMK 在 TCR 信号转导中对 PLCγ1 激活的贡献大部分仍未被阐明。本研究旨在阐明 IPMK 在 TCR 信号转导中的功能,并揭示 IPMK 在 PLCγ1 激活过程中介导信号作用的模式:方法:在CD4+ T细胞特异性IPMK基因敲除小鼠(IPMKΔCD4)中建立加奈伐林A(ConA)诱导的急性肝炎模型。进行组织学分析以评估肝损伤。利用幼稚 CD4+ T 细胞的原代培养物来揭示 IPMK 在体外的作用机制。通过 Western 印迹分析、定量实时 PCR 和流式细胞术分析 TCR 刺激诱导的 PLCγ1 活化和下游信号通路在幼稚 CD4+ T 细胞中的作用。通过酵母双杂交筛选和共免疫沉淀鉴定了IPMK结合蛋白和蛋白复合物:结果:IPMKΔCD4小鼠对ConA诱导的急性肝炎有缓解作用。这些小鼠的 CD4+ 辅助 T 细胞显示 PLCγ1 Y783 磷酸化减少,从而抑制了钙信号传导和 IL-2 的产生。研究发现,IPMK 在有丝分裂过程中与 68 kDa 的 Src 相关底物(Sam68)直接结合,从而促进了 PLCγ1 的激活。从机制上讲,IPMK 可稳定 Sam68 与 PLCγ1 之间的相互作用,从而促进 PLCγ1 磷酸化。用IPMK显性阴性肽干扰IPMK与Sam68的这种结合相互作用,会影响PLCγ1的磷酸化:我们的研究结果表明,IPMK通过稳定PLCγ1-Sam68复合物,非催化地促进了PLCγ1磷酸化。以 CD4+ T 细胞中的 IPMK 为靶点可能是治疗 TCR 过度刺激引起的免疫疾病的一种有前途的策略。
{"title":"A non-catalytic role of IPMK is required for PLCγ1 activation in T cell receptor signaling by stabilizing the PLCγ1-Sam68 complex.","authors":"Sehoon Hong, Kyurae Kim, Young-Ri Shim, Jiyeon Park, Sung Eun Choi, Hyungyu Min, Seulgi Lee, Ji-Joon Song, Suk-Jo Kang, Won-Il Jeong, Rho Hyun Seong, Seyun Kim","doi":"10.1186/s12964-024-01907-0","DOIUrl":"10.1186/s12964-024-01907-0","url":null,"abstract":"<p><strong>Background: </strong>Phospholipase C gamma 1 (PLCγ1) is an important mediator of the T cell receptor (TCR) and growth factor signaling. PLCγ1 is activated by Src family kinases (SFKs) and produces inositol 1,4,5-triphosphate (InsP<sub>3</sub>) from phosphatidylinositol 4,5-bisphosphate (PIP<sub>2</sub>). Inositol polyphosphate multikinase (IPMK) is a pleiotropic enzyme with broad substrate specificity and non-catalytic activities that mediate various functional protein-protein interactions. Therefore, IPMK plays critical functions in key biological events such as cell growth. However, the contribution of IPMK to the activation of PLCγ1 in TCR signaling remains mostly unelucidated. The current study aimed to elucidate the functions of IPMK in TCR signaling and to uncover the mode of IPMK-mediated signaling action in PLCγ1 activation.</p><p><strong>Methods: </strong>Concanavalin A (ConA)-induced acute hepatitis model was established in CD4<sup>+</sup> T cell-specific IPMK knockout mice (IPMK<sup>ΔCD4</sup>). Histological analysis was performed to assess hepatic injury. Primary cultures of naïve CD4<sup>+</sup> T cells were used to uncover the role of mechanisms of IPMK in vitro. Western blot analysis, quantitative real-time PCR, and flow cytometry were performed to analyze the TCR-stimulation-induced PLCγ1 activation and the downstream signaling pathway in naïve CD4<sup>+</sup> T cells. Yeast two-hybrid screening and co-immunoprecipitation were conducted to identify the IPMK-binding proteins and protein complexes.</p><p><strong>Results: </strong>IPMK<sup>ΔCD4</sup> mice showed alleviated ConA-induced acute hepatitis. CD4<sup>+</sup> helper T cells in these mice showed reduced PLCγ1 Y783 phosphorylation, which subsequently dampens calcium signaling and IL-2 production. IPMK was found to contribute to PLCγ1 activation via the direct binding of IPMK to Src-associated substrate during mitosis of 68 kDa (Sam68). Mechanistically, IPMK stabilizes the interaction between Sam68 and to PLCγ1, thereby promoting PLCγ1 phosphorylation. Interfering this IPMK-Sam68 binding interaction with IPMK dominant-negative peptides impaired PLCγ1 phosphorylation.</p><p><strong>Conclusions: </strong>Our results demonstrate that IPMK non-catalytically promotes PLCγ1 phosphorylation by stabilizing the PLCγ1-Sam68 complex. Targeting IPMK in CD4<sup>+</sup> T cells may be a promising strategy for managing immune diseases caused by excessive stimulation of TCR.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":"22 1","pages":"526"},"PeriodicalIF":8.2,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11524019/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549043","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
期刊
Cell Communication and Signaling
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