Objective: Identify transcription factors and target genes associated with prostate cancer, offering new therapy approaches.
Methods: Gene Set Enrichment Analysis (GSEA) investigates early 2 factor (E2F) transcription factor family roles in prostate cancer using the TCGA database. Survival analysis examined E2F factors and patient survival connections. Dataset analysis identified E2F1-involved key genes. Quantitative Real-time PCR (qPCR), which combines ultrasound-guided methods to collect clinical samples from prostate cancer patients, was utilized to determine the expression levels of E2F1 and its target genes in patient samples and cancer cells. The effect of E2F1 and its target gene expression alterations on prostate cell proliferation was examined utilizing the cell counting kit-8 (CCK8) technique. Double fluorescence enzyme experiment verified E2F1-target gene connections.
Results: E2F family genes induce prostate cancer and show correlated co-expression. E2F1, E2F2, E2F3, E2F5, and E2F7 were considerably over-expressed in prostate cancer tissues. While E2F4 and E2F6 were notably underexpressed, there was no statistically important change in the E2F8 expression between prostate cancer and surrounding tissues. High expression of E2F genes is associated with lower patient survival. The transmemrane protein 132 (TMEM132A) was identified as a key gene for E2F1 action and is associated with poor prognosis in patients. The essential gene for E2F1 function, TMEM132A, was discovered. According to the qPCR results, E2F1 and TMEM132A are considerably expressed in cancer cells and patient samples. Interfering with its expression significantly inhibited the proliferation ability of cancer cells. The double luciferase experiment showed that E2F1 regulates the expression level in phase by binding directly to the TMEM132A promoter.
Conclusions: The E2F transcription factor family induces prostate cancer and correlates with poor prognosis. E2F1 directly regulates TMEM132A by binding its promoter and controlling the degree of protein expression, thereby affecting cancer cell growth.
{"title":"Study of the Role of E2F1 and TMEM132A in Prostate Cancer Development.","authors":"Ying Wang, Haifeng Hu, Huilin Liu, Dandan Zhou, Yinghui Zhang, Lu Li, Chunxin Huang","doi":"10.31083/j.fbl2910360","DOIUrl":"https://doi.org/10.31083/j.fbl2910360","url":null,"abstract":"<p><strong>Objective: </strong>Identify transcription factors and target genes associated with prostate cancer, offering new therapy approaches.</p><p><strong>Methods: </strong>Gene Set Enrichment Analysis (GSEA) investigates early 2 factor (E2F) transcription factor family roles in prostate cancer using the TCGA database. Survival analysis examined E2F factors and patient survival connections. Dataset analysis identified E2F1-involved key genes. Quantitative Real-time PCR (qPCR), which combines ultrasound-guided methods to collect clinical samples from prostate cancer patients, was utilized to determine the expression levels of <i>E2F1</i> and its target genes in patient samples and cancer cells. The effect of <i>E2F1</i> and its target gene expression alterations on prostate cell proliferation was examined utilizing the cell counting kit-8 (CCK8) technique. Double fluorescence enzyme experiment verified E2F1-target gene connections.</p><p><strong>Results: </strong>E2F family genes induce prostate cancer and show correlated co-expression. <i>E2F1</i>, <i>E2F2</i>, <i>E2F3</i>, <i>E2F5</i>, and <i>E2F7</i> were considerably over-expressed in prostate cancer tissues. While <i>E2F4</i> and <i>E2F6</i> were notably underexpressed, there was no statistically important change in the <i>E2F8</i> expression between prostate cancer and surrounding tissues. High expression of <i>E2F</i> genes is associated with lower patient survival. The transmemrane protein 132 (<i>TMEM132A</i>) was identified as a key gene for <i>E2F1</i> action and is associated with poor prognosis in patients. The essential gene for <i>E2F1</i> function, <i>TMEM132A</i>, was discovered. According to the qPCR results, <i>E2F1</i> and <i>TMEM132A</i> are considerably expressed in cancer cells and patient samples. Interfering with its expression significantly inhibited the proliferation ability of cancer cells. The double luciferase experiment showed that <i>E2F1</i> regulates the expression level in phase by binding directly to the <i>TMEM132A</i> promoter.</p><p><strong>Conclusions: </strong>The E2F transcription factor family induces prostate cancer and correlates with poor prognosis. <i>E2F1</i> directly regulates <i>TMEM132A</i> by binding its promoter and controlling the degree of protein expression, thereby affecting cancer cell growth.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"29 10","pages":"360"},"PeriodicalIF":3.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Anna V Tugarova, Polina V Mamchenkova, Anastasiya A Vladimirova, Lilia P Petrova, Andrei V Shelud'ko, Alexander A Kamnev
Background: Many bacteria are capable of reducing selenium oxyanions, primarily selenite (SeO32-), in most cases forming selenium(0) nanostructures. The mechanisms of these transformations may vary for different bacterial species and have so far not yet been clarified in detail. Bacteria of the genus Azospirillum, including ubiquitous phytostimulating rhizobacteria, are widely studied and have potential for agricultural biotechnology and bioremediation of excessively seleniferous soils, as they are able to reduce selenite ions.
Methods: Cultures of A.brasilense Sp7 and its derivatives (mutant strains) were grown on the modified liquid malate salt medium in the presence or absence of selenite. The following methods were used: spectrophotometric monitoring of bacterial growth; inhibition of glutathione (GSH) synthesis in bacteria by L-buthionine-sulfoximine (BSO); optical selenite and nitrite reduction assays; transmission electron microscopy of cells grown with and without BSO and/or selenite.
Results: In a set of separate comparative studies of nitrite and selenite reduction by the wild-type strain A.brasilense Sp7 and its three specially selected derivatives (mutant strains) with different rates of nitrite reduction, a direct correlation was found between their nitrite and selenite reduction rates for all the strains used in the study. Moreover, for BSO it has been shown that its presence does not block selenite reduction in A.brasilense Sp7.
Conclusions: Evidence has been presented for the first time for bacteria of the genus Azospirillum that the denitrification pathway known to be inherent in these bacteria, including nitrite reductase, is likely to be involved in selenite reduction. The results using BSO also imply that detoxification of selenite through the GSH redox system (which is commonly considered as the primary mechanism of selenite reduction in many bacteria) does not play a significant role in A.brasilense. The acquired knowledge on the mechanisms underlying biogenic transformations of inorganic selenium in A.brasilense is a step forward both in understanding the biogeochemical selenium cycle and to a variety of potential nano- and biotechnological applications.
{"title":"Role of Denitrification in Selenite Reduction by <i>Azospirillum brasilense</i> with the Formation of Selenium Nanoparticles.","authors":"Anna V Tugarova, Polina V Mamchenkova, Anastasiya A Vladimirova, Lilia P Petrova, Andrei V Shelud'ko, Alexander A Kamnev","doi":"10.31083/j.fbl2910361","DOIUrl":"https://doi.org/10.31083/j.fbl2910361","url":null,"abstract":"<p><strong>Background: </strong>Many bacteria are capable of reducing selenium oxyanions, primarily selenite (SeO<sub>3</sub><sup>2-</sup>), in most cases forming selenium(0) nanostructures. The mechanisms of these transformations may vary for different bacterial species and have so far not yet been clarified in detail. Bacteria of the genus <i>Azospirillum</i>, including ubiquitous phytostimulating rhizobacteria, are widely studied and have potential for agricultural biotechnology and bioremediation of excessively seleniferous soils, as they are able to reduce selenite ions.</p><p><strong>Methods: </strong>Cultures of <i>A.brasilense</i> Sp7 and its derivatives (mutant strains) were grown on the modified liquid malate salt medium in the presence or absence of selenite. The following methods were used: spectrophotometric monitoring of bacterial growth; inhibition of glutathione (GSH) synthesis in bacteria by L-buthionine-sulfoximine (BSO); optical selenite and nitrite reduction assays; transmission electron microscopy of cells grown with and without BSO and/or selenite.</p><p><strong>Results: </strong>In a set of separate comparative studies of nitrite and selenite reduction by the wild-type strain <i>A.brasilense</i> Sp7 and its three specially selected derivatives (mutant strains) with different rates of nitrite reduction, a direct correlation was found between their nitrite and selenite reduction rates for all the strains used in the study. Moreover, for BSO it has been shown that its presence does not block selenite reduction in <i>A.brasilense</i> Sp7.</p><p><strong>Conclusions: </strong>Evidence has been presented for the first time for bacteria of the genus <i>Azospirillum</i> that the denitrification pathway known to be inherent in these bacteria, including nitrite reductase, is likely to be involved in selenite reduction. The results using BSO also imply that detoxification of selenite through the GSH redox system (which is commonly considered as the primary mechanism of selenite reduction in many bacteria) does not play a significant role in <i>A.brasilense</i>. The acquired knowledge on the mechanisms underlying biogenic transformations of inorganic selenium in <i>A.brasilense</i> is a step forward both in understanding the biogeochemical selenium cycle and to a variety of potential nano- and biotechnological applications.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"29 10","pages":"361"},"PeriodicalIF":3.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549251","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Chemokines bind to specific chemokine receptors, known as cell surface G protein-coupled receptors, constructing chemokine axes which lead to cell migration and invasion in developmental stage, pathophysiological process, and immune reactions. The chemokine axes in the tumor microenvironment are involved in tumor growth, angiogenesis, cancer stem-like cell properties, metastasis, and chemoresistance, modifying tumor immune contexture and cancer progression. Clinical features, including tumor state, grade, lymph node metastasis, and cancer subtypes, are related to the specific chemokine axes, which play a significant role in immune contexture and cell to cell interaction in the tumor microenvironment, followed by altered cancer prognosis and overall survival. The present review summarizes the role of chemokine axes in breast cancer, based on data obtained from cell line and animal models and human tumor samples. This review provides information that understand the important roles of each chemokine axis in breast cancer, probably offering a clue of adjuvant therapeutic options to improve the quality of life and survival for patients with breast cancer.
趋化因子与特定的趋化因子受体(即细胞表面 G 蛋白偶联受体)结合,构建出趋化因子轴,在发育阶段、病理生理过程和免疫反应中导致细胞迁移和侵袭。肿瘤微环境中的趋化因子轴参与肿瘤生长、血管生成、癌症干样细胞特性、转移和化疗抵抗,改变肿瘤免疫环境和癌症进展。临床特征,包括肿瘤状态、分级、淋巴结转移和癌症亚型,都与特定的趋化因子轴有关,这些趋化因子轴在肿瘤微环境的免疫环境和细胞间相互作用中发挥着重要作用,继而改变癌症预后和总生存期。本综述基于从细胞系、动物模型和人类肿瘤样本中获得的数据,总结了趋化因子轴在乳腺癌中的作用。本综述提供的信息有助于了解各趋化因子轴在乳腺癌中的重要作用,从而为改善乳腺癌患者的生活质量和生存率提供辅助治疗方案的线索。
{"title":"Roles of Chemokine Axes in Breast Cancer.","authors":"Deok-Soo Son, Samuel E Adunyah","doi":"10.31083/j.fbl2910358","DOIUrl":"https://doi.org/10.31083/j.fbl2910358","url":null,"abstract":"<p><p>Chemokines bind to specific chemokine receptors, known as cell surface G protein-coupled receptors, constructing chemokine axes which lead to cell migration and invasion in developmental stage, pathophysiological process, and immune reactions. The chemokine axes in the tumor microenvironment are involved in tumor growth, angiogenesis, cancer stem-like cell properties, metastasis, and chemoresistance, modifying tumor immune contexture and cancer progression. Clinical features, including tumor state, grade, lymph node metastasis, and cancer subtypes, are related to the specific chemokine axes, which play a significant role in immune contexture and cell to cell interaction in the tumor microenvironment, followed by altered cancer prognosis and overall survival. The present review summarizes the role of chemokine axes in breast cancer, based on data obtained from cell line and animal models and human tumor samples. This review provides information that understand the important roles of each chemokine axis in breast cancer, probably offering a clue of adjuvant therapeutic options to improve the quality of life and survival for patients with breast cancer.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"29 10","pages":"358"},"PeriodicalIF":3.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549252","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Ubiquitination plays a key role in various cancers, and F-box and WD repeat domain containing 7 (FBW7) is a tumor suppressor that targets several cancer-causing proteins for ubiquitination. This paper set out to pinpoint the role of FBW7 in hepatocellular carcinoma (HCC).
Methods: The target proteins of FBW7 and the expression of hromodomain helicase DNA binding protein 3 (CHD3) were analyzed in liver HCC (LIHC) samples using the BioSignal Data website. The effects of CHD3 and FBW7 on HCC cell viability, migration, invasion and stemness were investigated through cell counting kit (CCK)-8, wound healing, transwell and sphere formation assays. Detection on CHD3 and FBW7 expressions as well as their relationship was performed employing quantitative reverse transcription-polymerase chain reaction (qRT-PCR), immunoprecipitation, ubiquitination and western blot analyses.
Results: The prediction of Ubibrowser revealed CHD3 as a target protein of FBW7. The data of starBase exhibited a higher expression level of CHD3 in LIHC samples relative to normal samples. CHD3 was upregulated in HCC cells. CHD3 knockdown inhibited HCC cell proliferation, migration, invasion, stemness and oxaliplatin sensitivity. FBW7 targeted CHD3 for ubiquitination. FBW7 overexpression restrained HCC cell migration, invasion and stemness, and attenuated the effects of overexpressed CHD3 on promoting migration, invasion, stemness and oxaliplatin resistance in HCC cells.
Conclusion: FBW7 overexpression suppresses HCC cell metastasis, stemness and oxaliplatin resistance via targeting CHD3 for ubiquitylation and degradation.
{"title":"FBW7-Mediated Degradation of CHD3 Suppresses Hepatocellular Carcinoma Metastasis and Stemness to Enhance Oxaliplatin Sensitivity.","authors":"Shijie Li, Tingting Fan, Changjun Wu","doi":"10.31083/j.fbl2910357","DOIUrl":"https://doi.org/10.31083/j.fbl2910357","url":null,"abstract":"<p><strong>Background: </strong>Ubiquitination plays a key role in various cancers, and F-box and WD repeat domain containing 7 (FBW7) is a tumor suppressor that targets several cancer-causing proteins for ubiquitination. This paper set out to pinpoint the role of FBW7 in hepatocellular carcinoma (HCC).</p><p><strong>Methods: </strong>The target proteins of FBW7 and the expression of hromodomain helicase DNA binding protein 3 (<i>CHD3</i>) were analyzed in liver HCC (LIHC) samples using the BioSignal Data website. The effects of CHD3 and FBW7 on HCC cell viability, migration, invasion and stemness were investigated through cell counting kit (CCK)-8, wound healing, transwell and sphere formation assays. Detection on CHD3 and FBW7 expressions as well as their relationship was performed employing quantitative reverse transcription-polymerase chain reaction (qRT-PCR), immunoprecipitation, ubiquitination and western blot analyses.</p><p><strong>Results: </strong>The prediction of Ubibrowser revealed CHD3 as a target protein of FBW7. The data of starBase exhibited a higher expression level of <i>CHD3</i> in LIHC samples relative to normal samples. <i>CHD3</i> was upregulated in HCC cells. <i>CHD3</i> knockdown inhibited HCC cell proliferation, migration, invasion, stemness and oxaliplatin sensitivity. FBW7 targeted CHD3 for ubiquitination. <i>FBW7</i> overexpression restrained HCC cell migration, invasion and stemness, and attenuated the effects of overexpressed <i>CHD3</i> on promoting migration, invasion, stemness and oxaliplatin resistance in HCC cells.</p><p><strong>Conclusion: </strong>FBW7 overexpression suppresses HCC cell metastasis, stemness and oxaliplatin resistance via targeting CHD3 for ubiquitylation and degradation.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"29 10","pages":"357"},"PeriodicalIF":3.3,"publicationDate":"2024-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549327","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yongxia Li, Gang Shen, Jiarui Du, Wei Dai, Zuopeng Su
Background: As antioxidant and anti-inflammatory agents, carbonic anhydrase inhibitors can exert potentially useful therapeutic effects following central nervous system trauma, including intracerebral hemorrhage (ICH). However, the therapeutic efficacy of ethoxyzolamide (ETZ) as a novel carbonic anhydrase inhibitor for ICH has not yet been determined.
Methods: An autologous blood injection method was used to establish ICH models, which were then used to establish the effects of intraperitoneal injection of ETZ on ICH. Neuronal damage, apoptotic protein expression, oxidative and inflammatory factor content, microglia marker Iba-1 positivity, hepatic and renal pathological changes, and serum concentrations of hepatic and renal function indices were assessed by Nissl staining, western blotting, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, hematoxylin and eosin (HE) staining, and automatic biochemical analysis in brain tissues.
Results: The ICH group showed massive hemorrhagic foci; significant increases in brain water content, modified mouse neurological deficit scoring (mNSS) score, pro-apoptotic protein expression, oxidative factors, pro-inflammatory factors, and Iba-1 positivity; and significant reductions in Nissl body size, anti-apoptotic protein expression, and antioxidant factors, all of which were reversed by ETZ in a dose-dependent manner. ETZ has a good biosafety profile with no significant burden on the human liver or kidneys. The Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway was mildly activated in ICH mice, and was further increased after ETZ injection. Molecular docking experiments revealed that ETZ could dock onto the Nrf2-binding domain of keap1.
Conclusions: ETZ, as a novel carbonic anhydrase inhibitor, further activated the Keap1/Nrf2 pathway by docking with the Nrf2-binding domain of keap1, thereby exerting antioxidant, anti-inflammatory, anti-apoptotic, and cerebral neuroprotective effects in ICH mice.
背景:作为抗氧化剂和抗炎药,碳酸酐酶抑制剂可在中枢神经系统创伤(包括脑内出血)后发挥潜在的治疗作用。然而,乙氧唑胺(ETZ)作为一种新型碳酸酐酶抑制剂对 ICH 的疗效尚未确定:方法:采用自体血液注射法建立 ICH 模型,然后利用该模型确定腹腔注射 ETZ 对 ICH 的影响。方法:采用自体血注射法建立 ICH 模型,然后确定腹腔注射 ETZ 对 ICH 的影响,并通过 Nissl 染色、Western 印迹、酶联免疫吸附试验(ELISA)、免疫组化、苏木精和伊红(HE)染色以及脑组织的自动生化分析评估神经元损伤、凋亡蛋白表达、氧化因子和炎症因子含量、小胶质细胞标志物 Iba-1 阳性、肝脏和肾脏病理变化以及肝脏和肾脏功能指数的血清浓度:结果:ICH组出现大量出血灶;脑含水量、改良小鼠神经功能缺损评分(mNSS)、促凋亡蛋白表达、氧化因子、促炎因子和Iba-1阳性率显著增加;Nissl体大小、抗凋亡蛋白表达和抗氧化因子显著减少,所有这些均被ETZ以剂量依赖性方式逆转。ETZ 具有良好的生物安全性,对人体肝脏或肾脏无明显负担。ICH 小鼠的 Kelch-like ECH-associated protein 1 (Keap1) /nuclear factor erythroid 2-related factor 2 (Nrf2) 通路被轻度激活,注射 ETZ 后通路被进一步激活。分子对接实验显示,ETZ可与keap1的Nrf2结合结构域对接:结论:ETZ作为一种新型碳酸酐酶抑制剂,通过与keap1的Nrf2结合结构域对接,进一步激活了Keap1/Nrf2通路,从而在ICH小鼠体内发挥了抗氧化、抗炎、抗凋亡和脑神经保护作用。
{"title":"Neuroprotective Potential of Ethoxzolamide Targeting Oxidative Stress and Inflammation in Experimental Models of Intracerebral Hemorrhage.","authors":"Yongxia Li, Gang Shen, Jiarui Du, Wei Dai, Zuopeng Su","doi":"10.31083/j.fbl2910356","DOIUrl":"https://doi.org/10.31083/j.fbl2910356","url":null,"abstract":"<p><strong>Background: </strong>As antioxidant and anti-inflammatory agents, carbonic anhydrase inhibitors can exert potentially useful therapeutic effects following central nervous system trauma, including intracerebral hemorrhage (ICH). However, the therapeutic efficacy of ethoxyzolamide (ETZ) as a novel carbonic anhydrase inhibitor for ICH has not yet been determined.</p><p><strong>Methods: </strong>An autologous blood injection method was used to establish ICH models, which were then used to establish the effects of intraperitoneal injection of ETZ on ICH. Neuronal damage, apoptotic protein expression, oxidative and inflammatory factor content, microglia marker Iba-1 positivity, hepatic and renal pathological changes, and serum concentrations of hepatic and renal function indices were assessed by Nissl staining, western blotting, enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, hematoxylin and eosin (HE) staining, and automatic biochemical analysis in brain tissues.</p><p><strong>Results: </strong>The ICH group showed massive hemorrhagic foci; significant increases in brain water content, modified mouse neurological deficit scoring (mNSS) score, pro-apoptotic protein expression, oxidative factors, pro-inflammatory factors, and Iba-1 positivity; and significant reductions in Nissl body size, anti-apoptotic protein expression, and antioxidant factors, all of which were reversed by ETZ in a dose-dependent manner. ETZ has a good biosafety profile with no significant burden on the human liver or kidneys. The Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway was mildly activated in ICH mice, and was further increased after ETZ injection. Molecular docking experiments revealed that ETZ could dock onto the Nrf2-binding domain of keap1.</p><p><strong>Conclusions: </strong>ETZ, as a novel carbonic anhydrase inhibitor, further activated the Keap1/Nrf2 pathway by docking with the Nrf2-binding domain of keap1, thereby exerting antioxidant, anti-inflammatory, anti-apoptotic, and cerebral neuroprotective effects in ICH mice.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"29 10","pages":"356"},"PeriodicalIF":3.3,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549336","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Reply to Comment on Ou Y, <i>et al</i>. \"UBA2 as a Prognostic Biomarker and Potential Therapeutic Target in Glioma\". Frontiers in Bioscience-Landmark. 2024; 29: 144.","authors":"Yuhong Ou, Hongtao Luo, Qiuning Zhang, Tianqi Du, Ruifeng Liu, Dandan Wang, Junru Chen, Meng Dong, Yuhang Wang, Zhen Yang, Xiaohu Wang","doi":"10.31083/j.fbl2910354","DOIUrl":"https://doi.org/10.31083/j.fbl2910354","url":null,"abstract":"","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"29 10","pages":"354"},"PeriodicalIF":3.3,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549249","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
The sympathetic nervous system (SNS) consists largely of two different types of components: neurons that release the neurotransmitter norepinephrine (NE, noradrenaline) to modulate homeostasis of the innevrvated effector organ or tissue and adrenal chromaffin cells, which synthesize and secrete the hormone epinephrine (Epi, adrenaline) and some NE into the blood circulation to act at distant organs and tissues that are not directly innervated by the SNS. Like almost every physiological process in the human body, G protein-coupled receptors (GPCRs) tightly modulate both NE release from sympathetic neuronal terminals and catecholamine (CA) secretion from the adrenal medulla. Regulator of G protein Signaling (RGS) proteins, acting as guanosine triphosphatase (GTPase)-activating proteins (GAPs) for the Gα subunits of heterotrimeric guanine nucleotide-binding proteins (G proteins), play a central role in silencing G protein signaling from a plethora of GPCRs. Certain RGS proteins and, in particular, RGS4, have been implicated in regulation of SNS activity and of adrenal chromaffin cell CA secretion. More specifically, recent studies have implicated RGS4 in regulation of NE release from cardiac sympathetic neurons by means of terminating free fatty acid receptor (FFAR)-3 calcium signaling and in regulation of NE and Epi secretion from the adrenal medulla by means of terminating cholinergic calcium signaling in adrenal chromaffin cells. Thus, in this review, we provide an overview of the current literature on the involvement of RGS proteins, with a particular focus on RGS4, in these two processes, i.e., NE release from sympathetic nerve terminals & CA secretion from adrenal chromaffin cells. We also highlight the therapeutic potential of RGS4 pharmacological manipulation for diseases characterized by sympathetic dysfunction or SNS hyperactivity, such as heart failure and hypertension.
交感神经系统(SNS)主要由两种不同类型的成分组成:神经元释放神经递质去甲肾上腺素(NE,noradrenaline),以调节受其支配的效应器官或组织的稳态;肾上腺绒毛膜细胞合成并分泌激素肾上腺素(Epi,adrenaline),部分 NE 进入血液循环,以作用于不受 SNS 直接支配的远处器官和组织。与人体内几乎所有的生理过程一样,G 蛋白偶联受体(GPCR)对交感神经终端的 NE 释放和肾上腺髓质的儿茶酚胺(CA)分泌都有严格的调节作用。G 蛋白信号调节器(RGS)蛋白作为异三聚鸟嘌呤核苷酸结合蛋白(G 蛋白)Gα 亚基的鸟苷三磷酸酶(GTPase)-激活蛋白(GAP),在抑制来自大量 GPCR 的 G 蛋白信号方面发挥着核心作用。某些 RGS 蛋白,特别是 RGS4,与 SNS 活动和肾上腺绒毛细胞 CA 分泌的调节有关。更具体地说,最近的研究表明,RGS4 通过终止游离脂肪酸受体(FFAR)-3 的钙信号传导来调节心脏交感神经元的 NE 释放,并通过终止肾上腺绒毛细胞中胆碱能钙信号传导来调节肾上腺髓质的 NE 和 Epi 分泌。因此,在这篇综述中,我们概述了目前有关 RGS 蛋白参与这两个过程(即交感神经末梢释放 NE 和肾上腺绒毛细胞分泌 CA)的文献,尤其关注 RGS4。我们还强调了对以交感神经功能障碍或SNS亢进为特征的疾病(如心力衰竭和高血压)进行RGS4药物治疗的潜力。
{"title":"RGS Proteins in Sympathetic Nervous System Regulation: Focus on Adrenal RGS4.","authors":"Anastasios Lymperopoulos, Renee A Stoicovy","doi":"10.31083/j.fbl2910355","DOIUrl":"https://doi.org/10.31083/j.fbl2910355","url":null,"abstract":"<p><p>The sympathetic nervous system (SNS) consists largely of two different types of components: neurons that release the neurotransmitter norepinephrine (NE, noradrenaline) to modulate homeostasis of the innevrvated effector organ or tissue and adrenal chromaffin cells, which synthesize and secrete the hormone epinephrine (Epi, adrenaline) and some NE into the blood circulation to act at distant organs and tissues that are not directly innervated by the SNS. Like almost every physiological process in the human body, G protein-coupled receptors (GPCRs) tightly modulate both NE release from sympathetic neuronal terminals and catecholamine (CA) secretion from the adrenal medulla. Regulator of G protein Signaling (RGS) proteins, acting as guanosine triphosphatase (GTPase)-activating proteins (GAPs) for the Gα subunits of heterotrimeric guanine nucleotide-binding proteins (G proteins), play a central role in silencing G protein signaling from a plethora of GPCRs. Certain RGS proteins and, in particular, RGS4, have been implicated in regulation of SNS activity and of adrenal chromaffin cell CA secretion. More specifically, recent studies have implicated RGS4 in regulation of NE release from cardiac sympathetic neurons by means of terminating free fatty acid receptor (FFAR)-3 calcium signaling and in regulation of NE and Epi secretion from the adrenal medulla by means of terminating cholinergic calcium signaling in adrenal chromaffin cells. Thus, in this review, we provide an overview of the current literature on the involvement of RGS proteins, with a particular focus on RGS4, in these two processes, i.e., NE release from sympathetic nerve terminals & CA secretion from adrenal chromaffin cells. We also highlight the therapeutic potential of RGS4 pharmacological manipulation for diseases characterized by sympathetic dysfunction or SNS hyperactivity, such as heart failure and hypertension.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"29 10","pages":"355"},"PeriodicalIF":3.3,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549250","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Cox Regression in Glioma Prognosis Analysis: Challenges with the Proportional Hazards Assumption and Coping Strategies. Comment on Ou Y, <i>et al</i>. \"<i>UBA2</i> as a Prognostic Biomarker and Potential Therapeutic Target in Glioma\". Frontiers in Bioscience-Landmark. 2024; 29: 144.","authors":"Jihao Xue, Ligang Chen","doi":"10.31083/j.fbl2910353","DOIUrl":"https://doi.org/10.31083/j.fbl2910353","url":null,"abstract":"","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"29 10","pages":"353"},"PeriodicalIF":3.3,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549320","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Gastric cancer (GC) is a significant global health burden with limited treatment options. The purpose of this study was to investigate the role of SLC30A2, a zinc transporter, in GC development and its capacity as a target for therapy.
Methods: A comprehensive analysis of GC datasets (GSE54129 and stomach adenocarcinoma (STAD) from The Cancer Genome Atlas (TCGA)) was conducted using bioinformatics tools to examine differential gene expression, focusing on SLC30A2. Functional assays, including Cell counting kit-8 (CCK-8) and transwell assays, were carried out on GC cell lines to determine the impact of SLC30A2 knockdown on cell behavior. Flow cytometry was utilized to quantitatively observe cell apoptosis and cell cycle progression. The impact of zinc sulfate (ZnSO4) on GC cells was evaluated by detecting apoptosis markers, Wnt/β-catenin signaling pathway activity, and oxidative stress biomarkers, focusing on the regulatory effect of SLC30A2 overexpression.
Results: Our analysis revealed significant upregulation of SLC30A2 in GC samples compared to normal samples, and high SLC30A2 expression was linked to poor prognosis. SLC30A2 knockdown repressed proliferation, invasion, and migration of GC cells, induced apoptosis, as well as arrested the cell cycle. Additionally, ZnSO4 treatment induced cytotoxicity and oxidative stress in GC cells, while SLC30A2 overexpression rescued ZnSO4-induced, migration, invasion, and proliferation. Moreover, ZnSO4 had been shown to bolster apoptosis and trigger the Wnt/β-catenin signaling pathway, effects which were mitigated by the overexpression of SLC30A2.
Conclusion: Our results implied that SLC30A2 was essential for GC progression by modulating zinc homeostasis and cellular processes. Targeting SLC30A2 or zinc signaling may represent a potential therapeutic approach for GC treatment.
{"title":"<i>SLC30A2</i>-Mediated Zinc Metabolism Modulates Gastric Cancer Progression via the Wnt/β-Catenin Signaling Pathway.","authors":"Fan Li, Xiaohong Zhang, Li Feng, Xingxing Zhang","doi":"10.31083/j.fbl2910351","DOIUrl":"https://doi.org/10.31083/j.fbl2910351","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) is a significant global health burden with limited treatment options. The purpose of this study was to investigate the role of <i>SLC30A2</i>, a zinc transporter, in GC development and its capacity as a target for therapy.</p><p><strong>Methods: </strong>A comprehensive analysis of GC datasets (GSE54129 and stomach adenocarcinoma (STAD) from The Cancer Genome Atlas (TCGA)) was conducted using bioinformatics tools to examine differential gene expression, focusing on <i>SLC30A2</i>. Functional assays, including Cell counting kit-8 (CCK-8) and transwell assays, were carried out on GC cell lines to determine the impact of <i>SLC30A2</i> knockdown on cell behavior. Flow cytometry was utilized to quantitatively observe cell apoptosis and cell cycle progression. The impact of zinc sulfate (ZnSO<sub>4</sub>) on GC cells was evaluated by detecting apoptosis markers, Wnt/β-catenin signaling pathway activity, and oxidative stress biomarkers, focusing on the regulatory effect of <i>SLC30A2</i> overexpression.</p><p><strong>Results: </strong>Our analysis revealed significant upregulation of <i>SLC30A2</i> in GC samples compared to normal samples, and high <i>SLC30A2</i> expression was linked to poor prognosis. <i>SLC30A2</i> knockdown repressed proliferation, invasion, and migration of GC cells, induced apoptosis, as well as arrested the cell cycle. Additionally, ZnSO<sub>4</sub> treatment induced cytotoxicity and oxidative stress in GC cells, while <i>SLC30A2</i> overexpression rescued ZnSO<sub>4</sub>-induced, migration, invasion, and proliferation. Moreover, ZnSO4 had been shown to bolster apoptosis and trigger the Wnt/β-catenin signaling pathway, effects which were mitigated by the overexpression of <i>SLC30A2</i>.</p><p><strong>Conclusion: </strong>Our results implied that <i>SLC30A2</i> was essential for GC progression by modulating zinc homeostasis and cellular processes. Targeting <i>SLC30A2</i> or zinc signaling may represent a potential therapeutic approach for GC treatment.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"29 10","pages":"351"},"PeriodicalIF":3.3,"publicationDate":"2024-10-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549311","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Svetlana V Kostyuk, Elena M Malinovskaya, Pavel E Umriukhin, Elena N Mikheeva, Elizaveta S Ershova, Ekaterina A Savinova, Larisa V Kameneva, Pavel A Troshin, Olga A Kraevaya, Ivan V Rodionov, Svetlana E Kostyuk, Tatyana A Salimova, Sergey I Kutsev, Natalia N Veiko
Background: The new synthesized water-soluble derivatives of C60 fullerenes are of a great interest to researchers since they can potentially be promising materials for drug delivery, bioimaging, biosonding, and tissue engineering. Surface functionalization of fullerene derivatives changes their chemical and physical characteristics, increasing their solubility and suitability for different biological systems applications, however, any changes in functionalized fullerenes can modulate their cytotoxicity and antioxidant properties. The toxic or protective effect of fullerene derivatives on cells is realized through the activation or inhibition of genes and proteins of key signaling pathways in cells responsible for regulation of cellular reactive oxygen species (ROS) level, proliferation, and apoptosis.
Methods: The 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay was used to assess cells viability. Flow cytometry analyses was applied to measure proteins levels in human embryonic lung fibroblasts (HELF) cells. HELF is a standard, stable and well described human cell line that can be passaged many times. Quantitation of ROS was assessed using H2DCFH-DA. Fluorescence images were obtained using microscopy. Expression of BCL2, CCND1, CDKN2A, BRCA1, BAX, NFKB1, NOX4, NRF2, TBP (reference gene) was analyzed using real-time Polymerase chain reaction (PCR).
Results: We found that high and low concentrations of fullerene C60 derivatives with the five residues of potassium salt of 6-(3-phenylpropanamido)hexanoic (F1) or 6-(2-(thiophen-2-yl)acetamido)hexanoic (F2) acid and a chlorine atom attached directly to the cage cause diametrically opposite activation of genes and proteins of key signaling pathways regulating the level of oxidative stress and apoptosis in HELF. High concentrations of F1 and F2 have a genotoxic effect, causing NADPH oxidase 4 (NOX4) expression activation in 24-72 hours (2-4 fold increase), ROS synthesis induction (increase by 30-40%), DNA damage and breaks (2-2.5 fold 8-oxodG level increases), and activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) (by 40-80%) against the background of reduced NF-E2-related factor 2 (NRF2) expression (by 20-45%). Low concentrations of F1 and F2 produced a cytoprotective effect: in 24-72 hours they reduce the oxidative DNA damage (by 20-40%), decrease the number of double-strand DNA breaks (by 20-30%), increase the level of anti-apoptotic proteins and enhance the antioxidant response activating the NRF2 expression (NRF2 gene expression increases 1.5-2.3 fold, phosphorylated form of the NRF2 protein increases 2-3 fold).
Conclusions: Obtained results show that in low doses studied fullrens may serve as perspective DNA protectors against the damaging genotoxic factors.
背景:新合成的 C60 富勒烯水溶性衍生物引起了研究人员的极大兴趣,因为它们有可能成为药物输送、生物成像、生物降解和组织工程方面的理想材料。富勒烯衍生物的表面官能化改变了它们的化学和物理特性,增加了它们的溶解度,使其更适合不同的生物系统应用,然而,官能化富勒烯的任何变化都会改变它们的细胞毒性和抗氧化特性。富勒烯衍生物对细胞的毒性或保护作用是通过激活或抑制细胞中负责调节细胞活性氧(ROS)水平、增殖和凋亡的关键信号通路的基因和蛋白质来实现的:方法:采用 3-[4,5-二甲基噻唑-2-基]-2,5-二苯基溴化四氮唑(MTT)检测法评估细胞活力。流式细胞仪分析法用于测量人胚胎肺成纤维细胞(HELF)中的蛋白质水平。HELF 是一种标准、稳定和描述良好的人类细胞系,可以多次传代。使用 H2DCFH-DA 对 ROS 定量进行评估。荧光图像通过显微镜获得。使用实时聚合酶链反应(PCR)分析了 BCL2、CCND1、CDKN2A、BRCA1、BAX、NFKB1、NOX4、NRF2 和 TBP(参考基因)的表达:我们发现,高浓度和低浓度的富勒烯 C60 衍生物与 6-(3-苯基丙酰胺基)己酸钾盐(F1)或 6-(2-(噻吩-2-基)乙酰胺基)己酸钾盐(F2)的五个残基以及直接连接在笼子上的一个氯原子,会对 HELF 中调节氧化应激和细胞凋亡水平的关键信号通路的基因和蛋白质产生截然相反的激活作用。高浓度的 F1 和 F2 具有基因毒性作用,可在 24-72 小时内导致 NADPH 氧化酶 4(NOX4)表达激活(增加 2-4 倍)、ROS 合成诱导(增加 30-40%)、DNA 损伤和断裂(8-oxodG 水平增加 2-2.在 NF-E2 相关因子 2(NRF2)表达减少(20-45%)的背景下,活化 B 细胞的核因子卡巴轻链增强子(NF-κB)被激活(40-80%)。低浓度的 F1 和 F2 能产生细胞保护作用:在 24-72 小时内,它们能减少 DNA 的氧化损伤(20-40%),减少 DNA 双链断裂的数量(20-30%),提高抗凋亡蛋白的水平,增强激活 NRF2 表达的抗氧化反应(NRF2 基因表达增加 1.5-2.3 倍,NRF2 蛋白的磷酸化形式增加 2-3 倍):结论:研究结果表明,在低剂量情况下,所研究的富勒烯可作为 DNA 保护剂,抵御基因毒性因素的破坏。
{"title":"Dose-Response Effect of Various Concentrations of Cl-Containing Water-Soluble Derivatives of C<sub>60</sub> Fullerenes on a Selective Regulation of Gene Expression in Human Embryonic Lung Fibroblasts (HELF).","authors":"Svetlana V Kostyuk, Elena M Malinovskaya, Pavel E Umriukhin, Elena N Mikheeva, Elizaveta S Ershova, Ekaterina A Savinova, Larisa V Kameneva, Pavel A Troshin, Olga A Kraevaya, Ivan V Rodionov, Svetlana E Kostyuk, Tatyana A Salimova, Sergey I Kutsev, Natalia N Veiko","doi":"10.31083/j.fbl2910352","DOIUrl":"https://doi.org/10.31083/j.fbl2910352","url":null,"abstract":"<p><strong>Background: </strong>The new synthesized water-soluble derivatives of C<sub>60</sub> fullerenes are of a great interest to researchers since they can potentially be promising materials for drug delivery, bioimaging, biosonding, and tissue engineering. Surface functionalization of fullerene derivatives changes their chemical and physical characteristics, increasing their solubility and suitability for different biological systems applications, however, any changes in functionalized fullerenes can modulate their cytotoxicity and antioxidant properties. The toxic or protective effect of fullerene derivatives on cells is realized through the activation or inhibition of genes and proteins of key signaling pathways in cells responsible for regulation of cellular reactive oxygen species (ROS) level, proliferation, and apoptosis.</p><p><strong>Methods: </strong>The 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay was used to assess cells viability. Flow cytometry analyses was applied to measure proteins levels in human embryonic lung fibroblasts (HELF) cells. HELF is a standard, stable and well described human cell line that can be passaged many times. Quantitation of ROS was assessed using H2DCFH-DA. Fluorescence images were obtained using microscopy. Expression of <i>BCL2</i>, <i>CCND1</i>, <i>CDKN2A</i>, <i>BRCA1</i>, <i>BAX</i>, <i>NFKB1</i>, <i>NOX4</i>, <i>NRF2</i>, <i>TBP</i> (reference gene) was analyzed using real-time Polymerase chain reaction (PCR).</p><p><strong>Results: </strong>We found that high and low concentrations of fullerene C<sub>60</sub> derivatives with the five residues of potassium salt of 6-(3-phenylpropanamido)hexanoic (F1) or 6-(2-(thiophen-2-yl)acetamido)hexanoic (F2) acid and a chlorine atom attached directly to the cage cause diametrically opposite activation of genes and proteins of key signaling pathways regulating the level of oxidative stress and apoptosis in HELF. High concentrations of F1 and F2 have a genotoxic effect, causing NADPH oxidase 4 (NOX4) expression activation in 24-72 hours (2-4 fold increase), ROS synthesis induction (increase by 30-40%), DNA damage and breaks (2-2.5 fold 8-oxodG level increases), and activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) (by 40-80%) against the background of reduced NF-E2-related factor 2 (NRF2) expression (by 20-45%). Low concentrations of F1 and F2 produced a cytoprotective effect: in 24-72 hours they reduce the oxidative DNA damage (by 20-40%), decrease the number of double-strand DNA breaks (by 20-30%), increase the level of anti-apoptotic proteins and enhance the antioxidant response activating the NRF2 expression (NRF2 gene expression increases 1.5-2.3 fold, phosphorylated form of the NRF2 protein increases 2-3 fold).</p><p><strong>Conclusions: </strong>Obtained results show that in low doses studied fullrens may serve as perspective DNA protectors against the damaging genotoxic factors.</p>","PeriodicalId":73069,"journal":{"name":"Frontiers in bioscience (Landmark edition)","volume":"29 10","pages":"352"},"PeriodicalIF":3.3,"publicationDate":"2024-10-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142549323","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}