首页 > 最新文献

Biomolecules & Therapeutics最新文献

英文 中文
Mechanism of Action and Pharmacokinetics of Approved Bispecific Antibodies. 已获批准的双特异性抗体的作用机制和药代动力学。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-11-01 Epub Date: 2024-10-25 DOI: 10.4062/biomolther.2024.146
Seong Min Choi, Ju-Hee Lee, Soyeon Ko, Soon-Sun Hong, Hyo-Eon Jin

Bispecific antibodies represent a significant advancement in therapeutic antibody engineering, offering the ability to simultaneously target two distinct antigens. This dual-targeting capability enhances therapeutic efficacy, especially in complex diseases, such as cancer and autoimmune disorders, where drug resistance and incomplete target coverage are prevalent challenges. Bispecific antibodies facilitate immune cell engagement and disrupt multiple signaling pathways, providing a more comprehensive treatment approach than traditional monoclonal antibodies. However, the intricate structure of bispecific antibodies introduces unique pharmacokinetic challenges, including issues related to their absorption, distribution, metabolism, and excretion, which can significantly affect their efficacy and safety. This review provides an in-depth analysis of the structural design, mechanisms of action, and pharmacokinetics of the currently approved bispecific antibodies. It also highlights the engineering innovations that have been implemented to overcome these challenges, such as Fc modifications and advanced dimerization techniques, which enhance the stability and half-life of bispecific antibodies. Significant progress has been made in bispecific antibody technology; however, further research is necessary to broaden their clinical applications, enhance their safety profiles, and optimize their incorporation into combination therapies. Continuous advancements in this field are expected to enable bispecific antibodies to provide more precise and effective therapeutic strategies for a range of complex diseases, ultimately improving patient outcomes and advancing precision medicine.

双特异性抗体是治疗性抗体工程的一大进步,能够同时靶向两种不同的抗原。这种双重靶向能力提高了疗效,尤其是在癌症和自身免疫性疾病等复杂疾病中,耐药性和靶点覆盖不全是普遍面临的挑战。双特异性抗体可促进免疫细胞参与并破坏多种信号通路,提供比传统单克隆抗体更全面的治疗方法。然而,双特异性抗体错综复杂的结构带来了独特的药代动力学挑战,包括与吸收、分布、代谢和排泄相关的问题,这可能会严重影响其疗效和安全性。本综述深入分析了目前获批的双特异性抗体的结构设计、作用机制和药代动力学。它还重点介绍了为克服这些挑战而实施的工程创新,如 Fc 修饰和先进的二聚化技术,这些技术提高了双特异性抗体的稳定性和半衰期。双特异性抗体技术已取得重大进展,但仍需进一步研究,以扩大其临床应用,提高其安全性,并优化其在联合疗法中的应用。该领域的不断进步有望使双特异性抗体为一系列复杂疾病提供更精确、更有效的治疗策略,最终改善患者预后,推动精准医疗的发展。
{"title":"Mechanism of Action and Pharmacokinetics of Approved Bispecific Antibodies.","authors":"Seong Min Choi, Ju-Hee Lee, Soyeon Ko, Soon-Sun Hong, Hyo-Eon Jin","doi":"10.4062/biomolther.2024.146","DOIUrl":"10.4062/biomolther.2024.146","url":null,"abstract":"<p><p>Bispecific antibodies represent a significant advancement in therapeutic antibody engineering, offering the ability to simultaneously target two distinct antigens. This dual-targeting capability enhances therapeutic efficacy, especially in complex diseases, such as cancer and autoimmune disorders, where drug resistance and incomplete target coverage are prevalent challenges. Bispecific antibodies facilitate immune cell engagement and disrupt multiple signaling pathways, providing a more comprehensive treatment approach than traditional monoclonal antibodies. However, the intricate structure of bispecific antibodies introduces unique pharmacokinetic challenges, including issues related to their absorption, distribution, metabolism, and excretion, which can significantly affect their efficacy and safety. This review provides an in-depth analysis of the structural design, mechanisms of action, and pharmacokinetics of the currently approved bispecific antibodies. It also highlights the engineering innovations that have been implemented to overcome these challenges, such as Fc modifications and advanced dimerization techniques, which enhance the stability and half-life of bispecific antibodies. Significant progress has been made in bispecific antibody technology; however, further research is necessary to broaden their clinical applications, enhance their safety profiles, and optimize their incorporation into combination therapies. Continuous advancements in this field are expected to enable bispecific antibodies to provide more precise and effective therapeutic strategies for a range of complex diseases, ultimately improving patient outcomes and advancing precision medicine.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"708-722"},"PeriodicalIF":3.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11535297/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142494494","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Translation Initiation Factor-2S2 (eIF2S2) Contributes to Cervical Carcinogenesis by Inhibiting the TGF-β/SMAD4 Signaling Pathway. 翻译起始因子-2S2 (eIF2S2) 通过抑制 TGF-β/SMAD4 信号通路促进宫颈癌的发生
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-11-01 Epub Date: 2024-10-07 DOI: 10.4062/biomolther.2024.024
Juthika Kundu, Hobin Yang, Saerom Moon, Mi Ran Byun, Young Kee Shin, Kyoung Song, Joon-Seok Choi

The deregulation of protein translational machinery and the oncogenic role of several translation initiation factors have been extensively investigated. This study aimed to investigate the role of eukaryotic translation initiation factor 2S2 (eIF2S2, also known as eIF2β) in cervical carcinogenesis. Immunohistochemical analysis of human cervical carcinoma tissues revealed a stage-specific increase in eIF2S2 expression. The knockdown of eIF2S2 in human cervical cancer (SiHa) cells significantly reduced growth and migration properties, whereas its overexpression demonstrated the opposite effect. Immunoprecipitation and Bimolecular fluorescence complementation (BiFC) assay confirmed the previous photo array finding of the interaction between eIF2S2 and SMAD4 to understand the tumorigenic mechanism of eIF2S2. The results indicated that the N-terminus of eIF2S2 interacts with the MH-1 domain of SMAD4. The interaction effect between eIF2S2 and SMAD4 was further evaluated. The knockdown of eIF2S2 increased SMAD4 expression in cervical cancer cells without changing SMAD4 mRNA expression, whereas transient eIF2S2 overexpression reduced SMAD4 expression. This indicates the possibility of post-translational regulation of SMAD4 expression by eIF2S2. Additionally, eIF2S2 overexpression was confirmed to weaken the expression and/or promoter activity of p15 and p27, which are SMAD4-regulated antiproliferative proteins, by reducing SMAD4 levels. Therefore, our study indicated the pro-tumorigenic role of eIF2S2, which diminishes both SMAD4 expression and function as a transcriptional factor in cervical carcinogenesis.

蛋白质翻译机制的失调以及多种翻译起始因子的致癌作用已被广泛研究。本研究旨在探讨真核翻译起始因子 2S2(eIF2S2,又称 eIF2β)在宫颈癌发生中的作用。对人类宫颈癌组织的免疫组化分析表明,eIF2S2的表达呈阶段性增加。在人宫颈癌(SiHa)细胞中敲除 eIF2S2 能显著降低其生长和迁移特性,而过表达则表现出相反的效果。免疫沉淀和双分子荧光互补(BiFC)分析证实了之前的光阵发现的eIF2S2与SMAD4之间的相互作用,从而了解了eIF2S2的致瘤机制。结果表明,eIF2S2的N端与SMAD4的MH-1结构域相互作用。研究进一步评估了eIF2S2与SMAD4之间的相互作用效应。在不改变 SMAD4 mRNA 表达的情况下,敲除 eIF2S2 会增加宫颈癌细胞中 SMAD4 的表达,而一过性表达 eIF2S2 则会降低 SMAD4 的表达。这表明 eIF2S2 有可能对 SMAD4 的表达进行翻译后调控。此外,eIF2S2 的过表达被证实会通过降低 SMAD4 水平来削弱 p15 和 p27 的表达和/或启动子活性,而这两种蛋白是 SMAD4 调控的抗增殖蛋白。因此,我们的研究表明,eIF2S2 在宫颈癌发生过程中具有促肿瘤作用,它既能降低 SMAD4 的表达,又能降低其作为转录因子的功能。
{"title":"Translation Initiation Factor-2S2 (eIF2S2) Contributes to Cervical Carcinogenesis by Inhibiting the TGF-β/SMAD4 Signaling Pathway.","authors":"Juthika Kundu, Hobin Yang, Saerom Moon, Mi Ran Byun, Young Kee Shin, Kyoung Song, Joon-Seok Choi","doi":"10.4062/biomolther.2024.024","DOIUrl":"10.4062/biomolther.2024.024","url":null,"abstract":"<p><p>The deregulation of protein translational machinery and the oncogenic role of several translation initiation factors have been extensively investigated. This study aimed to investigate the role of eukaryotic translation initiation factor 2S2 (eIF2S2, also known as eIF2β) in cervical carcinogenesis. Immunohistochemical analysis of human cervical carcinoma tissues revealed a stage-specific increase in eIF2S2 expression. The knockdown of eIF2S2 in human cervical cancer (SiHa) cells significantly reduced growth and migration properties, whereas its overexpression demonstrated the opposite effect. Immunoprecipitation and Bimolecular fluorescence complementation (BiFC) assay confirmed the previous photo array finding of the interaction between eIF2S2 and SMAD4 to understand the tumorigenic mechanism of eIF2S2. The results indicated that the N-terminus of eIF2S2 interacts with the MH-1 domain of SMAD4. The interaction effect between eIF2S2 and SMAD4 was further evaluated. The knockdown of eIF2S2 increased SMAD4 expression in cervical cancer cells without changing SMAD4 mRNA expression, whereas transient eIF2S2 overexpression reduced SMAD4 expression. This indicates the possibility of post-translational regulation of SMAD4 expression by eIF2S2. Additionally, eIF2S2 overexpression was confirmed to weaken the expression and/or promoter activity of p15 and p27, which are SMAD4-regulated antiproliferative proteins, by reducing SMAD4 levels. Therefore, our study indicated the pro-tumorigenic role of eIF2S2, which diminishes both SMAD4 expression and function as a transcriptional factor in cervical carcinogenesis.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"767-777"},"PeriodicalIF":3.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11535292/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142380052","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Phytotherapeutic BS012 and Its Active Component Ameliorate Allergic Asthma via Inhibition of Th2-Mediated Immune Response and Apoptosis. 植物疗法 BS012 及其活性成分通过抑制 Th2 介导的免疫反应和细胞凋亡改善过敏性哮喘。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-11-01 Epub Date: 2024-10-07 DOI: 10.4062/biomolther.2024.058
Siqi Zhang, Joonki Kim, Gakyung Lee, Hong Ryul Ahn, Yeo Eun Kim, Hee Ju Kim, Jae Sik Yu, Miso Park, Keon Wook Kang, Hocheol Kim, Byung Hwa Jung, Sung Won Kwon, Dae Sik Jang, Hyun Ok Yang

Asthma is a chronic inflammatory disorder of the lungs that results in airway inflammation and narrowing. BS012 is an herbal remedy containing Asarum sieboldii, Platycodon grandiflorum, and Cinnamomum cassia extracts. To elucidate the anti-asthma effect of BS012, this study analyzed the immune response, respiratory protection, and changes in metabolic mechanisms in an ovalbumin-induced allergic asthma mouse model. Female BALB/c mice were exposed to ovalbumin to induce allergic asthma. Bronchoalveolar lavage fluid and plasma were analyzed for interleukin and immunoglobulin E levels. Histological analyses of the lungs were performed to measure morphological changes. Apoptosis-related mediators were assayed by western blotting. Plasma and lung tissue metabolomic analyses were performed to investigate the metabolic changes. A T-helper-2-like differentiated cell model was used to identify the active components of BS012. BS012 treatment improved inflammatory cell infiltration, mucus production, and goblet cell hyperplasia in lung tissues. BS012 also significantly downregulated ovalbumin-specific immunoglobulin E in plasma and T-helper-2-specific cytokines, interleukin-4 and -5, in bronchoalveolar lavage fluid. The lungs of ovalbumin-inhaled mice exhibited nerve growth factor-mediated apoptotic protein expression, which was significantly attenuated by BS012 treatment. Ovalbumin-induced abnormalities in amino acid and lipid metabolism were improved by BS012 in correlation with its anti-inflammatory properties and normalization of energy metabolism. Additionally, the differentiated cell model revealed that N-isobutyl-dodecatetraenamide is an active component that contributes to the anti-allergic properties of BS012. The current findings demonstrate the anti-allergic and respiratory protective functions of BS012 against allergic asthma, which can be considered a therapeutic candidate.

哮喘是一种慢性肺部炎症性疾病,会导致气道发炎和狭窄。BS012 是一种草药疗法,含有西洋菝葜、桔梗和肉桂提取物。为了阐明 BS012 的抗哮喘作用,本研究分析了卵清蛋白诱导的过敏性哮喘小鼠模型的免疫反应、呼吸保护和代谢机制的变化。雌性 BALB/c 小鼠接触卵清蛋白诱发过敏性哮喘。分析支气管肺泡灌洗液和血浆中的白细胞介素和免疫球蛋白 E 水平。对肺部进行组织学分析,以测量形态学变化。用 Western 印迹法测定与细胞凋亡相关的介质。血浆和肺组织代谢组分析用于研究代谢变化。使用 T-helper-2 样分化细胞模型来鉴定 BS012 的活性成分。BS012 治疗改善了肺组织的炎症细胞浸润、粘液分泌和上睑球细胞增生。BS012 还能显著下调血浆中的卵清蛋白特异性免疫球蛋白 E 和支气管肺泡灌洗液中的 T-helper-2 特异性细胞因子、白细胞介素-4 和白细胞介素-5。卵清蛋白吸入小鼠的肺部表现出神经生长因子介导的凋亡蛋白表达,BS012 治疗可显著减少这种表达。BS012 可改善卵清蛋白诱导的氨基酸和脂质代谢异常,这与 BS012 的抗炎特性和能量代谢正常化有关。此外,分化细胞模型显示,N-异丁基十二碳四烯酰胺是促进 BS012 抗过敏特性的活性成分。目前的研究结果表明,BS012 对过敏性哮喘具有抗过敏和呼吸保护功能,可被视为一种候选治疗药物。
{"title":"Phytotherapeutic BS012 and Its Active Component Ameliorate Allergic Asthma via Inhibition of Th2-Mediated Immune Response and Apoptosis.","authors":"Siqi Zhang, Joonki Kim, Gakyung Lee, Hong Ryul Ahn, Yeo Eun Kim, Hee Ju Kim, Jae Sik Yu, Miso Park, Keon Wook Kang, Hocheol Kim, Byung Hwa Jung, Sung Won Kwon, Dae Sik Jang, Hyun Ok Yang","doi":"10.4062/biomolther.2024.058","DOIUrl":"10.4062/biomolther.2024.058","url":null,"abstract":"<p><p>Asthma is a chronic inflammatory disorder of the lungs that results in airway inflammation and narrowing. BS012 is an herbal remedy containing <i>Asarum sieboldii</i>, <i>Platycodon grandiflorum</i>, and <i>Cinnamomum cassia</i> extracts. To elucidate the anti-asthma effect of BS012, this study analyzed the immune response, respiratory protection, and changes in metabolic mechanisms in an ovalbumin-induced allergic asthma mouse model. Female BALB/c mice were exposed to ovalbumin to induce allergic asthma. Bronchoalveolar lavage fluid and plasma were analyzed for interleukin and immunoglobulin E levels. Histological analyses of the lungs were performed to measure morphological changes. Apoptosis-related mediators were assayed by western blotting. Plasma and lung tissue metabolomic analyses were performed to investigate the metabolic changes. A T-helper-2-like differentiated cell model was used to identify the active components of BS012. BS012 treatment improved inflammatory cell infiltration, mucus production, and goblet cell hyperplasia in lung tissues. BS012 also significantly downregulated ovalbumin-specific immunoglobulin E in plasma and T-helper-2-specific cytokines, interleukin-4 and -5, in bronchoalveolar lavage fluid. The lungs of ovalbumin-inhaled mice exhibited nerve growth factor-mediated apoptotic protein expression, which was significantly attenuated by BS012 treatment. Ovalbumin-induced abnormalities in amino acid and lipid metabolism were improved by BS012 in correlation with its anti-inflammatory properties and normalization of energy metabolism. Additionally, the differentiated cell model revealed that <i>N</i>-isobutyl-dodecatetraenamide is an active component that contributes to the anti-allergic properties of BS012. The current findings demonstrate the anti-allergic and respiratory protective functions of BS012 against allergic asthma, which can be considered a therapeutic candidate.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"744-758"},"PeriodicalIF":3.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11535288/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142380050","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Multifaceted Role of Epithelial Membrane Protein 2 in Cancer: from Biomarker to Therapeutic Target. 上皮细胞膜蛋白 2 在癌症中的多方面作用:从生物标记物到治疗目标。
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-11-01 Epub Date: 2024-10-21 DOI: 10.4062/biomolther.2024.168
Ji Yun Jang, Mi Kyung Park, Chang Hoon Lee, Ho Lee

Tetraspanin superfamily proteins not only facilitate the trafficking of specific proteins to distinct plasma membrane domains but also influence cell-to-cell and cell-extracellular matrix interactions. Among these proteins, Epithelial Membrane Protein 2 (EMP2), a member of the growth arrest-specific gene 3/peripheral myelin protein 22 (GAS3/PMP22) family, is known to affect key cellular processes. Recent studies have revealed that EMP2 modulates critical signaling pathways and interacts with adhesion molecules and growth factor receptors, underscoring its potential as a biomarker for cancer diagnosis and prognosis. These findings suggest that EMP2 expression patterns could provide valuable insights into tumorigenesis and metastasis. Moreover, EMP2 has emerged as a promising therapeutic target, with approaches aimed at inhibiting or modulating its activity showing potential to disrupt tumor growth and metastasis. This review provides a comprehensive overview of recent advances in understanding the multifaceted roles of EMP2 in cancer, with a focus on its underlying mechanisms and clinical significance.

四跨蛋白超家族蛋白质不仅能促进特定蛋白质向不同质膜域的迁移,还能影响细胞与细胞之间以及细胞与细胞外基质之间的相互作用。在这些蛋白中,上皮细胞膜蛋白 2(EMP2)是生长停滞特异性基因 3/外周髓鞘蛋白 22(GAS3/PMP22)家族的成员,已知会影响关键的细胞过程。最近的研究发现,EMP2 可调节关键的信号通路,并与粘附分子和生长因子受体相互作用,这突显了它作为癌症诊断和预后生物标记物的潜力。这些研究结果表明,EMP2 的表达模式可为了解肿瘤发生和转移提供有价值的信息。此外,EMP2 已成为一个很有前景的治疗靶点,旨在抑制或调节其活性的方法显示出破坏肿瘤生长和转移的潜力。这篇综述全面概述了在了解 EMP2 在癌症中的多方面作用方面的最新进展,重点关注其潜在机制和临床意义。
{"title":"The Multifaceted Role of Epithelial Membrane Protein 2 in Cancer: from Biomarker to Therapeutic Target.","authors":"Ji Yun Jang, Mi Kyung Park, Chang Hoon Lee, Ho Lee","doi":"10.4062/biomolther.2024.168","DOIUrl":"10.4062/biomolther.2024.168","url":null,"abstract":"<p><p>Tetraspanin superfamily proteins not only facilitate the trafficking of specific proteins to distinct plasma membrane domains but also influence cell-to-cell and cell-extracellular matrix interactions. Among these proteins, Epithelial Membrane Protein 2 (EMP2), a member of the growth arrest-specific gene 3/peripheral myelin protein 22 (GAS3/PMP22) family, is known to affect key cellular processes. Recent studies have revealed that EMP2 modulates critical signaling pathways and interacts with adhesion molecules and growth factor receptors, underscoring its potential as a biomarker for cancer diagnosis and prognosis. These findings suggest that EMP2 expression patterns could provide valuable insights into tumorigenesis and metastasis. Moreover, EMP2 has emerged as a promising therapeutic target, with approaches aimed at inhibiting or modulating its activity showing potential to disrupt tumor growth and metastasis. This review provides a comprehensive overview of recent advances in understanding the multifaceted roles of EMP2 in cancer, with a focus on its underlying mechanisms and clinical significance.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"697-707"},"PeriodicalIF":3.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11535296/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142457187","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pyronaridine Inhibited MUC5AC Mucin Gene Expression by Regulation of Nuclear Factor Kappa B Signaling Pathway in Human Pulmonary Mucoepidermoid Cells. 吡咯烷酮通过调控核因子卡巴B信号通路抑制人肺黏液表皮样细胞中MUC5AC黏蛋白基因的表达
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-09-01 Epub Date: 2024-08-02 DOI: 10.4062/biomolther.2024.072
Rajib Hossain, Hyun Jae Lee, Choong Jae Lee

In this study, the potential effects of pyronaridine, an antimalarial agent, on airway MUC5AC mucin gene expression were investigated. The human pulmonary epithelial NCI-H292 cells were pretreated with pyronaridine for 30 min and then stimulated with phorbol 12-myristate 13-acetate (PMA) for 24 h. The effect of pyronaridine on the PMA-induced nuclear factor kappa B (NF-κB) signaling pathway was also examined. Pyronaridine inhibited glycoprotein production and mRNA expression of MUC5AC mucins induced by PMA through the inhibition of degradation of inhibitory kappa Bα and NF-κB p65 nuclear translocation. These results suggest that pyronaridine suppresses gene expression of mucin through regulation of the NF-κB signaling pathway in human pulmonary epithelial cells.

本研究探讨了抗疟药物吡咯烷酮对气道 MUC5AC 粘蛋白基因表达的潜在影响。研究还考察了吡咯烷对 PMA 诱导的核因子卡巴 B(NF-κB)信号通路的影响。吡咯那啶通过抑制抑制性卡巴Bα的降解和NF-κB p65的核转位,抑制了PMA诱导的糖蛋白生成和MUC5AC粘蛋白的mRNA表达。这些结果表明,吡咯烷酮通过调节人肺上皮细胞的 NF-κB 信号通路抑制粘蛋白的基因表达。
{"title":"Pyronaridine Inhibited <i>MUC5AC</i> Mucin Gene Expression by Regulation of Nuclear Factor Kappa B Signaling Pathway in Human Pulmonary Mucoepidermoid Cells.","authors":"Rajib Hossain, Hyun Jae Lee, Choong Jae Lee","doi":"10.4062/biomolther.2024.072","DOIUrl":"10.4062/biomolther.2024.072","url":null,"abstract":"<p><p>In this study, the potential effects of pyronaridine, an antimalarial agent, on airway <i>MUC5AC</i> mucin gene expression were investigated. The human pulmonary epithelial NCI-H292 cells were pretreated with pyronaridine for 30 min and then stimulated with phorbol 12-myristate 13-acetate (PMA) for 24 h. The effect of pyronaridine on the PMA-induced nuclear factor kappa B (NF-κB) signaling pathway was also examined. Pyronaridine inhibited glycoprotein production and mRNA expression of <i>MUC5AC</i> mucins induced by PMA through the inhibition of degradation of inhibitory kappa Bα and NF-κB p65 nuclear translocation. These results suggest that pyronaridine suppresses gene expression of mucin through regulation of the NF-κB signaling pathway in human pulmonary epithelial cells.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"540-545"},"PeriodicalIF":3.0,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11392666/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141874074","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Combination of Gefitinib and Acetaminophen Exacerbates Hepatotoxicity via ROS-Mediated Apoptosis. 吉非替尼与对乙酰氨基酚联用会通过ROS介导的细胞凋亡加剧肝毒性
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-09-01 Epub Date: 2024-06-14 DOI: 10.4062/biomolther.2023.209
Jiangxin Xu, Xiangliang Huang, Yourong Zhou, Zhifei Xu, Xinjun Cai, Bo Yang, Qiaojun He, Peihua Luo, Hao Yan, Jie Jin

Gefitinib is the well-tolerated first-line treatment of non-small cell lung cancer. As it need for analgesics during oncology treatment, particularly in the context ofthe coronavirus disease, where patients are more susceptible to contract high fever and sore throat. This has increased the likelihood of taking both gefitinib and antipyretic analgesic acetaminophen (APAP). Given that gefitinib and APAP overdose can predispose patients to liver injury or even acute liver failure, there is a risk of severe hepatotoxicity when these two drugs are used concomitantly. However, little is known regarding their safety at therapeutic doses. This study simulated the administration of gefitinib and APAP at clinically relevant doses in an animal model and confirmed that gefitinib in combination with APAP exhibited additional hepatotoxicity. We found that gefitinib plus APAP significantly exacerbated cell death, whereas each drug by itself had little or minor effect on hepatocyte survival. Mechanistically, combination of gefitinib and APAP induces hepatocyte death via the apoptotic pathway obviously. Reactive oxygen species (ROS) generation and DNA damage accumulation are involved in hepatocyte apoptosis. Gefitinib plus APAP also promotes the expression of Kelch-like ECH-associated protein 1 (Keap1) and downregulated the antioxidant factor, Nuclear factor erythroid 2-related factor 2 (Nrf2), by inhibiting p62 expression. Taken together, this study revealed the potential ROS-mediated apoptosis-dependent hepatotoxicity effect of the combination of gefitinib and APAP, in which the p62/Keap1/Nrf2 signaling pathway participates and plays an important regulatory role.

吉非替尼是治疗非小细胞肺癌的耐受性良好的一线药物。由于肿瘤治疗期间需要镇痛药,特别是在冠状病毒疾病的情况下,患者更容易感染高烧和咽喉痛。这增加了同时服用吉非替尼和解热镇痛药对乙酰氨基酚(APAP)的可能性。鉴于吉非替尼和对乙酰氨基酚过量服用会导致患者肝损伤甚至急性肝衰竭,因此同时使用这两种药物时存在严重肝毒性的风险。然而,人们对这两种药物在治疗剂量下的安全性知之甚少。本研究在动物模型中模拟了吉非替尼和 APAP 的临床相关剂量给药,结果证实吉非替尼与 APAP 联用会产生额外的肝毒性。我们发现,吉非替尼加 APAP 会明显加剧细胞死亡,而每种药物本身对肝细胞存活的影响很小或微乎其微。从机理上讲,吉非替尼和 APAP 联用明显通过凋亡途径诱导肝细胞死亡。活性氧(ROS)生成和 DNA 损伤积累参与了肝细胞凋亡。吉非替尼加 APAP 还能促进 Kelch 样 ECH 相关蛋白 1(Keap1)的表达,并通过抑制 p62 的表达下调抗氧化因子--核因子红细胞 2 相关因子 2(Nrf2)。综上所述,本研究揭示了吉非替尼和 APAP 联合用药潜在的 ROS 介导的凋亡依赖性肝毒性效应,p62/Keap1/Nrf2 信号通路在其中参与并发挥了重要的调控作用。
{"title":"The Combination of Gefitinib and Acetaminophen Exacerbates Hepatotoxicity via ROS-Mediated Apoptosis.","authors":"Jiangxin Xu, Xiangliang Huang, Yourong Zhou, Zhifei Xu, Xinjun Cai, Bo Yang, Qiaojun He, Peihua Luo, Hao Yan, Jie Jin","doi":"10.4062/biomolther.2023.209","DOIUrl":"10.4062/biomolther.2023.209","url":null,"abstract":"<p><p>Gefitinib is the well-tolerated first-line treatment of non-small cell lung cancer. As it need for analgesics during oncology treatment, particularly in the context ofthe coronavirus disease, where patients are more susceptible to contract high fever and sore throat. This has increased the likelihood of taking both gefitinib and antipyretic analgesic acetaminophen (APAP). Given that gefitinib and APAP overdose can predispose patients to liver injury or even acute liver failure, there is a risk of severe hepatotoxicity when these two drugs are used concomitantly. However, little is known regarding their safety at therapeutic doses. This study simulated the administration of gefitinib and APAP at clinically relevant doses in an animal model and confirmed that gefitinib in combination with APAP exhibited additional hepatotoxicity. We found that gefitinib plus APAP significantly exacerbated cell death, whereas each drug by itself had little or minor effect on hepatocyte survival. Mechanistically, combination of gefitinib and APAP induces hepatocyte death via the apoptotic pathway obviously. Reactive oxygen species (ROS) generation and DNA damage accumulation are involved in hepatocyte apoptosis. Gefitinib plus APAP also promotes the expression of Kelch-like ECH-associated protein 1 (Keap1) and downregulated the antioxidant factor, Nuclear factor erythroid 2-related factor 2 (Nrf2), by inhibiting p62 expression. Taken together, this study revealed the potential ROS-mediated apoptosis-dependent hepatotoxicity effect of the combination of gefitinib and APAP, in which the p62/Keap1/Nrf2 signaling pathway participates and plays an important regulatory role.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"647-657"},"PeriodicalIF":3.0,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11392667/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141316649","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
β-Lapachone Exerts Hypnotic Effects via Adenosine A1 Receptor in Mice. β-拉帕醌通过腺苷 A1 受体对小鼠产生催眠作用
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-09-01 Epub Date: 2024-08-21 DOI: 10.4062/biomolther.2024.106
Do Hyun Lee, Hye Jin Jee, Yi-Sook Jung

Sleep is one of the most essential physiological phenomena for maintaining health. Sleep disturbances, such as insomnia, are often accompanied by psychiatric or physical conditions such as impaired attention, anxiety, and stress. Medication used to treat insomnia have concerns about potential side effects with long-term use, so interest in the use of alternative medicine is increasing. In this study, we investigated the hypnotic effects of β-lapachone (β-Lap), a natural naphthoquinone compound, using pentobarbital-induced sleep test, immunohistochemistry, real-time PCR, and western blot in mice. Our results indicated that β-Lap exerts a significant hypnotic effect by showing a decrease in sleep onset latency and an increase in total sleep time in pentobarbital-induced sleep model. The results of c-Fos immunostaining showed that β-Lap decreased neuronal activity in the basal forebrain and lateral hypothalamus, which are wakefulness-promoting brain regions, while increasing in the ventrolateral preoptic nucleus, a sleep-promoting region; all these effects were significantly abolished by 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), an adenosine A1 receptor (A1R) antagonist. Western blot analysis showed that β-Lap increased extracellular signalregulated kinase phosphorylation and nuclear factor-kappa B translocation from the cytoplasm to the nucleus; these effects were inhibited by DPCPX. Additionally, β-Lap increased the mRNA levels of A1R. Taken together, these results suggest that β-Lap exerts hypnotic effects, potentially through A1R.

睡眠是维持健康最基本的生理现象之一。失眠等睡眠障碍通常伴随着精神或身体状况,如注意力不集中、焦虑和压力。用于治疗失眠的药物存在长期使用可能产生副作用的问题,因此人们对使用替代药物的兴趣与日俱增。在这项研究中,我们使用戊巴比妥诱导小鼠睡眠试验、免疫组织化学、实时 PCR 和 Western 印迹法研究了天然萘醌化合物 β-拉帕醌(β-Lap)的催眠作用。结果表明,在戊巴比妥诱导的睡眠模型中,β-Lap具有明显的催眠作用,能降低睡眠开始潜伏期,增加总睡眠时间。c-Fos免疫染色结果显示,β-Lap降低了前脑基底层和下丘脑外侧神经元的活性,而增加了视前核外侧神经元的活性,视前核外侧神经元是促进睡眠的区域;所有这些效应都被腺苷A1受体(A1R)拮抗剂8-环戊基-1,3-二丙基黄嘌呤(DPCPX)显著消除。Western 印迹分析显示,β-Lap 增加了细胞外信号调节激酶的磷酸化和核因子-kappa B 从细胞质到细胞核的转位;DPCPX 抑制了这些效应。此外,β-Lap 还增加了 A1R 的 mRNA 水平。综上所述,这些结果表明,β-Lap 可能通过 A1R 发挥催眠作用。
{"title":"β-Lapachone Exerts Hypnotic Effects via Adenosine A<sub>1</sub> Receptor in Mice.","authors":"Do Hyun Lee, Hye Jin Jee, Yi-Sook Jung","doi":"10.4062/biomolther.2024.106","DOIUrl":"10.4062/biomolther.2024.106","url":null,"abstract":"<p><p>Sleep is one of the most essential physiological phenomena for maintaining health. Sleep disturbances, such as insomnia, are often accompanied by psychiatric or physical conditions such as impaired attention, anxiety, and stress. Medication used to treat insomnia have concerns about potential side effects with long-term use, so interest in the use of alternative medicine is increasing. In this study, we investigated the hypnotic effects of β-lapachone (β-Lap), a natural naphthoquinone compound, using pentobarbital-induced sleep test, immunohistochemistry, real-time PCR, and western blot in mice. Our results indicated that β-Lap exerts a significant hypnotic effect by showing a decrease in sleep onset latency and an increase in total sleep time in pentobarbital-induced sleep model. The results of c-Fos immunostaining showed that β-Lap decreased neuronal activity in the basal forebrain and lateral hypothalamus, which are wakefulness-promoting brain regions, while increasing in the ventrolateral preoptic nucleus, a sleep-promoting region; all these effects were significantly abolished by 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), an adenosine A<sub>1</sub> receptor (A<sub>1</sub>R) antagonist. Western blot analysis showed that β-Lap increased extracellular signalregulated kinase phosphorylation and nuclear factor-kappa B translocation from the cytoplasm to the nucleus; these effects were inhibited by DPCPX. Additionally, β-Lap increased the mRNA levels of A<sub>1</sub>R. Taken together, these results suggest that β-Lap exerts hypnotic effects, potentially through A<sub>1</sub>R.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"531-539"},"PeriodicalIF":3.0,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11392670/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142008227","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Glycogen Phosphorylase Inhibitor Promotes Hair Growth via Protecting from Oxidative-Stress and Regulating Glycogen Breakdown in Human Hair follicles. 糖原磷酸化酶抑制剂通过保护人体毛囊免受氧化应激和调节糖原分解促进毛发生长
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-09-01 Epub Date: 2024-08-06 DOI: 10.4062/biomolther.2024.098
Bomi Park, Daeun Kim, Hairu Zhao, SoonRe Kim, Byung Cheol Park, Sanghwa Lee, Yurim Lee, Hee Dong Park, Dongchul Lim, Sunyoung Ryu, Jae Sung Hwang

Hair growth cycles are mainly regulated by human dermal papilla cells (hDPCs) and human outer root sheath cells (hORSCs). Protecting hDPCs from excessive oxidative stress and hORSCs from glycogen phosphorylase (PYGL) is crucial to maintaining the hair growth phase, anagen. In this study, we developed a new PYGL inhibitor, Hydroxytrimethylpyridinyl Methylindolecarboxamide (HTPI) and assessed its potential to prevent hair loss. HTPI reduced oxidative damage, preventing cell death and restored decreased level of anagen marker ALP and its related genes induced by hydrogen peroxide in hDPCs. Moreover, HTPI inhibited glycogen degradation and induced cell survival under glucose starvation in hORSCs. In ex-vivo culture, HTPI significantly enhanced hair growth compared to the control with minoxidil showing comparable results. Overall, these findings suggest that HTPI has significant potential as a therapeutic agent for the prevention and treatment of hair loss.

头发生长周期主要由人类真皮乳头细胞(hDPCs)和人类外根鞘细胞(hORSCs)调节。保护 hDPC 免受过度氧化应激,保护 hORSC 免受糖原磷酸化酶(PYGL)的侵害,对于维持头发生长期(生长期)至关重要。在这项研究中,我们开发了一种新的PYGL抑制剂--羟基三甲基吡啶甲基吲哚甲酰胺(HTPI),并评估了其预防脱发的潜力。HTPI 减少了氧化损伤,防止了细胞死亡,并恢复了过氧化氢诱导的脱发标志物 ALP 及其相关基因水平的下降。此外,HTPI 还能抑制糖原降解,并在葡萄糖饥饿条件下诱导 hORSCs 细胞存活。在体外培养中,与米诺西地相比,HTPI能显著促进头发生长。总之,这些研究结果表明,HTPI 在预防和治疗脱发方面具有巨大的治疗潜力。
{"title":"Glycogen Phosphorylase Inhibitor Promotes Hair Growth via Protecting from Oxidative-Stress and Regulating Glycogen Breakdown in Human Hair follicles.","authors":"Bomi Park, Daeun Kim, Hairu Zhao, SoonRe Kim, Byung Cheol Park, Sanghwa Lee, Yurim Lee, Hee Dong Park, Dongchul Lim, Sunyoung Ryu, Jae Sung Hwang","doi":"10.4062/biomolther.2024.098","DOIUrl":"10.4062/biomolther.2024.098","url":null,"abstract":"<p><p>Hair growth cycles are mainly regulated by human dermal papilla cells (hDPCs) and human outer root sheath cells (hORSCs). Protecting hDPCs from excessive oxidative stress and hORSCs from glycogen phosphorylase (PYGL) is crucial to maintaining the hair growth phase, anagen. In this study, we developed a new PYGL inhibitor, Hydroxytrimethylpyridinyl Methylindolecarboxamide (HTPI) and assessed its potential to prevent hair loss. HTPI reduced oxidative damage, preventing cell death and restored decreased level of anagen marker ALP and its related genes induced by hydrogen peroxide in hDPCs. Moreover, HTPI inhibited glycogen degradation and induced cell survival under glucose starvation in hORSCs. In ex-vivo culture, HTPI significantly enhanced hair growth compared to the control with minoxidil showing comparable results. Overall, these findings suggest that HTPI has significant potential as a therapeutic agent for the prevention and treatment of hair loss.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"640-646"},"PeriodicalIF":3.0,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11392663/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892804","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Gilteritinib Reduces FLT3 Expression in Acute Myeloid Leukemia Cells. 吉利替尼能减少急性髓性白血病细胞中的 FLT3 表达
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-09-01 Epub Date: 2024-08-02 DOI: 10.4062/biomolther.2023.215
Thị Lam Thái, Sun-Young Han

Acute myeloid leukemia (AML) is a genetically diverse and challenging malignancy, with mutations in the FLT3 gene being particularly common and deleterious. Gilteritinib, a potent FLT3 inhibitor, has been approved by the U.S. Food and Drug Administration (FDA) for the treatment of relapsed/refractory AML with FLT3 mutations. Although gilteritinib was developed based on its inhibitory activity against FLT3 kinase, it is important to understand the precise mechanisms of its antileukemic activity in managing drug resistance and discovering biomarkers. This study was designed to elucidate the effect of gilteritinib on the FLT3 expression level. The results showed that gilteritinib induced a dose-dependent decrease in both FLT3 phosphorylation and expression. This reduction was particularly pronounced after 48 h of treatment. The decrease in FLT3 expression was found to be independent of changes in FLT3 mRNA transcription, suggesting post-transcriptional regulatory mechanisms. Further studies were performed in various AML cell lines and cells with both FLT3 wild-type and FLT3 mutant exhibited FLT3 reduction by gilteritinib treatment. In addition, other FLT3 inhibitors were evaluated for their ability to reduce FLT3 expression. Other FLT3 inhibitors, midostaurin, crenolanib, and quizartinib, also reduced FLT3 expression, consistent with the effect of gilteritinib. These findings hold great promise for optimizing gilteritinib treatment in AML patients. However, it is important to recognize that further research is warranted to gain a full understanding of these mechanisms and their clinical implications in the context of FLT3 reduction.

急性髓性白血病(AML)是一种基因多样且极具挑战性的恶性肿瘤,其中FLT3基因突变尤为常见且具有毒性。Gilteritinib 是一种强效的 FLT3 抑制剂,已被美国食品药品管理局(FDA)批准用于治疗 FLT3 基因突变的复发/难治性急性髓性白血病。虽然吉特替尼是基于其对FLT3激酶的抑制活性而开发的,但了解其抗白血病活性的确切机制对于控制耐药性和发现生物标志物非常重要。本研究旨在阐明吉特替尼对FLT3表达水平的影响。结果显示,吉特替尼诱导了FLT3磷酸化和表达的剂量依赖性下降。这种降低在治疗48小时后尤为明显。研究发现,FLT3表达的降低与FLT3 mRNA转录的变化无关,这表明转录后调控机制的存在。在各种急性髓细胞系中进行了进一步研究,FLT3野生型和FLT3突变型细胞在吉特替尼治疗后都表现出FLT3降低。此外,还评估了其他 FLT3 抑制剂降低 FLT3 表达的能力。其他FLT3抑制剂、米多司他林、克瑞那尼和奎沙替尼也能降低FLT3的表达,与吉特替尼的效果一致。这些发现为优化吉特替尼治疗急性髓细胞性白血病患者带来了巨大希望。然而,重要的是要认识到,要全面了解这些机制及其在减少FLT3方面的临床意义,还需要进一步的研究。
{"title":"Gilteritinib Reduces FLT3 Expression in Acute Myeloid Leukemia Cells.","authors":"Thị Lam Thái, Sun-Young Han","doi":"10.4062/biomolther.2023.215","DOIUrl":"10.4062/biomolther.2023.215","url":null,"abstract":"<p><p>Acute myeloid leukemia (AML) is a genetically diverse and challenging malignancy, with mutations in the FLT3 gene being particularly common and deleterious. Gilteritinib, a potent FLT3 inhibitor, has been approved by the U.S. Food and Drug Administration (FDA) for the treatment of relapsed/refractory AML with FLT3 mutations. Although gilteritinib was developed based on its inhibitory activity against FLT3 kinase, it is important to understand the precise mechanisms of its antileukemic activity in managing drug resistance and discovering biomarkers. This study was designed to elucidate the effect of gilteritinib on the FLT3 expression level. The results showed that gilteritinib induced a dose-dependent decrease in both FLT3 phosphorylation and expression. This reduction was particularly pronounced after 48 h of treatment. The decrease in FLT3 expression was found to be independent of changes in FLT3 mRNA transcription, suggesting post-transcriptional regulatory mechanisms. Further studies were performed in various AML cell lines and cells with both FLT3 wild-type and FLT3 mutant exhibited FLT3 reduction by gilteritinib treatment. In addition, other FLT3 inhibitors were evaluated for their ability to reduce FLT3 expression. Other FLT3 inhibitors, midostaurin, crenolanib, and quizartinib, also reduced FLT3 expression, consistent with the effect of gilteritinib. These findings hold great promise for optimizing gilteritinib treatment in AML patients. However, it is important to recognize that further research is warranted to gain a full understanding of these mechanisms and their clinical implications in the context of FLT3 reduction.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"577-581"},"PeriodicalIF":3.0,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11392668/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141874070","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Domperidone, a Dopamine Receptor D2 Antagonist, Induces Apoptosis by Inhibiting the ERK/STAT3-Mediated Pathway in Human Colon Cancer HCT116 Cells. 多巴胺受体 D2 拮抗剂多潘立酮通过抑制 ERK/STAT3 介导的途径诱导人结肠癌 HCT116 细胞凋亡
IF 3 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2024-09-01 Epub Date: 2024-06-25 DOI: 10.4062/biomolther.2024.048
So Jin Sim, Jeong-Hoon Jang, Joon-Seok Choi, Kyung-Soo Chun

Colorectal cancer (CRC) continues to demonstrate high incidence and mortality rates, emphasizing that implementing strategic measures for prevention and treatment is crucial. Recently, the dopamine receptor D2 (DRD2), a G protein-coupled receptor, has been reported to play multiple roles in growth of tumor cells. This study investigated the anticancer potential of domperidone, a dopamine receptor D2 antagonist, in HCT116 human CRC cells. Domperidone demonstrated concentration- and time-dependent reductions in cell viability, thereby inducing apoptosis. The molecular mechanism revealed that domperidone modulated the mitochondrial pathway, decreasing mitochondrial Bcl-2 levels, elevating cytosolic cytochrome C expression, and triggering caspase- 3, -7, and -9 cleavage. Domperidone decreased in formation of β-arrestin2/MEK complex, which contributing to inhibition of ERK activation. Additionally, treatment with domperidone diminished JAK2 and STAT3 activation. Treatment of U0126, the MEK inhibitor, resulted in reduced phosphorylation of MEK, ERK, and STAT3 without alteration of JAK2 activation, indicating that domperidone targeted both MEK-ERK-STAT3 and JAK2-STAT3 signaling pathways, respectively. Immunoblot analysis revealed that domperidone also downregulated DRD2 expression. Domperidone-induced reactive oxygen species (ROS) generation and N-acetylcysteine treatment mitigated ROS levels and restored cell viability. An in vivo xenograft study verified the significant antitumor effects of domperidone. These results emphasize the multifaceted anticancer effects of domperidone, highlighting its potential as a promising therapeutic agent for human CRC.

结肠直肠癌(CRC)的发病率和死亡率居高不下,因此实施战略性预防和治疗措施至关重要。最近有报道称,多巴胺受体 D2(DRD2)是一种 G 蛋白偶联受体,在肿瘤细胞生长过程中发挥多种作用。本研究探讨了多巴胺受体D2拮抗剂多潘立酮在HCT116人CRC细胞中的抗癌潜力。多潘立酮可在浓度和时间上降低细胞活力,从而诱导细胞凋亡。分子机制显示,多潘立酮调节了线粒体通路,降低了线粒体Bcl-2水平,提高了细胞膜细胞色素C的表达,并引发了caspase-3、-7和-9的裂解。多潘立酮减少了β-arrestin2/MEK复合物的形成,从而抑制了ERK的激活。此外,多潘立酮还能减少 JAK2 和 STAT3 的活化。用MEK抑制剂U0126处理可减少MEK、ERK和STAT3的磷酸化,而不改变JAK2的活化,这表明多潘立酮分别针对MEK-ERK-STAT3和JAK2-STAT3信号通路。免疫印迹分析显示,多潘立酮还下调了DRD2的表达。多潘立酮诱导了活性氧(ROS)的产生,而N-乙酰半胱氨酸治疗可降低ROS水平并恢复细胞活力。一项体内异种移植研究验证了多潘立酮的显著抗肿瘤作用。这些结果强调了多潘立酮的多方面抗癌作用,凸显了它作为人类 CRC 治疗药物的潜力。
{"title":"Domperidone, a Dopamine Receptor D2 Antagonist, Induces Apoptosis by Inhibiting the ERK/STAT3-Mediated Pathway in Human Colon Cancer HCT116 Cells.","authors":"So Jin Sim, Jeong-Hoon Jang, Joon-Seok Choi, Kyung-Soo Chun","doi":"10.4062/biomolther.2024.048","DOIUrl":"10.4062/biomolther.2024.048","url":null,"abstract":"<p><p>Colorectal cancer (CRC) continues to demonstrate high incidence and mortality rates, emphasizing that implementing strategic measures for prevention and treatment is crucial. Recently, the dopamine receptor D2 (DRD2), a G protein-coupled receptor, has been reported to play multiple roles in growth of tumor cells. This study investigated the anticancer potential of domperidone, a dopamine receptor D2 antagonist, in HCT116 human CRC cells. Domperidone demonstrated concentration- and time-dependent reductions in cell viability, thereby inducing apoptosis. The molecular mechanism revealed that domperidone modulated the mitochondrial pathway, decreasing mitochondrial Bcl-2 levels, elevating cytosolic cytochrome C expression, and triggering caspase- 3, -7, and -9 cleavage. Domperidone decreased in formation of β-arrestin2/MEK complex, which contributing to inhibition of ERK activation. Additionally, treatment with domperidone diminished JAK2 and STAT3 activation. Treatment of U0126, the MEK inhibitor, resulted in reduced phosphorylation of MEK, ERK, and STAT3 without alteration of JAK2 activation, indicating that domperidone targeted both MEK-ERK-STAT3 and JAK2-STAT3 signaling pathways, respectively. Immunoblot analysis revealed that domperidone also downregulated DRD2 expression. Domperidone-induced reactive oxygen species (ROS) generation and <i>N</i>-acetylcysteine treatment mitigated ROS levels and restored cell viability. An <i>in vivo</i> xenograft study verified the significant antitumor effects of domperidone. These results emphasize the multifaceted anticancer effects of domperidone, highlighting its potential as a promising therapeutic agent for human CRC.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"568-576"},"PeriodicalIF":3.0,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11392662/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141445357","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","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
Biomolecules & Therapeutics
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