Pub Date : 2025-09-01Epub Date: 2025-08-19DOI: 10.4062/biomolther.2025.081
Hye Won Lee, Yun Jin Ju, Seeun Choi, Kiyon Rhew, Samantha Serafin Sevilleno, Min Sik Choi
Atopic dermatitis (AD) is a chronic relapsing inflammatory skin disorder characterized by pruritus, skin barrier dysfunction, and immune dysregulation. It significantly impacts the quality of life and increases the risk of infections, sleep disturbances, and psychological distress. AD pathogenesis involves genetic predisposition, environmental triggers, microbiome alterations, and immune dysfunction. Traditional treatments such as topical corticosteroids, calcineurin inhibitors, and systemic immunosuppressants provide symptomatic relief but often fail to provide long-term disease control. The emergence of targeted biologics and Janus kinase inhibitors has transformed AD management by offering more precise and effective therapeutic options. However, treatment responses vary, highlighting the need for biomarker-driven personalized therapies. In this review, we explore the evolving therapeutic landscape of AD, emphasizing the emerging role of biomarker-guided treatment strategies. We highlight recent discoveries of therapeutic (OX40, IgE, IL-5, IL-31, IL-22, thymic stromal lymphopoietin) and diagnostic (TARC/CCL17, MDC/CCL2, filaggrin, sphingosine-1-phosphate, CXCL2) biomarkers that offer promising avenues for patient stratification and treatment monitoring. This review offers novel insight into how the convergence of biomarker research and therapeutic innovation can address current gaps in AD care. Future research should focus on refining biomarker-guided treatment strategies, optimizing therapeutic combinations, and addressing unmet patient needs. The integration of biomarker-guided strategies into routine clinical practice represents a critical step toward long-term disease control and improved quality of life for AD patients.
{"title":"Atopic Dermatitis Management: from Conventional Therapies to Biomarker-Driven Treatment Approaches.","authors":"Hye Won Lee, Yun Jin Ju, Seeun Choi, Kiyon Rhew, Samantha Serafin Sevilleno, Min Sik Choi","doi":"10.4062/biomolther.2025.081","DOIUrl":"10.4062/biomolther.2025.081","url":null,"abstract":"<p><p>Atopic dermatitis (AD) is a chronic relapsing inflammatory skin disorder characterized by pruritus, skin barrier dysfunction, and immune dysregulation. It significantly impacts the quality of life and increases the risk of infections, sleep disturbances, and psychological distress. AD pathogenesis involves genetic predisposition, environmental triggers, microbiome alterations, and immune dysfunction. Traditional treatments such as topical corticosteroids, calcineurin inhibitors, and systemic immunosuppressants provide symptomatic relief but often fail to provide long-term disease control. The emergence of targeted biologics and Janus kinase inhibitors has transformed AD management by offering more precise and effective therapeutic options. However, treatment responses vary, highlighting the need for biomarker-driven personalized therapies. In this review, we explore the evolving therapeutic landscape of AD, emphasizing the emerging role of biomarker-guided treatment strategies. We highlight recent discoveries of therapeutic (OX40, IgE, IL-5, IL-31, IL-22, thymic stromal lymphopoietin) and diagnostic (TARC/CCL17, MDC/CCL2, filaggrin, sphingosine-1-phosphate, CXCL2) biomarkers that offer promising avenues for patient stratification and treatment monitoring. This review offers novel insight into how the convergence of biomarker research and therapeutic innovation can address current gaps in AD care. Future research should focus on refining biomarker-guided treatment strategies, optimizing therapeutic combinations, and addressing unmet patient needs. The integration of biomarker-guided strategies into routine clinical practice represents a critical step toward long-term disease control and improved quality of life for AD patients.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"813-829"},"PeriodicalIF":3.2,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12408207/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144871264","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}
Pub Date : 2025-09-01Epub Date: 2025-08-06DOI: 10.4062/biomolther.2025.062
Mücahit Varlı, Eun-Young Lee, So-Yeon Park, Yi Yang, Prima F Hillman, Rui Zhou, Jae-Seoun Hur, Sang-Jip Nam, Hangun Kim
Endolichenic fungi (ELF), symbionts of lichens, have been reported to produce diverse bioactive secondary metabolites with promising pharmaceutical potential. In this study, we isolated and identified an ELF, EL001668 (KACC 83020BP), from Cetraria laevigata Rass., and assessed its crude extract and bioactive compounds against colorectal cancer (CRC) stem cell activity. cis-10-nonadecenoic acid (c-NDA), isolated through bioactivity-guided fractionation exerted substantial inhibitory effects on CRC stemness, such as the suppression of spheroid formation and the downregulation of the key stem cell markers ALDH1, CD44, and CD133. Comparative analysis with the omega-3 fatty acids EPA and DHA, with well-established properties, showed that c- NDA exerted comparable or superior inhibitory effects against the markers and phenotypic traits of stemness. Besides, the crude extract of EL001668 exhibited greater suppression of certain markers in comparison to the individual compounds. These findings suggest that c-NDA, in conjunction with ELF-derived compounds, holds potential as a novel therapeutic candidate targeting CRC stem cells. Taken together, the current study demonstrated that c-NDA, similar to EPA and DHA, may possess adjunct or complementary effects in cancer treatment and other diseases.
{"title":"An Endolichenic Fungi-Derived Fatty Acid, cis-10-Nonadecenoic acid, Suppresses Colorectal Cancer Stemness.","authors":"Mücahit Varlı, Eun-Young Lee, So-Yeon Park, Yi Yang, Prima F Hillman, Rui Zhou, Jae-Seoun Hur, Sang-Jip Nam, Hangun Kim","doi":"10.4062/biomolther.2025.062","DOIUrl":"10.4062/biomolther.2025.062","url":null,"abstract":"<p><p>Endolichenic fungi (ELF), symbionts of lichens, have been reported to produce diverse bioactive secondary metabolites with promising pharmaceutical potential. In this study, we isolated and identified an ELF, EL001668 (KACC 83020BP), from <i>Cetraria laevigata</i> Rass., and assessed its crude extract and bioactive compounds against colorectal cancer (CRC) stem cell activity. cis-10-nonadecenoic acid (c-NDA), isolated through bioactivity-guided fractionation exerted substantial inhibitory effects on CRC stemness, such as the suppression of spheroid formation and the downregulation of the key stem cell markers ALDH1, CD44, and CD133. Comparative analysis with the omega-3 fatty acids EPA and DHA, with well-established properties, showed that c- NDA exerted comparable or superior inhibitory effects against the markers and phenotypic traits of stemness. Besides, the crude extract of EL001668 exhibited greater suppression of certain markers in comparison to the individual compounds. These findings suggest that c-NDA, in conjunction with ELF-derived compounds, holds potential as a novel therapeutic candidate targeting CRC stem cells. Taken together, the current study demonstrated that c-NDA, similar to EPA and DHA, may possess adjunct or complementary effects in cancer treatment and other diseases.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"842-851"},"PeriodicalIF":3.2,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12408206/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144788178","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}
Pub Date : 2025-09-01Epub Date: 2025-08-31DOI: 10.4062/biomolther.2025.056
Jaebeom Cho
Tumor dormancy represents a clinically significant but poorly understood state in which disseminated cancer cells persist in a quiescent, non-proliferative state, evading conventional therapies and driving late relapse. This review summarizes recent advancements in experimental models-both in vitro and in vivo-that recapitulate the full spectrum of dormancy, including its induction, maintenance, and reactivation. Crucial intrinsic pathways such as ERK/p38 signaling shifts, epigenetic remodeling, and metabolic adaptations and microenvironmental and immune-mediated cues that regulate dormant cell fate are discussed. Therapeutic strategies aimed at maintaining dormancy, reactivating dormant cells for elimination, or directly targeting their survival pathways have been highlighted. By integrating insights from model systems, molecular regulation, and therapy, this review aims to provide a comprehensive framework that informs future efforts to target dormant cancer cells and ultimately reduce recurrence and improve patient outcomes.
{"title":"Understanding Tumor Dormancy: from Experimental Models to Mechanisms and Therapeutic Strategies.","authors":"Jaebeom Cho","doi":"10.4062/biomolther.2025.056","DOIUrl":"10.4062/biomolther.2025.056","url":null,"abstract":"<p><p>Tumor dormancy represents a clinically significant but poorly understood state in which disseminated cancer cells persist in a quiescent, non-proliferative state, evading conventional therapies and driving late relapse. This review summarizes recent advancements in experimental models-both <i>in vitro</i> and <i>in vivo</i>-that recapitulate the full spectrum of dormancy, including its induction, maintenance, and reactivation. Crucial intrinsic pathways such as ERK/p38 signaling shifts, epigenetic remodeling, and metabolic adaptations and microenvironmental and immune-mediated cues that regulate dormant cell fate are discussed. Therapeutic strategies aimed at maintaining dormancy, reactivating dormant cells for elimination, or directly targeting their survival pathways have been highlighted. By integrating insights from model systems, molecular regulation, and therapy, this review aims to provide a comprehensive framework that informs future efforts to target dormant cancer cells and ultimately reduce recurrence and improve patient outcomes.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":"33 5","pages":"770-784"},"PeriodicalIF":3.2,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12408196/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144941462","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}
Pub Date : 2025-09-01Epub Date: 2025-08-13DOI: 10.4062/biomolther.2025.071
Yoon Tae Kim, Geunyeol Jeong, Yihyo Kim, HyeonJu Roh, Susung Lim, Jinah Jang, Yun Hui Choi, Kyoung Chul Park, Mirim Jin
Sepsis is a leading cause of mortality in hospitals with a lack of reliable biomarkers and specialized therapeutics. Recently, highly secreted tryptophanyl-tRNA synthetase 1 (WARS1), an endogenous ligand for Toll-like receptor (TLR) 2 and TLR4, was found to be a potential theranostic target for hypercytokinemic severe sepsis. In this study, using the minipig sepsis model inoculated with cecum slurry, we demonstrated that increases in WARS1 levels were associated with severity of sepsis and showed strong correlations with RBC count and the levels of HGB, HCT, EPO, lactate, and PLT count in the acute phase of sepsis. Further, administration of the WARS1 neutralizing antibody to the septic minipigs inhibited the increase in the overall SOFA score with a significantly lower P/F ratio, which was accompanied by the suppression of proinflammatory cytokine and chemokine expressions as well as EPO production, a decrease in AST and ALT levels, and inflammatory immune cell infiltration in the lung. Taken together, these findings provide a novel insight into the pathophysiology of acute phase of sepsis and suggest the clinical application of WARS1 neutralizing therapeutics in the treatment of sepsis.
{"title":"Control of Overly Secreted Tryptophanyl tRNA Synthetase Attenuates Sepsis Severity in a Porcine Model.","authors":"Yoon Tae Kim, Geunyeol Jeong, Yihyo Kim, HyeonJu Roh, Susung Lim, Jinah Jang, Yun Hui Choi, Kyoung Chul Park, Mirim Jin","doi":"10.4062/biomolther.2025.071","DOIUrl":"10.4062/biomolther.2025.071","url":null,"abstract":"<p><p>Sepsis is a leading cause of mortality in hospitals with a lack of reliable biomarkers and specialized therapeutics. Recently, highly secreted tryptophanyl-tRNA synthetase 1 (WARS1), an endogenous ligand for Toll-like receptor (TLR) 2 and TLR4, was found to be a potential theranostic target for hypercytokinemic severe sepsis. In this study, using the minipig sepsis model inoculated with cecum slurry, we demonstrated that increases in WARS1 levels were associated with severity of sepsis and showed strong correlations with RBC count and the levels of HGB, HCT, EPO, lactate, and PLT count in the acute phase of sepsis. Further, administration of the WARS1 neutralizing antibody to the septic minipigs inhibited the increase in the overall SOFA score with a significantly lower P/F ratio, which was accompanied by the suppression of proinflammatory cytokine and chemokine expressions as well as EPO production, a decrease in AST and ALT levels, and inflammatory immune cell infiltration in the lung. Taken together, these findings provide a novel insight into the pathophysiology of acute phase of sepsis and suggest the clinical application of WARS1 neutralizing therapeutics in the treatment of sepsis.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"852-865"},"PeriodicalIF":3.2,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12408201/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144833846","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}
The accumulation of free fatty acids (FFAs) in hepatocytes is a key characteristic of metabolic dysfunction-associated steatotic liver disease (MASLD), which leads to lipid peroxidation and ultimately results in ferroptosis. Currently, there is an absence of efficacious therapeutic options available for the management of MASLD. Consequently, an in-depth exploration of the roles of FFAs and ferroptosis in the progression of MASLD may reveal hitherto unidentified therapeutic targets. In the study, we established an early lesion model of MASLD, namely NAFL, and comprehensive analyses of lipid metabolism, hepatocellular injury, iron homeostasis, and ferroptosis were performed. The HFD and FFAs treatment significantly elevated the expression of enzymes associated with lipid synthesis, including ACC1 and FASN, leading to enhanced lipid accumulation in hepatocytes. Additionally, HFD and FFAs resulted in increased iron loading and a reduction in the levels of the antioxidant enzyme GPX4, which ultimately triggers ferroptosis. In contrast, the administration of melatonin effectively inhibited the activity of lipid synthesis-related enzymes, decreased hepatic lipid deposition, alleviated free fatty acid-induced iron dysregulation, and mitigated liver damage. Mechanistically, melatonin has been shown to attenuate hepatocyte ferroptosis by modulating the KEAP1/NRF2/HO-1 pathway, which in turn diminishes free fatty acids-induced oxidative stress. In conclusion, melatonin alleviates MASLD progression by curbing FFAs-induced oxidative stress and ferroptosis. These findings provide valuable insights into the mechanisms underlying MASLD progression and highlight melatonin as a potential therapeutic agent for the management of MASLD.
{"title":"Melatonin Prevents the Progression of MASLD via Inhibiting FFAs-Induced Ferroptosis through KEAP1/NRF2/HO-1 Pathway.","authors":"Shuojiao Li, Peng Rao, Wenxian Yu, Yue Tang, Xuanpeng Jiang, Jiatao Liu","doi":"10.4062/biomolther.2025.037","DOIUrl":"10.4062/biomolther.2025.037","url":null,"abstract":"<p><p>The accumulation of free fatty acids (FFAs) in hepatocytes is a key characteristic of metabolic dysfunction-associated steatotic liver disease (MASLD), which leads to lipid peroxidation and ultimately results in ferroptosis. Currently, there is an absence of efficacious therapeutic options available for the management of MASLD. Consequently, an in-depth exploration of the roles of FFAs and ferroptosis in the progression of MASLD may reveal hitherto unidentified therapeutic targets. In the study, we established an early lesion model of MASLD, namely NAFL, and comprehensive analyses of lipid metabolism, hepatocellular injury, iron homeostasis, and ferroptosis were performed. The HFD and FFAs treatment significantly elevated the expression of enzymes associated with lipid synthesis, including ACC1 and FASN, leading to enhanced lipid accumulation in hepatocytes. Additionally, HFD and FFAs resulted in increased iron loading and a reduction in the levels of the antioxidant enzyme GPX4, which ultimately triggers ferroptosis. In contrast, the administration of melatonin effectively inhibited the activity of lipid synthesis-related enzymes, decreased hepatic lipid deposition, alleviated free fatty acid-induced iron dysregulation, and mitigated liver damage. Mechanistically, melatonin has been shown to attenuate hepatocyte ferroptosis by modulating the KEAP1/NRF2/HO-1 pathway, which in turn diminishes free fatty acids-induced oxidative stress. In conclusion, melatonin alleviates MASLD progression by curbing FFAs-induced oxidative stress and ferroptosis. These findings provide valuable insights into the mechanisms underlying MASLD progression and highlight melatonin as a potential therapeutic agent for the management of MASLD.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"876-889"},"PeriodicalIF":3.2,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12408204/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144871265","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}
Pub Date : 2025-09-01Epub Date: 2025-08-12DOI: 10.4062/biomolther.2024.211
Young-Sool Hah, Sun-Young Han
Small molecules that induce protein polymerization represent an emerging class of compounds with diverse therapeutic potential. This review provides a comprehensive overview of five such molecules: arsenic trioxide (As2O3), BI-3802, NVS-STG2, paclitaxel, and verteporfin. These compounds target different proteins (PML-RARα, BCL6, STING, β-tubulin, and p62, respectively) and exhibit varied mechanisms of action. Some, like As2O3 and BI-3802, induce polymerization leading to protein degradation, while others, such as NVS-STG2, activate protein function through polymerization. Paclitaxel, distinct from these, induces the stabilization of tubulin polymers. Verteporfin, on the other hand, uniquely causes covalent cross-linking of its target and other cellular proteins. This review explores the molecular mechanisms, structural insights, and therapeutic implications of these compounds, highlighting their potential in targeted protein degradation, cancer treatment, and modulation of cellular processes, such as autophagy and immune response. The diverse effects of these molecules underscore the complexity of protein polymerization in cellular function and disease, opening new avenues for drug discovery and development.
{"title":"Small-Molecule-Induced Protein Polymerization: Mechanisms and Therapeutic Implications.","authors":"Young-Sool Hah, Sun-Young Han","doi":"10.4062/biomolther.2024.211","DOIUrl":"10.4062/biomolther.2024.211","url":null,"abstract":"<p><p>Small molecules that induce protein polymerization represent an emerging class of compounds with diverse therapeutic potential. This review provides a comprehensive overview of five such molecules: arsenic trioxide (As<sub>2</sub>O<sub>3</sub>), BI-3802, NVS-STG2, paclitaxel, and verteporfin. These compounds target different proteins (PML-RARα, BCL6, STING, β-tubulin, and p62, respectively) and exhibit varied mechanisms of action. Some, like As<sub>2</sub>O<sub>3</sub> and BI-3802, induce polymerization leading to protein degradation, while others, such as NVS-STG2, activate protein function through polymerization. Paclitaxel, distinct from these, induces the stabilization of tubulin polymers. Verteporfin, on the other hand, uniquely causes covalent cross-linking of its target and other cellular proteins. This review explores the molecular mechanisms, structural insights, and therapeutic implications of these compounds, highlighting their potential in targeted protein degradation, cancer treatment, and modulation of cellular processes, such as autophagy and immune response. The diverse effects of these molecules underscore the complexity of protein polymerization in cellular function and disease, opening new avenues for drug discovery and development.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"804-812"},"PeriodicalIF":3.2,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12408205/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144820484","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}
In this study, we explored the effects of taxifolin, a flavonoid compound, on the expression of the MUC5AC mucin gene in airway epithelial cells. Human pulmonary epithelial NCI-H292 cells were pretreated with taxifolin for 30 min prior to stimulation with phorbol 12-myristate 13-acetate (PMA) for 24 h. We also investigated the influence of taxifolin on the PMA-induced activation of the NF-κB signaling pathway. Our results demonstrated that taxifolin inhibited both glycoprotein production and MUC5AC mRNA expression triggered by PMA. This inhibition occurred through the prevention of IκBα degradation and the nuclear translocation of NF-κB p65. These findings suggest that taxifolin suppresses mucin gene expression by modulating the NF-κB signaling pathway in human pulmonary epithelial cells.
{"title":"Taxifolin Regulates the Gene Expression of MUC5AC Mucin via Affecting IκBα/NF-κB p65 Signaling in Human Airway Epithelial Cells.","authors":"Rajib Hossain, Md Solayman Hossain, Hyun Jae Lee, Choong Jae Lee","doi":"10.4062/biomolther.2025.101","DOIUrl":"10.4062/biomolther.2025.101","url":null,"abstract":"<p><p>In this study, we explored the effects of taxifolin, a flavonoid compound, on the expression of the MUC5AC mucin gene in airway epithelial cells. Human pulmonary epithelial NCI-H292 cells were pretreated with taxifolin for 30 min prior to stimulation with phorbol 12-myristate 13-acetate (PMA) for 24 h. We also investigated the influence of taxifolin on the PMA-induced activation of the NF-κB signaling pathway. Our results demonstrated that taxifolin inhibited both glycoprotein production and MUC5AC mRNA expression triggered by PMA. This inhibition occurred through the prevention of IκBα degradation and the nuclear translocation of NF-κB p65. These findings suggest that taxifolin suppresses mucin gene expression by modulating the NF-κB signaling pathway in human pulmonary epithelial cells.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"901-906"},"PeriodicalIF":3.2,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12408199/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144871266","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}
Pub Date : 2025-07-01Epub Date: 2025-06-19DOI: 10.4062/biomolther.2025.022
Betül Ertural, Büşra Nur Çiçek, Işıl Aksan Kurnaz
RNA therapeutics represent a disruptive technology that has transformed drug discovery and manufacturing, gaining significant prominence during the COVID-19 pandemic. RNA therapeutics encompass diverse molecules like antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), microRNAs (miRNAs), RNA aptamers, and messenger RNAs (mRNAs), which can function through different mechanisms. RNA therapeutics are increasingly used to treat various diseases, including neurological disorders. For example, ASO therapies such as nusinersen for spinal muscular atrophy and eteplirsen for Duchenne muscular dystrophy are successful applications of RNA-based treatment. Emerging ASO treatments for Huntington's disease and amyotrophic lateral sclerosis are also promising, with ongoing clinical trials demonstrating significant reductions in disease-associated proteins. Still, delivery of these molecules remains a pivotal challenge in RNA therapeutics, especially for ASOs in penetrating the blood-brain barrier to target neurological disorders effectively. Nanoparticle-based formulations have emerged as leading strategies to enhance RNA stability, reduce immunogenicity, and improve cellular uptake. Despite these advances, significant hurdles remain, including optimizing pharmacokinetics, minimizing off-target effects, and ensuring sustained therapeutic efficacy. Regulatory frameworks are evolving to accommodate the unique challenges of RNA-based therapies, including ASOs with efforts underway to establish comprehensive guidelines for RNA therapeutics, yet there are also sustainable manufacturing issues that need to be considered for long-term feasibility. By addressing these challenges, RNA therapeutics hold immense potential to revolutionize treatment paradigms for neurological disorders. Looking forward, the future of RNA therapeutics in neurology appears promising but requires continued interdisciplinary collaboration and technological innovation.
{"title":"RNA Therapeutics: Focus on Antisense Oligonucleotides in the Nervous System.","authors":"Betül Ertural, Büşra Nur Çiçek, Işıl Aksan Kurnaz","doi":"10.4062/biomolther.2025.022","DOIUrl":"10.4062/biomolther.2025.022","url":null,"abstract":"<p><p>RNA therapeutics represent a disruptive technology that has transformed drug discovery and manufacturing, gaining significant prominence during the COVID-19 pandemic. RNA therapeutics encompass diverse molecules like antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), microRNAs (miRNAs), RNA aptamers, and messenger RNAs (mRNAs), which can function through different mechanisms. RNA therapeutics are increasingly used to treat various diseases, including neurological disorders. For example, ASO therapies such as nusinersen for spinal muscular atrophy and eteplirsen for Duchenne muscular dystrophy are successful applications of RNA-based treatment. Emerging ASO treatments for Huntington's disease and amyotrophic lateral sclerosis are also promising, with ongoing clinical trials demonstrating significant reductions in disease-associated proteins. Still, delivery of these molecules remains a pivotal challenge in RNA therapeutics, especially for ASOs in penetrating the blood-brain barrier to target neurological disorders effectively. Nanoparticle-based formulations have emerged as leading strategies to enhance RNA stability, reduce immunogenicity, and improve cellular uptake. Despite these advances, significant hurdles remain, including optimizing pharmacokinetics, minimizing off-target effects, and ensuring sustained therapeutic efficacy. Regulatory frameworks are evolving to accommodate the unique challenges of RNA-based therapies, including ASOs with efforts underway to establish comprehensive guidelines for RNA therapeutics, yet there are also sustainable manufacturing issues that need to be considered for long-term feasibility. By addressing these challenges, RNA therapeutics hold immense potential to revolutionize treatment paradigms for neurological disorders. Looking forward, the future of RNA therapeutics in neurology appears promising but requires continued interdisciplinary collaboration and technological innovation.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"572-581"},"PeriodicalIF":3.0,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12215037/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144324438","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}
Pub Date : 2025-07-01Epub Date: 2025-06-10DOI: 10.4062/biomolther.2024.167
Oh Seong Kwon, Kyu-Taek Hwang, Won Seok Choi, Ji-Yun Lee
Asthma is an allergic inflammatory disease of the lungs characterized by eosinophilic inflammation, mucus hypersecretion, and airway hyperresponsiveness (AHR). Exposure to environmental endotoxins, such as lipopolysaccharide (LPS), can exacerbate asthma severity. Phosphodiesterase (PDE) inactivates cyclic adenosine 3',5'-monophosphate and cyclic guanosine 3',5'-monophosphate, thereby aggravating inflammation. Accordingly, PDE inhibitors could be used to treat asthma. Herein, we studied the effects of BRL-50481 (BRL), a PDE7 inhibitor, in a murine model of ovalbumin (OVA)-induced allergic asthma with co-exposure to LPS. Mice were sensitized, challenged with OVA, and subsequently exposed to LPS. Mice were administered with BRL prior to the OVA challenge. We observed that BRL treatment could suppress hallmark features of asthma, including mediators of eosinophilic and neutrophilic inflammation, such as expression of antigen-specific immunoglobulin (Ig) E, interleukin (IL)-13, IL-6, and mucus hypersecretion. Mice co-exposed to OVA and LPS exhibited marked AHR, which was improved in BRL-treated mice because of inhibition of mucus overproduction. In conclusion, given that PDE7 inhibitors can regulate allergic inflammatory responses, these agents could be potential candidates for treating asthma.
{"title":"BRL-50481 Ameliorates Lung Inflammation in a Murine Model of Ovalbumin-Induced Allergic Asthma with Co-Exposure to Lipopolysaccharide.","authors":"Oh Seong Kwon, Kyu-Taek Hwang, Won Seok Choi, Ji-Yun Lee","doi":"10.4062/biomolther.2024.167","DOIUrl":"10.4062/biomolther.2024.167","url":null,"abstract":"<p><p>Asthma is an allergic inflammatory disease of the lungs characterized by eosinophilic inflammation, mucus hypersecretion, and airway hyperresponsiveness (AHR). Exposure to environmental endotoxins, such as lipopolysaccharide (LPS), can exacerbate asthma severity. Phosphodiesterase (PDE) inactivates cyclic adenosine 3',5'-monophosphate and cyclic guanosine 3',5'-monophosphate, thereby aggravating inflammation. Accordingly, PDE inhibitors could be used to treat asthma. Herein, we studied the effects of BRL-50481 (BRL), a PDE7 inhibitor, in a murine model of ovalbumin (OVA)-induced allergic asthma with co-exposure to LPS. Mice were sensitized, challenged with OVA, and subsequently exposed to LPS. Mice were administered with BRL prior to the OVA challenge. We observed that BRL treatment could suppress hallmark features of asthma, including mediators of eosinophilic and neutrophilic inflammation, such as expression of antigen-specific immunoglobulin (Ig) E, interleukin (IL)-13, IL-6, and mucus hypersecretion. Mice co-exposed to OVA and LPS exhibited marked AHR, which was improved in BRL-treated mice because of inhibition of mucus overproduction. In conclusion, given that PDE7 inhibitors can regulate allergic inflammatory responses, these agents could be potential candidates for treating asthma.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"692-703"},"PeriodicalIF":3.0,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12215039/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144257303","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}
Pub Date : 2025-07-01Epub Date: 2025-06-23DOI: 10.4062/biomolther.2025.044
Mikyoung Kwon, Hye Jin Jung
Liver cancer stem cells (LCSCs) play a significant role in the development, metastasis, treatment resistance, and recurrence of hepatocellular carcinoma (HCC). Targeting LCSCs offers a novel strategy to overcome treatment resistance in HCC. Myricetin, a flavonol from the flavonoid family, is known for its diverse biological activities, including anticancer effects. However, its potential for eradicating LCSCs had not been thoroughly investigated prior to this study. This study evaluated the effects of myricetin on LCSCs derived from Huh7 and Hep3B cell lines both in vitro and in vivo. LCSCs were treated with myricetin to assess cell proliferation, cell cycle arrest, apoptosis induction, autophagy regulation, stemness and EMT marker expression, and tumor growth suppression using a chicken embryo CAM model. Additionally, the combination therapy of myricetin with chloroquine, an autophagy inhibitor, was explored. Myricetin significantly inhibited the proliferation of Huh7- and Hep3B-derived LCSCs and suppressed tumor growth in the CAM model. It induced cell cycle arrest at the G0/G1 phase and triggered apoptosis through intrinsic and extrinsic pathways. Myricetin also stimulated autophagy by inhibiting the PI3K/AKT/mTOR pathway, reduced the expression of stemness markers, including Sox2, Oct4, Nanog, and ALDH1A1, and suppressed EMT. Combining myricetin with chloroquine enhanced apoptotic effects and further downregulated stemness markers by inhibiting STAT3 activation, demonstrating greater efficacy than myricetin alone. The findings establish myricetin, either as a standalone treatment or in combination with chloroquine, as a promising therapeutic candidate for targeting LCSC growth and overcoming chemotherapy resistance in HCC.
肝癌干细胞(Liver cancer stem cells, LCSCs)在肝细胞癌(hepatellular carcinoma, HCC)的发生、转移、治疗抵抗和复发中起着重要作用。靶向LCSCs提供了一种克服HCC治疗耐药的新策略。杨梅素是类黄酮家族中的一种黄酮醇,因其多种生物活性而闻名,包括抗癌作用。然而,在这项研究之前,其根除LCSCs的潜力尚未得到彻底的调查。本研究在体外和体内研究了杨梅素对Huh7和Hep3B细胞系LCSCs的影响。采用鸡胚CAM模型,用杨梅素处理LCSCs,观察细胞增殖、细胞周期阻滞、细胞凋亡诱导、自噬调节、干细胞性和EMT标志物表达以及肿瘤生长抑制作用。此外,还探讨了杨梅素与自噬抑制剂氯喹的联合治疗。在CAM模型中,杨梅素显著抑制Huh7-和hep3b来源的LCSCs的增殖,抑制肿瘤生长。它诱导细胞周期阻滞于G0/G1期,并通过内源性和外源性途径引发细胞凋亡。杨梅素还通过抑制PI3K/AKT/mTOR通路刺激自噬,降低Sox2、Oct4、Nanog、ALDH1A1等干性标志物的表达,抑制EMT。杨梅素联合氯喹可增强细胞凋亡效应,并通过抑制STAT3激活进一步下调茎干标志物,其效果优于单用杨梅素。研究结果表明,杨梅素无论是单独治疗还是与氯喹联合治疗,都是靶向肝细胞癌生长和克服肝细胞癌化疗耐药的有希望的治疗候选药物。
{"title":"Anticancer Potential of Myricetin against Huh7- and Hep3B-Derived Liver Cancer Stem Cells through the Regulation of Apoptosis, Autophagy, and Stemness.","authors":"Mikyoung Kwon, Hye Jin Jung","doi":"10.4062/biomolther.2025.044","DOIUrl":"10.4062/biomolther.2025.044","url":null,"abstract":"<p><p>Liver cancer stem cells (LCSCs) play a significant role in the development, metastasis, treatment resistance, and recurrence of hepatocellular carcinoma (HCC). Targeting LCSCs offers a novel strategy to overcome treatment resistance in HCC. Myricetin, a flavonol from the flavonoid family, is known for its diverse biological activities, including anticancer effects. However, its potential for eradicating LCSCs had not been thoroughly investigated prior to this study. This study evaluated the effects of myricetin on LCSCs derived from Huh7 and Hep3B cell lines both <i>in vitro</i> and <i>in vivo</i>. LCSCs were treated with myricetin to assess cell proliferation, cell cycle arrest, apoptosis induction, autophagy regulation, stemness and EMT marker expression, and tumor growth suppression using a chicken embryo CAM model. Additionally, the combination therapy of myricetin with chloroquine, an autophagy inhibitor, was explored. Myricetin significantly inhibited the proliferation of Huh7- and Hep3B-derived LCSCs and suppressed tumor growth in the CAM model. It induced cell cycle arrest at the G0/G1 phase and triggered apoptosis through intrinsic and extrinsic pathways. Myricetin also stimulated autophagy by inhibiting the PI3K/AKT/mTOR pathway, reduced the expression of stemness markers, including Sox2, Oct4, Nanog, and ALDH1A1, and suppressed EMT. Combining myricetin with chloroquine enhanced apoptotic effects and further downregulated stemness markers by inhibiting STAT3 activation, demonstrating greater efficacy than myricetin alone. The findings establish myricetin, either as a standalone treatment or in combination with chloroquine, as a promising therapeutic candidate for targeting LCSC growth and overcoming chemotherapy resistance in HCC.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"636-651"},"PeriodicalIF":3.0,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12215034/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144367859","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}