Curzerene, a sesquiterpene compound isolated from Curcuma Radix, exhibits various therapeutic effects, such as anti-tumor and anti-hyperlipidemic properties. However, its neuroprotective effects have not yet been reported. This study focused on exploring the neuroprotective effect of curzerene and elucidate its potential mechanism by combining molecular biotechnology with multi-omics approaches. Curzerene was orally administered to LPS-induced depressive-like behaviors and cognitive impairment in mice for 14 days, and the related biochemical parameters were evaluated. The possible mechanisms were elucidated using qRT-PCR, Western Blot, immunofluorescence, untarget metabolomics, GC-MS and 16S rDNA comprehensively. Curzerene ameliorated depression symptoms and cognitive impairment by increasing the preference for sucrose in SPT and the central area and total distance traveled in OFT, reducing the immobility time in TST and FST, as well as rising the spontaneous alternation ratio in Y maze. Multiple molecular biology techniques analysis indicated the ameliorative effect of curzerene via HMGB1/RAGE/ TLR4 pathway. Moreover, curzerene primarily regulates purine metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, phenylalanine metabolism, pyrimidine metabolism, etc. Furthermore, intervention increased the relative abundance of Parabacteroides, Clostridia_UCG-014_unclassified, and Rhodospirillales_unclassified, and enhanced the production of SCFAs. This work demonstrated that curzerene effectively protects against LPS-induced neurological damage, potentially by inhibiting the HMGB1/ RAGE/TLR4 pathway through the restoration of gut microbiota homeostasis, modulation of metabolites, and enhancement of SCFAs. In conclusion, this study offers new perspectives on the therapeutic possibilities of curzerene in mitigating depressive-like behaviors and cognitive impairment.
{"title":"Curzerene Ameliorates Depression-Like Behaviors and Cognitive Impairment by Modulating the Gut Microbiota and HMGB1/RAGE/TLR4 Pathway.","authors":"Fengjing Huang, Xiaohong Ma, Xiao Xu, Jingwen Zhang, Chunlai Wang, Ruoxi Song, Xiangxiang Wang, Mingxing Chen, Yu Qi","doi":"10.4062/biomolther.2025.120","DOIUrl":"https://doi.org/10.4062/biomolther.2025.120","url":null,"abstract":"<p><p>Curzerene, a sesquiterpene compound isolated from Curcuma Radix, exhibits various therapeutic effects, such as anti-tumor and anti-hyperlipidemic properties. However, its neuroprotective effects have not yet been reported. This study focused on exploring the neuroprotective effect of curzerene and elucidate its potential mechanism by combining molecular biotechnology with multi-omics approaches. Curzerene was orally administered to LPS-induced depressive-like behaviors and cognitive impairment in mice for 14 days, and the related biochemical parameters were evaluated. The possible mechanisms were elucidated using qRT-PCR, Western Blot, immunofluorescence, untarget metabolomics, GC-MS and 16S rDNA comprehensively. Curzerene ameliorated depression symptoms and cognitive impairment by increasing the preference for sucrose in SPT and the central area and total distance traveled in OFT, reducing the immobility time in TST and FST, as well as rising the spontaneous alternation ratio in Y maze. Multiple molecular biology techniques analysis indicated the ameliorative effect of curzerene <i>via</i> HMGB1/RAGE/ TLR4 pathway. Moreover, curzerene primarily regulates purine metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, phenylalanine metabolism, pyrimidine metabolism, etc. Furthermore, intervention increased the relative abundance of <i>Parabacteroides</i>, <i>Clostridia_UCG-014_unclassified</i>, and <i>Rhodospirillales_unclassified</i>, and enhanced the production of SCFAs. This work demonstrated that curzerene effectively protects against LPS-induced neurological damage, potentially by inhibiting the HMGB1/ RAGE/TLR4 pathway through the restoration of gut microbiota homeostasis, modulation of metabolites, and enhancement of SCFAs. In conclusion, this study offers new perspectives on the therapeutic possibilities of curzerene in mitigating depressive-like behaviors and cognitive impairment.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145720760","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-11DOI: 10.4062/biomolther.2025.125
Qiang Deng, Xiaoping Men, Duo Jin, Yuzhuo Bai
Thyroid cancer is one of the most prevalent cancers in the world, accounting for the increased sensitivity of diagnostic assessments, the environment, and extensive imaging. While open thyroidectomy still forms the base surgical treatment of differentiated thyroid carcinoma (DTC), its visible scarring and postoperative morbidity have mandatorily propelled a longitudinal shift to minimally invasive techniques. Robotic thyroidectomy using the bilateral axillo-breast approach (BABA) is an emerging transformative technique that combines robotic precision and visualization with oncologic efficacy and superior cosmetic outcomes to enhance recovery. Molecular diagnostics such as next-generation sequencing (NGS) and microRNA classifiers have transformed preoperative planning. These tools aid in accurate risk stratification, enabling clinicians to determine the course of surgery and avoid overtreatment. Artificial intelligence (AI) enhances precision medicine by improving nodule classification, predicting surgical risks, providing intraoperative navigation guidance, and supporting postoperative histopathological evaluations. Despite these innovations, cost, accessibility, and ethical governance issues highlight persistent challenges. This review consolidates the current state of robotic-assisted surgery for thyroid cancer with molecular profiling and AI. It proposes future aims and strides toward precision surgery that is accessible worldwide.
{"title":"Integrating Robotic Bilateral Axillo-Breast Approach Thyroidectomy with Molecular Diagnostics and Artificial Intelligence in Thyroid Cancer Care.","authors":"Qiang Deng, Xiaoping Men, Duo Jin, Yuzhuo Bai","doi":"10.4062/biomolther.2025.125","DOIUrl":"https://doi.org/10.4062/biomolther.2025.125","url":null,"abstract":"<p><p>Thyroid cancer is one of the most prevalent cancers in the world, accounting for the increased sensitivity of diagnostic assessments, the environment, and extensive imaging. While open thyroidectomy still forms the base surgical treatment of differentiated thyroid carcinoma (DTC), its visible scarring and postoperative morbidity have mandatorily propelled a longitudinal shift to minimally invasive techniques. Robotic thyroidectomy using the bilateral axillo-breast approach (BABA) is an emerging transformative technique that combines robotic precision and visualization with oncologic efficacy and superior cosmetic outcomes to enhance recovery. Molecular diagnostics such as next-generation sequencing (NGS) and microRNA classifiers have transformed preoperative planning. These tools aid in accurate risk stratification, enabling clinicians to determine the course of surgery and avoid overtreatment. Artificial intelligence (AI) enhances precision medicine by improving nodule classification, predicting surgical risks, providing intraoperative navigation guidance, and supporting postoperative histopathological evaluations. Despite these innovations, cost, accessibility, and ethical governance issues highlight persistent challenges. This review consolidates the current state of robotic-assisted surgery for thyroid cancer with molecular profiling and AI. It proposes future aims and strides toward precision surgery that is accessible worldwide.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145720787","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03DOI: 10.4062/biomolther.2025.126
Li Cao, Jing Wang
Osteoblasts primarily originate from mesenchymal stem cells (MSCs) within the bone marrow. These stem cells possess the ability to differentiate into osteoblasts, which are responsible for secreting bone matrix, promoting bone formation, and contributing to bone remodeling. Dysfunction in osteoblast activity can lead to various bone-related disorders, such as osteoporosis, delayed fracture healing, and skeletal deformities. In recent years, due to the adverse effects associated with the use of parathyroid hormone (PTH) analogs, bisphosphonates, and calmodulin-targeting drugs, there has been growing interest in exploring the mechanisms underlying osteoblast differentiation. Researchers are increasingly focusing on identifying natural compounds as potential treatments for osteoporosis. Among the signaling pathways involved, the Wnt/β-catenin pathway is recognized as a key regulator of osteoblast differentiation and a crucial therapeutic target in osteoporosis. However, both upregulation and downregulation of this pathway have been implicated in various diseases, including cancers. This review highlights the role of the Wnt/β-catenin signaling pathway in osteoblast differentiation, examines the association between pathway-related proteins and human diseases, and summarizes recent advancements in the development of natural compounds targeting this pathway for osteoporosis therapy.
{"title":"Roles of Wnt/β-Catenin Signaling in Osteoporosis, Disease Pathogenesis, and Natural Compound Intervention.","authors":"Li Cao, Jing Wang","doi":"10.4062/biomolther.2025.126","DOIUrl":"https://doi.org/10.4062/biomolther.2025.126","url":null,"abstract":"<p><p>Osteoblasts primarily originate from mesenchymal stem cells (MSCs) within the bone marrow. These stem cells possess the ability to differentiate into osteoblasts, which are responsible for secreting bone matrix, promoting bone formation, and contributing to bone remodeling. Dysfunction in osteoblast activity can lead to various bone-related disorders, such as osteoporosis, delayed fracture healing, and skeletal deformities. In recent years, due to the adverse effects associated with the use of parathyroid hormone (PTH) analogs, bisphosphonates, and calmodulin-targeting drugs, there has been growing interest in exploring the mechanisms underlying osteoblast differentiation. Researchers are increasingly focusing on identifying natural compounds as potential treatments for osteoporosis. Among the signaling pathways involved, the Wnt/β-catenin pathway is recognized as a key regulator of osteoblast differentiation and a crucial therapeutic target in osteoporosis. However, both upregulation and downregulation of this pathway have been implicated in various diseases, including cancers. This review highlights the role of the Wnt/β-catenin signaling pathway in osteoblast differentiation, examines the association between pathway-related proteins and human diseases, and summarizes recent advancements in the development of natural compounds targeting this pathway for osteoporosis therapy.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145660263","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-02DOI: 10.4062/biomolther.2025.134
Jae-Yong Kim, Rak Ho Son, Sang-Yoon Kim, Ji Hoon Kim, Sunhoo Kim, Chul Young Kim
Lycium Radicis Cortex (LRC), derived from the root bark of Lycium chinense Mill., has traditionally been used in East Asian medicine to mitigate heat in the blood and consumptive fever. This study investigates LRC's effects on skeletal muscle in aged mice subjected to forced exercise and examines the protective properties of its primary constituents, kukoamines A (KA) and B (KB), against dexamethasone (DEX)-induced muscle atrophy. Sixteen-month-old male C57BL/6 mice underwent regular swimming and received oral LRC supplementation for 8 weeks. The effects of KA and KB on muscle atrophy were further explored using C2C12 myotubes treated with DEX. LRC administration significantly enhanced muscle mass, strength, and endurance, while reducing plasma lactate and creatinine levels compared to the control group. LRC also upregulated mRNA expression of MyoD, myogenin, MHC, Akt, and mTOR, and downregulated myostatin, FoxO3a, MuRF1, and atrogin-1 in gastrocnemius and soleus muscles. Furthermore, KA and KB alleviated DEX-induced muscle atrophy in C2C12 myotubes by reducing proteolysis and ROS production, enhancing SOD activity, and improving mitochondrial function. Taken together, LRC may be a useful supplement in exercise-based muscle strengthening and amelioration of muscle disorders, and KA and KB have shown potential as preventive and therapeutic agents for muscle atrophy, indirectly suggesting that the efficacy of LRC is attributed to KA and KB.
枸杞根皮(Lycium Radicis Cortex, LRC),由枸杞根皮提取。传统上,它在东亚医学中被用来缓解血液中的热量和消耗性发烧。本研究探讨了LRC对受强迫运动的老年小鼠骨骼肌的影响,并研究了其主要成分kukoamines A (KA)和B (KB)对地塞米松(DEX)诱导的肌肉萎缩的保护作用。16月龄雄性C57BL/6小鼠定期游泳并口服LRC 8周。用DEX处理C2C12肌管,进一步探讨KA和KB对肌萎缩的影响。与对照组相比,LRC显著增强了肌肉质量、力量和耐力,同时降低了血浆乳酸和肌酐水平。LRC还上调了腓骨肌和比目鱼肌中MyoD、myogenin、MHC、Akt和mTOR的mRNA表达,下调了肌生长抑制素、FoxO3a、MuRF1和atroggin -1的表达。此外,KA和KB通过减少蛋白水解和ROS产生、提高SOD活性和改善线粒体功能,减轻了dex诱导的C2C12肌管肌肉萎缩。综上所述,LRC可能是基于运动的肌肉强化和改善肌肉疾病的有益补充,而KA和KB已显示出作为肌肉萎缩预防和治疗药物的潜力,间接表明LRC的功效归因于KA和KB。
{"title":"Lycium Radicis Cortex and Its Kukoamine Constituents Attenuate Sarcopenia by Modulating Anabolic and Catabolic Pathways.","authors":"Jae-Yong Kim, Rak Ho Son, Sang-Yoon Kim, Ji Hoon Kim, Sunhoo Kim, Chul Young Kim","doi":"10.4062/biomolther.2025.134","DOIUrl":"https://doi.org/10.4062/biomolther.2025.134","url":null,"abstract":"<p><p>Lycium Radicis Cortex (LRC), derived from the root bark of <i>Lycium chinense</i> Mill., has traditionally been used in East Asian medicine to mitigate heat in the blood and consumptive fever. This study investigates LRC's effects on skeletal muscle in aged mice subjected to forced exercise and examines the protective properties of its primary constituents, kukoamines A (KA) and B (KB), against dexamethasone (DEX)-induced muscle atrophy. Sixteen-month-old male C57BL/6 mice underwent regular swimming and received oral LRC supplementation for 8 weeks. The effects of KA and KB on muscle atrophy were further explored using C2C12 myotubes treated with DEX. LRC administration significantly enhanced muscle mass, strength, and endurance, while reducing plasma lactate and creatinine levels compared to the control group. LRC also upregulated mRNA expression of MyoD, myogenin, MHC, Akt, and mTOR, and downregulated myostatin, FoxO3a, MuRF1, and atrogin-1 in gastrocnemius and soleus muscles. Furthermore, KA and KB alleviated DEX-induced muscle atrophy in C2C12 myotubes by reducing proteolysis and ROS production, enhancing SOD activity, and improving mitochondrial function. Taken together, LRC may be a useful supplement in exercise-based muscle strengthening and amelioration of muscle disorders, and KA and KB have shown potential as preventive and therapeutic agents for muscle atrophy, indirectly suggesting that the efficacy of LRC is attributed to KA and KB.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145653448","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mitochondrial biogenesis represents a promising therapeutic target in triple-negative breast cancer (TNBC) due to its essential role in cancer cell metabolism and survival. The natural compound γ-Elemene exhibits potent anti-tumor activity, but its effects on mitochondrial regulation in TNBC remain unclear. In this study, we demonstrate that γ-Elemene induces dose-dependent cytotoxicity in MDA-MB-468 and HCC1806 TNBC cells while significantly impairing mitochondrial function, as shown by reduced membrane potential, oxidative phosphorylation capacity, and ATP production. γ-Elemene treatment markedly suppressed mitochondrial biogenesis, decreasing mitochondrial DNA content and downregulating key mitochondrial genes and proteins. These effects were associated with reduced expression of the master regulators NRF1 and TFAM, but independent of PGC-1α expression levels. Mechanistically, γ-Elemene upregulated the acetyltransferase GCN5, leading to enhanced PGC-1α acetylation. This upregulation occurs primarily through increased GCN5 transcription. Genetic ablation of GCN5 completely reversed γ-Elemene-induced PGC- 1α acetylation and restored mitochondrial biogenesis and cell viability, establishing a critical role for GCN5 in mediating these effects. Our findings reveal a novel mechanism whereby γ-Elemene disrupts mitochondrial function in TNBC through GCN5-mediated PGC-1α acetylation, providing new insights into its anti-cancer properties and potential therapeutic applications against TNBC.
{"title":"γ-Elemene Impairs Mitochondrial Biogenesis in Breast Cancer Cells by Upregulating GCN5-Mediated PGC-1α Acetylation.","authors":"Ling Tang, Mingyan Wang, Jia Liu, Qiong Yu, Chen Chen, Lihua Jia, Jiyi Xia","doi":"10.4062/biomolther.2025.119","DOIUrl":"https://doi.org/10.4062/biomolther.2025.119","url":null,"abstract":"<p><p>Mitochondrial biogenesis represents a promising therapeutic target in triple-negative breast cancer (TNBC) due to its essential role in cancer cell metabolism and survival. The natural compound γ-Elemene exhibits potent anti-tumor activity, but its effects on mitochondrial regulation in TNBC remain unclear. In this study, we demonstrate that γ-Elemene induces dose-dependent cytotoxicity in MDA-MB-468 and HCC1806 TNBC cells while significantly impairing mitochondrial function, as shown by reduced membrane potential, oxidative phosphorylation capacity, and ATP production. γ-Elemene treatment markedly suppressed mitochondrial biogenesis, decreasing mitochondrial DNA content and downregulating key mitochondrial genes and proteins. These effects were associated with reduced expression of the master regulators NRF1 and TFAM, but independent of PGC-1α expression levels. Mechanistically, γ-Elemene upregulated the acetyltransferase GCN5, leading to enhanced PGC-1α acetylation. This upregulation occurs primarily through increased GCN5 transcription. Genetic ablation of GCN5 completely reversed γ-Elemene-induced PGC- 1α acetylation and restored mitochondrial biogenesis and cell viability, establishing a critical role for GCN5 in mediating these effects. Our findings reveal a novel mechanism whereby γ-Elemene disrupts mitochondrial function in TNBC through GCN5-mediated PGC-1α acetylation, providing new insights into its anti-cancer properties and potential therapeutic applications against TNBC.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145653527","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-02DOI: 10.4062/biomolther.2025.090
Ga Hee Lee, Jin Pyo Lee, Akram Abdul Wahab, Na Yoon Heo, Chang Eun Park, Dong-Ha Lee
Chrysoeriol, a flavonoid naturally found in several plants, including Danggui Susan, a traditional herbal medicine, exhibits promising anti-inflammatory and antioxidant properties. Its potential to prevent cardiovascular diseases, primarily through inhibiting platelet activation and aggregation, has attracted significant interest. This study aimed to investigate the molecular mechanisms underlying the antiplatelet effects of chrysoeriol. The compound effectively suppressed collagen-induced platelet aggregation without inducing cytotoxicity. Chrysoeriol elevated intracellular levels of cyclic AMP (cAMP) and cyclic GMP (cGMP), enhanced inositol 1,4,5-trisphosphate receptor (IP3R) phosphorylation, and reduced cytosolic calcium (Ca2+) mobilization, all of which contributed to its antiplatelet action. Furthermore, chrysoeriol inhibited the phosphorylation of PI3K, Akt, JNK, and p38 MAPK, pathways involved in the activation of cytosolic phospholipase A2 (cPLA2) and thromboxane A2 (TXA2) production. These effects were accompanied by reduced TXA2 production and secretion of dense granules (ATP and serotonin). Chrysoeriol also impaired thrombin-induced clot retraction, further suggesting its capacity to regulate platelet responses and cytoskeletal rearrangements. These findings highlight chrysoeriol's multi-target mechanisms, including modulation of cyclic nucleotides, kinase pathways, and platelet function, offering potential as a therapeutic agent to prevent thrombotic cardiovascular events.
{"title":"Chrysoeriol Exerts Antiplatelet Effects by Regulating cAMP/cGMP and PI3K/MAPK Pathway.","authors":"Ga Hee Lee, Jin Pyo Lee, Akram Abdul Wahab, Na Yoon Heo, Chang Eun Park, Dong-Ha Lee","doi":"10.4062/biomolther.2025.090","DOIUrl":"https://doi.org/10.4062/biomolther.2025.090","url":null,"abstract":"<p><p>Chrysoeriol, a flavonoid naturally found in several plants, including Danggui Susan, a traditional herbal medicine, exhibits promising anti-inflammatory and antioxidant properties. Its potential to prevent cardiovascular diseases, primarily through inhibiting platelet activation and aggregation, has attracted significant interest. This study aimed to investigate the molecular mechanisms underlying the antiplatelet effects of chrysoeriol. The compound effectively suppressed collagen-induced platelet aggregation without inducing cytotoxicity. Chrysoeriol elevated intracellular levels of cyclic AMP (cAMP) and cyclic GMP (cGMP), enhanced inositol 1,4,5-trisphosphate receptor (IP<sub>3</sub>R) phosphorylation, and reduced cytosolic calcium (Ca<sup>2+</sup>) mobilization, all of which contributed to its antiplatelet action. Furthermore, chrysoeriol inhibited the phosphorylation of PI3K, Akt, JNK, and p38 MAPK, pathways involved in the activation of cytosolic phospholipase A2 (cPLA<sub>2</sub>) and thromboxane A2 (TXA<sub>2</sub>) production. These effects were accompanied by reduced TXA<sub>2</sub> production and secretion of dense granules (ATP and serotonin). Chrysoeriol also impaired thrombin-induced clot retraction, further suggesting its capacity to regulate platelet responses and cytoskeletal rearrangements. These findings highlight chrysoeriol's multi-target mechanisms, including modulation of cyclic nucleotides, kinase pathways, and platelet function, offering potential as a therapeutic agent to prevent thrombotic cardiovascular events.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":""},"PeriodicalIF":3.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145653380","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-01Epub Date: 2025-10-15DOI: 10.4062/biomolther.2025.143
Kyoung Ah Kang, Mei Jing Piao, Herath Mudiyanselage Maheshika Madhuwanthi Senavirathna, Hye-Jin Boo, Sang Pil Yoon, Joo Mi Yi, Jinny Park, Jin Won Hyun
Anticancer drug resistance remains a significant challenge to the efficacy of cancer treatment, with DNA repair enzymes contributing to this resistance. We hypothesized that thymine DNA glycosylases (TDGs) may be involved in anticancer drug resistance given their dual function of DNA repair and demethylation as well as investigated their possible involvement in the induction of β-catenin in SNUC5 cells resistant to 5-fluorouracil (SNUC5/5-FUR) and oxaliplatin (SNUC5/OXTR). The expression of TDG and phospho-β-catenin increased in both resistant cell types when compared to that in SNUC5 cells. Moreover, knockdown of TDG significantly suppressed phospho-β-catenin expression in both resistant cell types, resulting in enhanced sensitivity to anticancer drugs. TDG binding to the β-catenin promoter was stronger in both resistant cell types than in SNUC5 cells, showing a decreased methylation pattern in the CpG islands of the β-catenin promoter. Furthermore, another DNA demethylase, ten-eleven translocation 1 (TET1), showed the same pattern as TDG in both resistant cell types. Additionally, TDG significantly interacted more with TET1 in both resistant cell types than in SNUC5 cells, enhancing binding to the same locus in the β-catenin promoter. These findings suggest that TDG may be a promising target molecule for overcoming drug resistance in colorectal cancer.
{"title":"Involvement of the DNA Demethylase Thymine DNA Glycosylase in Anticancer Drug Resistance.","authors":"Kyoung Ah Kang, Mei Jing Piao, Herath Mudiyanselage Maheshika Madhuwanthi Senavirathna, Hye-Jin Boo, Sang Pil Yoon, Joo Mi Yi, Jinny Park, Jin Won Hyun","doi":"10.4062/biomolther.2025.143","DOIUrl":"10.4062/biomolther.2025.143","url":null,"abstract":"<p><p>Anticancer drug resistance remains a significant challenge to the efficacy of cancer treatment, with DNA repair enzymes contributing to this resistance. We hypothesized that thymine DNA glycosylases (TDGs) may be involved in anticancer drug resistance given their dual function of DNA repair and demethylation as well as investigated their possible involvement in the induction of β-catenin in SNUC5 cells resistant to 5-fluorouracil (SNUC5/5-FUR) and oxaliplatin (SNUC5/OXTR). The expression of TDG and phospho-β-catenin increased in both resistant cell types when compared to that in SNUC5 cells. Moreover, knockdown of TDG significantly suppressed phospho-β-catenin expression in both resistant cell types, resulting in enhanced sensitivity to anticancer drugs. TDG binding to the β-catenin promoter was stronger in both resistant cell types than in SNUC5 cells, showing a decreased methylation pattern in the CpG islands of the β-catenin promoter. Furthermore, another DNA demethylase, ten-eleven translocation 1 (TET1), showed the same pattern as TDG in both resistant cell types. Additionally, TDG significantly interacted more with TET1 in both resistant cell types than in SNUC5 cells, enhancing binding to the same locus in the β-catenin promoter. These findings suggest that TDG may be a promising target molecule for overcoming drug resistance in colorectal cancer.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"986-1003"},"PeriodicalIF":3.2,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12580638/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145290898","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 : 2025-11-01Epub Date: 2025-10-27DOI: 10.4062/biomolther.2025.153
Saerom Moon, Kyoung Song
Chimeric Antigen Receptor (CAR)-based cell and gene therapies have become transformative treatments, offering targeted and durable responses, especially in hematologic malignancies. This review analyzes 1,744 CAR clinical trials registered on Clinical-Trials.gov as of 2024, focusing on platform types, indications, target antigens, therapeutic strategies, and late-phase development. CAR-T therapies predominate, followed by CAR-NK, CAR-NKT, CAR-M and CAR-DC platforms. Approximately 92% of trials target tumors, with hematologic malignancies accounting for 65% of indications; CD19 and BCMA are primary targets in Phase 3 studies. Solid tumor applications are expanding steadily, driven by unmet clinical needs and advances in CAR engineering. Although monospecific CARs dominate, dual, bispecific, and universal designs are gaining traction to overcome antigen heterogeneity and tumor escape. Combination therapies, such as CAR-T with chemotherapy or monoclonal antibodies, are increasingly used to improve efficacy. CAR-NK therapies, while in early development, show promise due to favorable safety profiles and off-the-shelf allogeneic potential. The United States and China lead global development, supported by robust research ecosystems and industrial investment. Overall, CAR-based therapeutics are evolving from hematologic specialization toward broader clinical application, addressing challenges and guiding future strategies.
{"title":"CAR-Based Cell and Gene Therapies: Global Clinical Landscape and Emerging Therapeutic Strategies from ClinicalTrials.gov.","authors":"Saerom Moon, Kyoung Song","doi":"10.4062/biomolther.2025.153","DOIUrl":"10.4062/biomolther.2025.153","url":null,"abstract":"<p><p>Chimeric Antigen Receptor (CAR)-based cell and gene therapies have become transformative treatments, offering targeted and durable responses, especially in hematologic malignancies. This review analyzes 1,744 CAR clinical trials registered on Clinical-Trials.gov as of 2024, focusing on platform types, indications, target antigens, therapeutic strategies, and late-phase development. CAR-T therapies predominate, followed by CAR-NK, CAR-NKT, CAR-M and CAR-DC platforms. Approximately 92% of trials target tumors, with hematologic malignancies accounting for 65% of indications; CD19 and BCMA are primary targets in Phase 3 studies. Solid tumor applications are expanding steadily, driven by unmet clinical needs and advances in CAR engineering. Although monospecific CARs dominate, dual, bispecific, and universal designs are gaining traction to overcome antigen heterogeneity and tumor escape. Combination therapies, such as CAR-T with chemotherapy or monoclonal antibodies, are increasingly used to improve efficacy. CAR-NK therapies, while in early development, show promise due to favorable safety profiles and off-the-shelf allogeneic potential. The United States and China lead global development, supported by robust research ecosystems and industrial investment. Overall, CAR-based therapeutics are evolving from hematologic specialization toward broader clinical application, addressing challenges and guiding future strategies.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"907-923"},"PeriodicalIF":3.2,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12580648/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145372089","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 : 2025-11-01Epub Date: 2025-10-01DOI: 10.4062/biomolther.2025.046
Maha AlDoughaim, Nada AlSuhebany, Mohammed AlZahrani, Atheer AlDairem, Sahar S Alghamdi
Suzetrigine is a novel non-opioid analgesic that selectively inhibits the Nav1.8 sodium channel, which plays a key role in peripheral pain signaling. By blocking action potential propagation in nociceptors, it effectively reduces pain without affecting the central nervous system, thus avoiding the risks associated with opioids, such as addiction and respiratory depression. In two phase 2 clinical trials, suzetrigine demonstrated superior pain control compared to placebo and showed comparable effectiveness to hydrocodone/acetaminophen for treating moderate to severe acute pain after abdominoplasty and bunionectomy with an acceptable safety profile. Current findings support suzetrigine's potential role as a safer alternative to opioids in managing moderate to severe pain.
{"title":"Transforming Pain Management: Suzetrigine, a Novel Non-Opioid Analgesic.","authors":"Maha AlDoughaim, Nada AlSuhebany, Mohammed AlZahrani, Atheer AlDairem, Sahar S Alghamdi","doi":"10.4062/biomolther.2025.046","DOIUrl":"10.4062/biomolther.2025.046","url":null,"abstract":"<p><p>Suzetrigine is a novel non-opioid analgesic that selectively inhibits the Nav1.8 sodium channel, which plays a key role in peripheral pain signaling. By blocking action potential propagation in nociceptors, it effectively reduces pain without affecting the central nervous system, thus avoiding the risks associated with opioids, such as addiction and respiratory depression. In two phase 2 clinical trials, suzetrigine demonstrated superior pain control compared to placebo and showed comparable effectiveness to hydrocodone/acetaminophen for treating moderate to severe acute pain after abdominoplasty and bunionectomy with an acceptable safety profile. Current findings support suzetrigine's potential role as a safer alternative to opioids in managing moderate to severe pain.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"934-939"},"PeriodicalIF":3.2,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12580649/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145198017","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 : 2025-11-01Epub Date: 2025-10-27DOI: 10.4062/biomolther.2025.163
Jeong Mi Lee, Young Hun Hwang, Bo Mi Park, Hee Bin Seo, Da Yeong Nam, Eung Won Kim, So Min Kang, Jae Sung Hwang
Topically applied hyaluronic acid (HA) hydrates the skin without efficient penetration. This study compared the penetration efficiency of liposomal hyaluronic acid (LPS-HA) against that of conventional HA across different models. Dynamic light scattering revealed that particles of LPS-HA (226.1 nm, PDI 0.2898) were smaller than those of HA (798.4 nm, PDI 0.8709). In Strat-M® membrane assays, permeability over 24 h was higher with LPS-HA (629.37 ± 103.26%) than that of HA (508.04 ± 93.80%; p<0.05). In keratinocytes, LPS-HA increased differentiation markers filaggrin and caspase-14 in a concentration-dependent manner, with maximal induction at 1% (186.6 ± 6.99% and 249.3 ± 8.60%) vs. HA (117.9 ± 7.64% and 130.1 ± 2.90%; p<0.05). In fibroblasts, LPS-HA increased the expression of type I and type III collagens (138.4 and 133.6%) without increasing that of elastin (68.3-94.7%) and reduced UVB-induced IL-6 (79.1-90.2% of UVB; p<0.05). Ex vivo, LPS-HA enhanced HAS-3 mRNA (3.03 ± 0.19-fold vs. 1.31 ± 0.13-fold with HA; p<0.05) and increased epidermal hyaluronan staining. In PM10-treated human skin, LPS-HA reduced inflammatory cytokines (TNF-α, IL-6, IL-8, and IL-1β) and suppressed mast cell degranulation, similar to dexamethasone, and reduced ROS formation (124.46 ± 8.45% vs. 169.35 ± 9.40% in PM10-only, p<0.01) without histological abnormalities. In a 20-subject clinical study, corneometric hydration with LPS-HA was higher than that of control (96.99% vs. 36.31%; RM-ANOVA, p<0.001). Collectively, LPS-HA enhanced skin permeation, hydration, and anti-inflammatory responses, supporting its potential as a cosmetic moisturizing ingredient.
{"title":"Liposomal Hyaluronic Acid Enhances Skin Permeation and Hydration: Evidence from In Vitro, Ex Vivo, and In Vivo Studies.","authors":"Jeong Mi Lee, Young Hun Hwang, Bo Mi Park, Hee Bin Seo, Da Yeong Nam, Eung Won Kim, So Min Kang, Jae Sung Hwang","doi":"10.4062/biomolther.2025.163","DOIUrl":"10.4062/biomolther.2025.163","url":null,"abstract":"<p><p>Topically applied hyaluronic acid (HA) hydrates the skin without efficient penetration. This study compared the penetration efficiency of liposomal hyaluronic acid (LPS-HA) against that of conventional HA across different models. Dynamic light scattering revealed that particles of LPS-HA (226.1 nm, PDI 0.2898) were smaller than those of HA (798.4 nm, PDI 0.8709). In Strat-M® membrane assays, permeability over 24 h was higher with LPS-HA (629.37 ± 103.26%) than that of HA (508.04 ± 93.80%; <i>p</i><0.05). In keratinocytes, LPS-HA increased differentiation markers filaggrin and caspase-14 in a concentration-dependent manner, with maximal induction at 1% (186.6 ± 6.99% and 249.3 ± 8.60%) <i>vs</i>. HA (117.9 ± 7.64% and 130.1 ± 2.90%; <i>p</i><0.05). In fibroblasts, LPS-HA increased the expression of type I and type III collagens (138.4 and 133.6%) without increasing that of elastin (68.3-94.7%) and reduced UVB-induced IL-6 (79.1-90.2% of UVB; <i>p</i><0.05). <i>Ex vivo</i>, LPS-HA enhanced HAS-3 mRNA (3.03 ± 0.19-fold <i>vs</i>. 1.31 ± 0.13-fold with HA; <i>p</i><0.05) and increased epidermal hyaluronan staining. In PM10-treated human skin, LPS-HA reduced inflammatory cytokines (TNF-α, IL-6, IL-8, and IL-1β) and suppressed mast cell degranulation, similar to dexamethasone, and reduced ROS formation (124.46 ± 8.45% <i>vs</i>. 169.35 ± 9.40% in PM10-only, <i>p</i><0.01) without histological abnormalities. In a 20-subject clinical study, corneometric hydration with LPS-HA was higher than that of control (96.99% <i>vs</i>. 36.31%; RM-ANOVA, <i>p</i><0.001). Collectively, LPS-HA enhanced skin permeation, hydration, and anti-inflammatory responses, supporting its potential as a cosmetic moisturizing ingredient.</p>","PeriodicalId":8949,"journal":{"name":"Biomolecules & Therapeutics","volume":" ","pages":"1073-1084"},"PeriodicalIF":3.2,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12580642/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145372136","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}