Ji Eun Kim, Yun Kim, Jiwon Bae, Eileen Laurel Yoon, Hyun Sung Kim, Sung Ryol Lee, Tae Hyun Yoon, Dae Won Jun
{"title":"A novel 11β-HSD1 inhibitor ameliorates liver fibrosis by inhibiting the notch signaling pathway and increasing NK cell population.","authors":"Ji Eun Kim, Yun Kim, Jiwon Bae, Eileen Laurel Yoon, Hyun Sung Kim, Sung Ryol Lee, Tae Hyun Yoon, Dae Won Jun","doi":"10.1007/s12272-025-01534-4","DOIUrl":"https://doi.org/10.1007/s12272-025-01534-4","url":null,"abstract":"<p><p>11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) regulates hepatic glucose output and is implicated in liver fibrosis. We aimed to investigate the anti-fibrotic effect of a novel 11β-HSD1 inhibitor in a thioacetamide (TAA)-induced liver fibrosis mouse model. Mice were administered TAA for 19 weeks and treated with 11β-HSD1 inhibitor for the last 9 weeks. Treatment with 11β-HSD1 inhibitor significantly reduced fibrosis area, alanine aminotransferase, and aspartate aminotransferase levels compared to the TAA-only group. Inhibition of 11β-HSD1 led to a decrease in intracellular cortisol levels, which suppressed the activation of hepatic stellate cells. RNA sequencing revealed significant downregulation of the Notch signaling pathway, including reduced expression of Notch ligands and receptors, as well as downstream genes. Furthermore, 11β-HSD1 inhibition enhanced NK cell-mediated immune responses, as indicated by the upregulation of NK cell-related genes and increased NK cell populations confirmed by mass cytometry. This increase in NK cell activity contributed to the clearance of activated HSCs and the attenuation of fibrosis. These findings suggest that 11β-HSD1 inhibition alleviates liver fibrosis through Notch pathway suppression and enhancement of NK cell-mediated immune responses. Our results support the therapeutic potential of a novel 11β-HSD1 inhibitor for treating liver fibrosis.</p>","PeriodicalId":8287,"journal":{"name":"Archives of Pharmacal Research","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143424898","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}
Mona F Mahmoud, Mohamed El-Nagar, Hany M El-Bassossy
{"title":"Retraction Note: Anti-inflammatory effect of atorvastatin on vascular reactivity and insulin resistance in fructose fed rats.","authors":"Mona F Mahmoud, Mohamed El-Nagar, Hany M El-Bassossy","doi":"10.1007/s12272-025-01535-3","DOIUrl":"https://doi.org/10.1007/s12272-025-01535-3","url":null,"abstract":"","PeriodicalId":8287,"journal":{"name":"Archives of Pharmacal Research","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143397902","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}
Ahmed Elkamhawy, Usama M Ammar, Minkyoung Kim, Anam Rana Gul, Tae Jung Park, Kyeong Lee
{"title":"Discovery of novel naphthalene-based diarylamides as pan-Raf kinase inhibitors with promising anti-melanoma activity: rational design, synthesis, in vitro and in silico screening.","authors":"Ahmed Elkamhawy, Usama M Ammar, Minkyoung Kim, Anam Rana Gul, Tae Jung Park, Kyeong Lee","doi":"10.1007/s12272-025-01533-5","DOIUrl":"https://doi.org/10.1007/s12272-025-01533-5","url":null,"abstract":"<p><p>Raf kinase enzymes are often dysregulated in melanoma. While sorafenib demonstrates strong activity against wild-type B-Raf, it fails to effectively inhibit the mutated form of B-Raf. In this study, sorafenib served as a lead compound for the development of new derivatives designed to enhance inhibitory activity across multiple Raf isoforms (pan-Raf inhibitors). Novel naphthalene-based diarylamide derivatives were subsequently designed, synthesized, and evaluated for their biological activity against various Raf kinase isoforms and the melanoma A375 cell line. Among these, compound 9a, containing a difluoromethoxy group, demonstrated strong inhibitory activity across B-Raf<sup>WT</sup>, B-Raf<sup>V600E</sup>, and c-Raf. Additionally, it induced G2/M phase arrest and triggered dose-dependent apoptosis, effectively suppressing both cell proliferation and survival. Compound 9a also exhibited high selectivity for Raf isoforms with minimal off-target effects, underscoring its specificity and therapeutic potential for Raf-driven malignancies.</p>","PeriodicalId":8287,"journal":{"name":"Archives of Pharmacal Research","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143370348","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}
{"title":"Tumor microenvironment regulation by reactive oxygen species-mediated inflammasome activation.","authors":"Jeong-Hoon Jang, Do-Hee Kim, Kyung-Soo Chun","doi":"10.1007/s12272-025-01532-6","DOIUrl":"10.1007/s12272-025-01532-6","url":null,"abstract":"<p><p>Tumor microenvironment (TME) is composed of diverse cell types whose interactions, both direct and indirect, significantly influence tumorigenesis and therapeutic outcomes. Within TME, reactive oxygen species (ROS) are produced by various cells and exhibit a dual role: moderate ROS levels promote tumor initiation and progression, whereas excessive levels induce cancer cell death, influencing the efficacy of anticancer therapies. Inflammasomes, cytosolic multiprotein complexes, are pivotal in multiple stages of tumorigenesis and play a crucial role in establishing the inflammatory TME. By releasing cytokines such as IL-1β and IL-18, inflammasomes contribute to immune cell recruitment and sustain a chronic inflammatory state that supports tumor growth. ROS are critical regulators of inflammasome activation, with the impact of ROS-mediated activation differing across cell types, leading to distinct influences on tumor progression and therapeutic responses. This review explores how ROS drive inflammasome activation in various TME-associated cells and the reciprocal ROS generation induced by inflammasomes, examining their multifaceted impact on tumorigenesis and therapeutic efficacy. By elucidating the complex interplay between ROS and inflammasomes in TME, we provide insights into potential therapeutic approaches that could modulate cancer progression and enhance treatment outcomes.</p>","PeriodicalId":8287,"journal":{"name":"Archives of Pharmacal Research","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143063445","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}
{"title":"Physiologically based pharmacokinetic (PBPK) modeling of gliclazide for different genotypes of CYP2C9 and CYP2C19.","authors":"Hye-Jung Park, Sang-Ho Lee, Pureum Kang, Chang-Keun Cho, Choon-Gon Jang, Seok-Yong Lee, Yun Jeong Lee, Jung-Woo Bae, Chang-Ik Choi","doi":"10.1007/s12272-024-01528-8","DOIUrl":"https://doi.org/10.1007/s12272-024-01528-8","url":null,"abstract":"<p><p>Gliclazide is a sulfonylurea hypoglycemic agent used to treat type 2 diabetes. Cytochrome P450 (CYP) 2C9 and CYP2C19 are primarily involved in the hepatic metabolism of gliclazide. The two CYP isozymes are highly polymorphic, and their genetic polymorphisms are known to significantly impact the pharmacokinetics of gliclazide. In the present study, the physiologically based pharmacokinetic (PBPK) model was developed using data from subjects whose pharmacokinetic parameters were influenced by the genetic polymorphisms of the CYP metabolic enzymes. All predicted plasma concentration-time profiles generated by the model showed visual agreement with the observed data, and the pharmacokinetic results were within the twofold error range. Individual simulation results showed additional metrics: average fold error (- 0.19 to 0.07), geometric mean fold error (1.13-1.56), and mean relative deviation (1.18-1.58) for AUC, C<sub>max</sub>, T<sub>1/2</sub>, T<sub>max</sub>, CL/F, and V<sub>d</sub> values. These results met the standard evaluation criteria. The validation across a total of 8 studies and 7 races also satisfied the twofold error range for AUC, C<sub>max</sub>, and T<sub>1/2</sub>. Therefore, variations in gliclazide exposure according to individuals' CYP2C9 and CYP2C19 genotypes were properly captured through PBPK modeling in this study. This PBPK model may allow us to predict the gliclazide pharmacokinetics of patients with genetic polymorphisms in CYP2C9 and CYPC19 under various conditions, ultimately contributing to the realization of individualized drug therapy.</p>","PeriodicalId":8287,"journal":{"name":"Archives of Pharmacal Research","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142930473","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}
{"title":"Canine mammary tumors as a promising adjunct preclinical model for human breast cancer research: similarities, opportunities, and challenges","authors":"Zeeshan Ahmad Bhutta, Kyung-Chul Choi","doi":"10.1007/s12272-024-01524-y","DOIUrl":"10.1007/s12272-024-01524-y","url":null,"abstract":"<div><p>Despite significant progress in the field of human breast cancer research and treatment, there is a consistent increase in the incidence rate of 0.5 percent annually, posing challenges in the development of effective novel therapeutic strategies. The failure rate of drugs in clinical trials stands at approximately 95%, primarily attributed to the limitations and lack of reliability of existing preclinical models, such as mice, which do not mimic human tumor biology. This article examines the potential utility of canine mammary tumors as an adjunct preclinical model for investigating human breast cancer. Given the numerous similarities between canine and human breast cancer, canines present a promising alternative model. The discussion delves into the intricate molecular and clinical aspects of human breast cancer and canine mammary tumors, shedding light on the tumors' molecular profiles, identifying specific molecular markers, and the application of radiological imaging modalities. Furthermore, the manuscript addresses the current constraints of preclinical cancer studies, the benefits of using canines as models, and the obstacles linked to the canine mammary tumors model. By concentrating on these elements, this review aims to highlight the viability of canine models in enhancing our understanding and management of human breast cancer.</p></div>","PeriodicalId":8287,"journal":{"name":"Archives of Pharmacal Research","volume":"48 1","pages":"43 - 61"},"PeriodicalIF":6.9,"publicationDate":"2025-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142920716","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}
{"title":"Effects of fluconazole on the pharmacokinetics of celecoxib and its carboxylic acid metabolite in different CYP2C9 genotypes.","authors":"Chang-Keun Cho, Pureum Kang, Choon-Gon Jang, Yun Jeong Lee, Jung-Woo Bae, Chang-Ik Choi, Seok-Yong Lee","doi":"10.1007/s12272-024-01531-z","DOIUrl":"https://doi.org/10.1007/s12272-024-01531-z","url":null,"abstract":"<p><p>This study aimed to investigate the effects of fluconazole, a moderate inhibitor of CYP2C9 and CYP3A4, on the pharmacokinetics of celecoxib and its carboxylic acid metabolite in different CYP2C9 genotypes. A total of thirty-nine healthy Korean male volunteers were divided into three different CYP2C9 genotype groups (CYP2C9*1/*1, *1/*3 and *3/*3 genotypes) and were enrolled in the celecoxib alone trial, celecoxib with fluconazole trial, or both. In the celecoxib alone trial, participants received a single oral dose of 200 mg celecoxib. In the celecoxib with fluconazole trial, participants received 300 mg fluconazole on day 1, 150 mg fluconazole once daily for four consecutive days (day 2-5), and a coadministration of 200 mg celecoxib with 150 mg fluconazole on day 6. Plasma concentrations of celecoxib and celecoxib carboxylic acid were determined by using HPLC-MS/MS. In the CYP2C9*1/*1 genotype group, fluconazole treatment increased AUC<sub>inf</sub> of celecoxib by 2.61-fold, and decreased CL/F by 60.4% (both p < 0.001). In the CYP2C9*1/*3 genotype group, fluconazole treatment increased AUC<sub>inf</sub> of celecoxib by 2.44-fold (p < 0.001), prolonged t<sub>1/2</sub> by 1.36-fold (p < 0.05), and decreased CL/F by 60.4% (p < 0.001). Fluconazole treatment increased AUC<sub>inf</sub> of celecoxib by 2.23-fold, prolonged t<sub>1/2</sub> by 1.64-fold, and decreased CL/F by 53.8% in the subject with CYP2C9*3/*3 genotype. C<sub>max</sub> of celecoxib carboxylic acid significantly decreased in CYP2C9*1/*1 and *1/*3 genotypes (p < 0.01 and p < 0.05, respectively), following fluconazole treatment, whereas AUC<sub>inf</sub> showed no significant changes in any CYP2C9 genotype group. In conclusion, fluconazole affected the pharmacokinetics of celecoxib in different CYP2C9 genotypes.</p>","PeriodicalId":8287,"journal":{"name":"Archives of Pharmacal Research","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2024-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142891715","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}
Nam-Hee Kim, So-Jin Sim, Hong-Gyu Han, Jeong-Hyuk Yoon, Yong-Hyun Han
{"title":"Immunosenescence and age-related immune cells: causes of age-related diseases.","authors":"Nam-Hee Kim, So-Jin Sim, Hong-Gyu Han, Jeong-Hyuk Yoon, Yong-Hyun Han","doi":"10.1007/s12272-024-01529-7","DOIUrl":"https://doi.org/10.1007/s12272-024-01529-7","url":null,"abstract":"<p><p>Immunosenescence is a weakening of the immune system due to aging, characterized by changes in immune cells and dysregulated immune function. Age-related immune cells are increasing with aging. They are associated with chronic prolonged inflammation, causing tissue dysfunction and age-related diseases. Here, we discuss increased pro-inflammatory activity of aged macrophages, accumulation of lymphocytes with an age-associated phenotype, and specific alterations in both functions and characteristics of these immune cells. These cellular changes are associated with development of age-related diseases. Additionally, we reviewed various therapeutic strategies targeting age-related immunosenescence, providing pathways to mitigate effects of age-related diseases.</p>","PeriodicalId":8287,"journal":{"name":"Archives of Pharmacal Research","volume":" ","pages":""},"PeriodicalIF":6.9,"publicationDate":"2024-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142891716","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}
Shrawani Lamichhane, Jo-Eun Seo, Ji Hoon Jeong, Sooyeun Lee, Sangkil Lee
{"title":"Ideal animal models according to multifaceted mechanisms and peculiarities in neurological disorders: present and challenges","authors":"Shrawani Lamichhane, Jo-Eun Seo, Ji Hoon Jeong, Sooyeun Lee, Sangkil Lee","doi":"10.1007/s12272-024-01527-9","DOIUrl":"10.1007/s12272-024-01527-9","url":null,"abstract":"<div><p>Neurological disorders, encompassing conditions such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS), pose a significant global health challenge, affecting millions worldwide. With an aging population and increased life expectancy, the prevalence of these disorders is escalating rapidly, leading to substantial economic burdens exceeding trillions of dollars annually. Animal models play a crucial role in understanding the underlying mechanisms of these disorders and developing effective treatments. Various species, including rodents, non-human primates, and fruit flies, are utilized to replicate specific aspects of human neurological conditions. However, selecting the ideal animal model requires careful consideration of its proximity to human disease conditions and its ability to mimic disease pathobiology and pharmacological responses. An Animal Model Quality Assessment (AMQA) tool has been developed to facilitate this selection process, focusing on assessing models based on their similarity to human conditions and disease pathobiology. Therefore, integrating intrinsic and extrinsic factors linked to the disease into the study's objectives aids in constructing a biological information matrix for comparing disease progression between the animal model and human disease. Ultimately, selecting an ideal animal disease model depends on its predictive, face, and construct validity, ensuring relevance and reliability in translational research efforts.</p></div>","PeriodicalId":8287,"journal":{"name":"Archives of Pharmacal Research","volume":"48 1","pages":"62 - 88"},"PeriodicalIF":6.9,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142845706","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}