1
|
Wei H, Zhao Q. CYP2D6 polymorphism rs1065852 significantly increases the risk of type 2 diabetes. Ann Med 2025; 57:2470956. [PMID: 40028882 DOI: 10.1080/07853890.2025.2470956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Genetic variations within the cytochrome P450 (CYP) gene family are significant determinants of type 2 diabetes mellitus (T2DM) susceptibility. This study aimed to investigate the association between CYP2C8 and CYP2D6 gene variants and the risk of T2DM. METHODS We conducted a case-control study involving 512 individuals with T2DM and 515 controls. Genotyping of CYP2C8 and CYP2D6 polymorphisms was performed using the Agena MassARRAY system. Logistic regression analysis was employed to estimate the odds ratios (ORs) and 95% confidence intervals (CIs), thereby assessing the relationship between these genetic variants and T2DM risk. Additionally, multifactor dimensionality reduction (MDR) was utilized to assess the potential interaction effects of SNPs on T2DM risk. RESULTS The study found a strong correlation between rs1065852 and increased risk of T2DM in overall (A vs. G: OR = 1.22, 95% CI: 1.03-1.45, p = .024; AA vs. GG: OR = 1.46, 95% CI: 1.04-2.06, p = .031; AA-AG vs. GG: OR = 1.36, 95% CI: 1.04-1.79, p = .026; additive: OR = 1.21, 95% CI: 1.02-1.44, p = .027), males and age < 59 subgroups. However, there is no significant association between the CYP2C8 polymorphisms (rs1934953, rs1934951, rs2275620 and rs17110453) and T2DM risk. MDR analysis results showed that the best model was the one locus model (rs1065852, testing accuracy = 0.534; OR = 1.39; 95% CI: 1.05-1.85; p = .023; CVC = 10/10), indicating that rs1065852 is an independent risk factor for T2DM. CONCLUSIONS This study suggests that rs1065852 (CYP2D6) is an independent risk factor for T2DM. Further research is warranted to validate these results and explore their clinical implications.
Collapse
Affiliation(s)
- Huiyi Wei
- Medical College of Yan'an University, Yan'an, China
| | - Qingbin Zhao
- Department of Geratology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
2
|
Zhang Y, Mo C, Ai P, He X, Xiao Q, Yang X. Pharmacomicrobiomics: a new field contributing to optimizing drug therapy in Parkinson's disease. Gut Microbes 2025; 17:2454937. [PMID: 39875349 PMCID: PMC11776486 DOI: 10.1080/19490976.2025.2454937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/19/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
Gut microbiota, which act as a determinant of pharmacokinetics, have long been overlooked. In recent years, a growing body of evidence indicates that the gut microbiota influence drug metabolism and efficacy. Conversely, drugs also exert a substantial influence on the function and composition of the gut microbiota. Pharmacomicrobiomics, an emerging field focusing on the interplay of drugs and gut microbiota, provides a potential foundation for making certain advances in personalized medicine. Understanding the communication between gut microbiota and antiparkinsonian drugs is critical for precise treatment of Parkinson's disease. Here, we provide a historical overview of the interplay between gut microbiota and antiparkinsonian drugs. Moreover, we discuss potential mechanistic insights into the complex associations between gut microbiota and drug metabolism. In addition, we also draw attention to microbiota-based biomarkers for predicting antiparkinsonian drug efficacy and examine current state-of-the-art knowledge of microbiota-based strategies to optimize drug therapy in Parkinson's disease.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengjun Mo
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Penghui Ai
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoqin He
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Xiao
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodong Yang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Sun L, Feng K, Bai D, Yu Y, Shen WT, Zhang JA, Fang RH, Gao W, Zhang L. Hepatic endoplasmic reticulum-derived nanodiscs for broad-spectrum drug detoxification. Biomaterials 2025; 318:123188. [PMID: 39954312 DOI: 10.1016/j.biomaterials.2025.123188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Drug overdose is a pressing global public health challenge, with current detoxification treatments often lacking the broad-spectrum efficacy needed for emergency applications. Inspired by the unique advantages of cell membrane-derived nanodiscs (CNDs), including their compact size, rapid distribution, and preservation of native cell membrane functions, we developed endoplasmic reticulum (ER)-derived nanodiscs (ER-NDs) from the ER membranes of mouse hepatic cells for broad-spectrum drug detoxification. ER-NDs retain natural cytochrome P450 (CYP) enzymes, enabling effective detoxification of three model drugs: bupropion, haloperidol, and propranolol. Cell-based assays demonstrated ER-NDs' ability to mitigate drug-induced cytotoxicity, reduce oxidative stress, and restore antioxidant defenses. In mouse models of drug intoxication, ER-ND treatment significantly improved survival rates and alleviated drug-induced oxidative damage. Importantly, ER-NDs showed no evidence of acute toxicity in vivo. These findings underscore the potential of ER-NDs as a versatile platform for broad-spectrum drug detoxification and as a promising tool for managing drug intoxication in emergency and clinical settings.
Collapse
Affiliation(s)
- Lei Sun
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kailin Feng
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Dean Bai
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yiyan Yu
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Wei-Ting Shen
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jiayuan Alex Zhang
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H Fang
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA
| | - Weiwei Gao
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Liangfang Zhang
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
4
|
Husain I, Gurley BJ, Kothapalli HB, Wang YH, Vedova LD, Chittiboyina AG, Khan IA, Khan SI. Evaluation of bioaccessibility, metabolic clearance and interaction with xenobiotic receptors (PXR and AhR) of cinnamaldehyde. FOOD CHEMISTRY. MOLECULAR SCIENCES 2025; 10:100237. [PMID: 39845339 PMCID: PMC11751564 DOI: 10.1016/j.fochms.2024.100237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/02/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
Cinnamon is one of the oldest known spices used in various food delicacies and herbal formulations. Cinnamaldehyde is a primary active constituent of cinnamon and substantially contributes to the food additive and medicinal properties of cinnamon. This report deals with cinnamaldehyde bioaccessibility, metabolic clearance, and interaction with human xenobiotic receptors (PXR and AhR). Results showed the bioaccessibility of cinnamaldehyde was 100 % in both fasted and fed-state gastric and intestinal fluids. Upon incubation with human liver microsomes (HLMs) and human liver S-9 fraction, cinnamaldehyde (alone or in cinnamon oil) rapidly oxidized into cinnamic acid. Cinnamon oil dose-dependently activated AhR in human AhR-reporter cells, but cinnamaldehyde and cinnamic acid did not affect AhR. In addition, cinnamon oil and cinnamic acid dose-dependently activated PXR in human hepatic (HepG2) and intestinal (LS174T) cells. Both cinnamon oil and cinnamaldehyde inhibited the catalytic activity of CYP2C9 and CYP1A2. Our findings indicated that cinnamaldehyde (alone or in cinnamon oil) possesses high bioaccessibility and adequate metabolic stability. Hence, while controlled ingestion of cinnamon-containing foods or supplements may have beneficial effects but overconsumption could induce PXR or AhR-dependent herb-drug interactions (HDIs) which can bring deleterious effects on human health, particularly in individuals with chronic health conditions.
Collapse
Affiliation(s)
- Islam Husain
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, MS 38677, United States
| | - Bill J. Gurley
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, MS 38677, United States
| | - Hari Babu Kothapalli
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, MS 38677, United States
| | - Yan-Hong Wang
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, MS 38677, United States
| | - Larissa Della Vedova
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133 Milan, Italy
| | - Amar G. Chittiboyina
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, MS 38677, United States
| | - Ikhlas A. Khan
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, MS 38677, United States
- Department of Bio-Molecular Sciences, School of Pharmacy, The University of Mississippi, MS 38677, United States
| | - Shabana I. Khan
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, MS 38677, United States
- Department of Bio-Molecular Sciences, School of Pharmacy, The University of Mississippi, MS 38677, United States
| |
Collapse
|
5
|
Coimbra PPS, Teixeira ADC, Trindade MEF, Brito GO, Antonio ADS, Souza L, Silva-E-Silva ACAGD, Pereira HMG, Veiga-Junior VFD, Felzenszwalb I, Teodoro AJ, Araujo-Lima CF. Beetroot peel flour: Characterization, betalains profile, in silico ADMET properties and in vitro biological activity. Food Chem 2025; 476:143402. [PMID: 39965349 DOI: 10.1016/j.foodchem.2025.143402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
The use of vegetable residues as a source of bioactive components is a global trend. The production of flours reintroduces these materials into the productive chain and extend their shelf-life. Processing may reduce the diversity of pigments present in the fresh matter. We analysed a beetroot peel flour (BPF) that presented relevant protein and fibre contents and preserved the colour of the in natura beetroot (Beta vulgaris L.) due to the presence of betacyanins and betaxanthins. The bioavailability, pharmacokinetics and mutagenicity of the pigments were predicted using bioinformatics. No mutagenicity was confirmed according to the OECD guidelines. A chemoprotective effect and cancer cell anti-clone activities were observed. BPF processing ensured a good nutritional value and maintained this product as a good source of bioactive compounds and of pigments with antitumor activity, suggesting this vegetable residue as a food industry pigments source for use in the elaboration of functional products.
Collapse
Affiliation(s)
- Pedro Paulo Saldanha Coimbra
- Food and Nutrition Graduate Program, Federal University of State of Rio de Janeiro, Rio de Janeiro, Brazil; Laboratory of Environmental Mutagenesis, Department of Biophysics and Biometry, Rio de Janeiro State University, Rio de Janeiro, Brazil; Integrated Environmental Mutagenesis Laboratory, Department of Genetics and Molecular Biology, Federal University of State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adrielli de Carvalho Teixeira
- Laboratory of Environmental Mutagenesis, Department of Biophysics and Biometry, Rio de Janeiro State University, Rio de Janeiro, Brazil; Integrated Environmental Mutagenesis Laboratory, Department of Genetics and Molecular Biology, Federal University of State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Gabriel Oliveira Brito
- Laboratory of Environmental Mutagenesis, Department of Biophysics and Biometry, Rio de Janeiro State University, Rio de Janeiro, Brazil; Integrated Environmental Mutagenesis Laboratory, Department of Genetics and Molecular Biology, Federal University of State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ananda da Silva Antonio
- Laboratory for the Support of Technological Development, Chemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lays Souza
- Laboratory of Environmental Mutagenesis, Department of Biophysics and Biometry, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Henrique Marcelo Gualberto Pereira
- Laboratory for the Support of Technological Development, Chemistry Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Israel Felzenszwalb
- Laboratory of Environmental Mutagenesis, Department of Biophysics and Biometry, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Anderson Junger Teodoro
- Food and Nutrition Graduate Program, Federal University of State of Rio de Janeiro, Rio de Janeiro, Brazil; Department of Nutrition and Dietetics, Faculty of Nutrition, Fluminense Federal University, Rio de Janeiro, Brazil
| | - Carlos Fernando Araujo-Lima
- Food and Nutrition Graduate Program, Federal University of State of Rio de Janeiro, Rio de Janeiro, Brazil; Laboratory of Environmental Mutagenesis, Department of Biophysics and Biometry, Rio de Janeiro State University, Rio de Janeiro, Brazil; Integrated Environmental Mutagenesis Laboratory, Department of Genetics and Molecular Biology, Federal University of State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
6
|
Sharma S, Pandey G. Understanding the impact of triazoles on female fertility and embryo development: Mechanisms and implications. Toxicol Rep 2025; 14:101948. [PMID: 39996041 PMCID: PMC11848504 DOI: 10.1016/j.toxrep.2025.101948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025] Open
Abstract
Triazoles are among the most widely used fungicides that were launched in 1980s and are one of the most important pesticide groups used in agriculture as plant growth regulators and stress protectors. Triazoles are also frequently used in the pharmaceutical industry to treat fungal and bacterial infections as well as to treat and prevent some forms of pneumonia. Humans are normally exposed to triazoles through food, water, and medications, which raises concerns about their potential adverse effects on health. Therefore, this review was planned to examine the impact of triazole fungicides on female fertility, as well as their teratogenic and embryotoxic effects. Various search engines such as PubMed, Google Scholar, Elsevier, IEEE were used to search the relevant articles published between 2006 and 2024 using the following keywords: "azoles," "female infertility," "reproductive toxicity," "teratogenicity," "triazoles," and "embryo toxicity." The findings suggest that triazoles might negatively affect female fertility and embryonic development through multiple mechanisms including inhibition or interference with key enzymes such as CYP17A1 and CYP19A1 (aromatase) involved in steroid hormone synthesis, endocrine disruption, oxidative stress, disruption of signaling pathways, and apoptosis. This review consolidates current knowledge on the teratogenic and embryotoxic properties of triazole fungicides, providing a comprehensive understanding of their health implications and addressing critical research gaps.
Collapse
Affiliation(s)
- Sonal Sharma
- Department of Zoology, IIS (deemed to be University), Jaipur, Rajasthan 302020, India
| | - Geeta Pandey
- Department of Zoology, IIS (deemed to be University), Jaipur, Rajasthan 302020, India
| |
Collapse
|
7
|
Ogwu MC, Malík M, Tlustoš P, Patočka J. The psychostimulant drug, fenethylline (captagon): Health risks, addiction and the global impact of illicit trade. DRUG AND ALCOHOL DEPENDENCE REPORTS 2025; 15:100323. [PMID: 40151181 PMCID: PMC11946500 DOI: 10.1016/j.dadr.2025.100323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025]
Abstract
Fenethylline (street name, captagon) is a synthetic amphetamine-type stimulant that is emerging as a significant public health and security concern, particularly in the Middle East. This systematic review synthesizes original research articles, epidemiological studies, systematic reviews, policy analyses, and case reports to provide a comprehensive analysis of fenethylline's health impacts, addiction potential, and dynamics of illicit trade. Initially developed for therapeutic use, fenethylline illicit production and use have escalated, raising concern about its physiological, psychological, and socio-economic impacts. This stimulant profoundly affects the central nervous system, enhancing wakefulness, concentration, and physical stamina while inducing euphoria. These effects come at the cost of serious adverse health outcomes, particularly with prolonged or heavy use, including cardiovascular complications, neurological damage, and addiction. The dependence-forming nature of captagon contributes to escalating substance use disorders, impacting healthcare systems. Beyond its biomedical implications, fenethylline trafficking has become a global issue, with supply chains deeply intertwined with politically unstable regions where illicit economies thrive. The geopolitical dimensions of captagon's trade amplify its global security threat, influencing international relations and regional stability. This paper underscores the urgent need for systematic data collection and coordinated efforts to regulate illicit fenethylline production and distribution. Strategies such as improved surveillance, public health interventions, and international cooperation are essential to mitigate its escalating risks. Addressing this issue requires a multidisciplinary approach, integrating public health, law enforcement, and policy development to curb its impact on global health and security.
Collapse
Affiliation(s)
- Matthew Chidozie Ogwu
- Goodnight Family Department of Sustainable Development, Appalachian State University, 212 Living Learning Center, 305 Bodenheimer Drive, Boone, NC 28608, United States
| | - Matěj Malík
- Department of Agroenvironmental Chemistry and Plant Nutrition, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, Suchdol, Praha 165 00, Czech Republic
| | - Pavel Tlustoš
- Department of Agroenvironmental Chemistry and Plant Nutrition, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, Suchdol, Praha 165 00, Czech Republic
| | - Jiří Patočka
- Department of Radiology, Toxicology and Civil Protection, Faculty of Health and Social Studies, University of South Bohemia, J. Boreckého 1167/27, České Budějovice 370 11, Czech Republic
| |
Collapse
|
8
|
Quaye JA, Moni BM, Kugblenu JE, Gadda G. Oxidation of α-hydroxy acids by D-2-hydroxyglutarate dehydrogenase enzymes. Arch Biochem Biophys 2025; 768:110355. [PMID: 39993590 DOI: 10.1016/j.abb.2025.110355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/15/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
α-Hydroxy acids are naturally occurring organic molecules with various medical and industrial applications. However, some α-hydroxy acids, like D-2-hydroxyglutarate (D2HG), have been implicated in cancers and neurometabolic disorders such as D2HG aciduria. Several studies on the D2HG oxidizing enzyme D-2-hydroxyglutarate dehydrogenase (D2HGDH) from various eukaryotic and prokaryotic sources focus on the use and application of the enzyme as biosensors for detecting D2HG. A recent gene knockout study on the bacterial D2HGDH homologs from Pseudomonas stutzeri and Pseudomonas aeruginosa identified the D2HGDH to be essential for bacterial survival by driving l-serine biosynthesis. Thus, D2HGDH is a good candidate for a therapeutic target against the multidrug-resistant P. aeruginosa. However, there is no consensus on the D2HGDH catalytic mechanism, and several D2HGDH homologs have not been characterized in their structural properties, which are two crucial features for therapeutic design. P. aeruginosa D2HGDH, the most extensively studied D2HGDH homolog, is emerging as a paradigm for D2HGDH and flavoproteins with metal ions in their active site. In this review, we have explored the structures of all published D2HGDH homologs from 12 species using AlphaFold 3 and highlighted the fully conserved structure and active site topologies of all D2HGDH homologs. Additionally, evolutionary and functional studies coupled with analyses of enzymatic activities reveal that prokaryotic and eukaryotic D2HGDH homologs, diverging from two distinct ancestors, may have differentially evolved to specialize in their α-hydroxy acid catalysis. Additionally, this review identifies all D2HGDH homologs as metal and FAD-dependent enzymes that employ a metal-triggered FAD reduction in their catalysis. Elucidation of the D2HGDH mechanism will allow designing antibiotics that target these enzymes as potential therapeutics against pathogenic bacteria like P. aeruginosa in addition to the application of D2HGDH homologs as biosensors.
Collapse
Affiliation(s)
- Joanna Afokai Quaye
- Departments of Chemistry, Georgia State University, Atlanta, GA, 30302-3965, USA
| | - Bilkis Mehrin Moni
- Departments of Chemistry, Georgia State University, Atlanta, GA, 30302-3965, USA; The Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, 30302-3965, USA
| | | | - Giovanni Gadda
- Departments of Chemistry, Georgia State University, Atlanta, GA, 30302-3965, USA; Departments of Biology, Georgia State University, Atlanta, GA, 30302-3965, USA; The Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA, 30302-3965, USA.
| |
Collapse
|
9
|
Bhattacharjee A, Kumar A, Ojha PK, Kar S. Artificial intelligence to predict inhibitors of drug-metabolizing enzymes and transporters for safer drug design. Expert Opin Drug Discov 2025:1-21. [PMID: 40241626 DOI: 10.1080/17460441.2025.2491669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
INTRODUCTION Drug-metabolizing enzymes (DMEs) and transporters (DTs) play integral roles in drug metabolism and drug-drug interactions (DDIs) which directly impact drug efficacy and safety. It is well-established that inhibition of DMEs and DTs often leads to adverse drug reactions (ADRs) and therapeutic failure. As such, early prediction of such inhibitors is vital in drug development. In this context, the limitations of the traditional in vitro assays and QSAR models methods have been addressed by harnessing artificial intelligence (AI) techniques. AREAS COVERED This narrative review presents the insights gained from the application of AI for predicting DME and DT inhibitors over the past decade. Several case studies demonstrate successful AI applications in enzyme-transporter interaction prediction, and the authors discuss workflows for integrating these predictions into drug design and regulatory frameworks. EXPERT OPINION The application of AI in predicting DME and DT inhibitors has demonstrated significant potential toward enhancing drug safety and effectiveness. However, critical challenges involve the data quality, biases, and model transparency. The availability of diverse, high-quality datasets alongside the integration of pharmacokinetic and genomic data are essential. Lastly, the collaboration among computational scientists, pharmacologists, and regulatory bodies is pyramidal in tailoring AI tools for personalized medicine and safer drug development.
Collapse
Affiliation(s)
- Arnab Bhattacharjee
- Drug Discovery and Development Laboratory (DDD Lab), Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Ankur Kumar
- Drug Discovery and Development Laboratory (DDD Lab), Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Probir Kumar Ojha
- Drug Discovery and Development Laboratory (DDD Lab), Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Supratik Kar
- Chemometrics and Molecular Modeling Laboratory, Department of Chemistry and Physics, Kean University, Union, NJ, USA
| |
Collapse
|
10
|
Li S, Wang S, Zhang L, Ka Y, Zhou M, Wang Y, Tang Z, Zhang J, Wang W, Liu W. Research progress on pharmacokinetics, anti-inflammatory and immunomodulatory effects of kaempferol. Int Immunopharmacol 2025; 152:114387. [PMID: 40054326 DOI: 10.1016/j.intimp.2025.114387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/24/2025]
Abstract
Chronic inflammation (an abnormal state) and autoimmune disease (AD) can both cause multiple organ damage. AD is a heterogeneous group of diseases due to immune dysfunction. Chronic inflammation is closely related to AD and is an important part of AD. With the increasing prevalence of AD, researchers are constantly exploring new drugs with small side effects, considerable curative effects, and lower costs. Kaempferol, a flavonoid, possesses a range of biological functions, including antioxidant, anti-inflammatory, anti-neoplastic, and immunomodulatory capabilities. This compound is prevalent in a variety of plant sources, such as vegetables, fruits, and medicinal herbs traditionally used in Chinese medicine. A plethora of empirical evidence from animal-based research supports the assertion that this particular substance exhibits both anti-inflammatory and immunomodulatory effects, with the curative effect being significant and application prospects. This article mainly summarizes and discusses the pharmacokinetics, drug delivery system, and the mechanism of kaempferol on immune cells, cytokines, signaling pathways, and other aspects. This paper summarizes the existing kaempferol drug delivery system, analyzes the possibility and limitations of kaempferol as a new anti-inflammatory and immunomodulatory drug, and discusses how to apply it in clinical practice. Therefore, kaempferol can more effectively exert its anti-inflammatory and immune-modulating effects, thereby demonstrating therapeutic potential in clinical settings, while reducing patient burden.
Collapse
Affiliation(s)
- Suiran Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Siwei Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Lei Zhang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, China
| | - Yuxiu Ka
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Meijiao Zhou
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yiwen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Zhuo Tang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Jiamin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wen Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
11
|
Pratt CL, Hahn NM. Review of pharmacology of medications during pregnancy. Semin Perinatol 2025:152073. [PMID: 40221297 DOI: 10.1016/j.semperi.2025.152073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 04/14/2025]
Abstract
Ninety percent of pregnant patients take at least one medication during pregnancy. Physiological changes during pregnancy can alter drug exposure. Understanding basic physiological principles including absorption, distribution, metabolism and excretion can guide medication management during pregnancy. While physiologic changes related to medications used to treat hypothyroidism, depression and epilepsy are highlighted in this article, the principles of pharmacokinetic changes during pregnancy can be applied to other medical conditions where less data may exist. Given the complexity of pharmacology, physiological changes of pregnancy, and nuances of managing medical conditions, a team approach to managing medications in pregnancy is recommended. Comprehensive care including general and specialized physicians, pharmacists, genetic counselors and more can help provide the most appropriate care to this multifaceted patient population.
Collapse
Affiliation(s)
- Christy L Pratt
- Pharmacy Department, Kaiser Permanente Colorado, 16601 East Centretech Parkway, Aurora, CO 80011, USA; Primary Care and Obstetrics and Gynecology Departments, Kaiser Permanente Colorado, 280 Exempla Circle, Lafayette, Colorado 80026, CO, USA.
| | - Nicole M Hahn
- Pharmacy Department, Kaiser Permanente Colorado, 16601 East Centretech Parkway, Aurora, CO 80011, USA; Neurology Department, Kaiser Permanente Colorado, 1375 E 20th Avenue, Denver, Colorado 80205 CO, USA
| |
Collapse
|
12
|
Wang J, Nithianantham S, Chai SC, Jung YH, Yang L, Ong HW, Li Y, Zhang Y, Miller DJ, Chen T. Decoding the selective chemical modulation of CYP3A4. Nat Commun 2025; 16:3423. [PMID: 40210880 PMCID: PMC11985932 DOI: 10.1038/s41467-025-58749-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Drug-drug interactions associate with concurrent uses of multiple medications. Cytochrome P450 (CYP) 3A4 metabolizes a large portion of marketed drugs. To maintain the efficacy of drugs metabolized by CYP3A4, pan-CYP3A inhibitors such as ritonavir are often co-administered. Although selective CYP3A4 inhibitors have greater therapeutic benefits as they avoid inhibiting unintended CYPs and undesirable clinical consequences, the high homology between CYP3A4 and CYP3A5 has hampered the development of such selective inhibitors. Here, we report a series of selective CYP3A4 inhibitors with scaffolds identified by high-throughput screening. Structural, functional, and computational analyses reveal that the differential C-terminal loop conformations and two distinct ligand binding surfaces disfavor the binding of selective CYP3A4 inhibitors to CYP3A5. Structure-guided design of compounds validates the model and yields analogs that are selective for CYP3A4 versus other major CYPs. These findings demonstrate the feasibility to selectively inhibit CYP3A4 and provide guidance for designing better CYP3A4 selective inhibitors.
Collapse
Affiliation(s)
- Jingheng Wang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stanley Nithianantham
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sergio C Chai
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Young-Hwan Jung
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Han Wee Ong
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yong Li
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yifan Zhang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Darcie J Miller
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
13
|
Zhao X, Su S, Zhou J, Gao J, Tang X, Wen B. Metabolism and Excretion of 8-O-Acetylharpagide in Rats and Identification of Its Potential Anti-Breast Cancer Active Metabolites. Drug Des Devel Ther 2025; 19:2795-2815. [PMID: 40231196 PMCID: PMC11995923 DOI: 10.2147/dddt.s487898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 03/28/2025] [Indexed: 04/16/2025] Open
Abstract
Background Ajuga decumbens, a traditional Chinese medicine, possesses anti-breast cancer effects. Its main component, 8-O-acetylharpagide, exhibits potential anticancer activity; however, the active metabolites and mechanisms underlying its effects remain unclear. Methods The metabolism and excretion of 8-O-acetylharpagide in rats were investigated using ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry analysis of bile, urine, and feces. Active metabolites were identified and evaluated using network pharmacology, molecular docking, and Western blotting assays. Results A total of 21 metabolites were identified, with demethylation, hydrolysis, and glucuronidation being the primary metabolic pathways. M3 and M5 were identified as key metabolites, showing strong binding affinity to cancer-related targets, such as AKT1, MMP9, and STAT3. M5 displayed strong pharmacokinetic properties, including better lipid solubility and reduced polarity. Network pharmacology analysis indicated that these metabolites exert anticancer effects by modulating the PI3K/AKT signaling pathway. In vivo experiments demonstrated that oral administration of 8-O-acetylharpagide significantly inhibited the proliferation of 4T1 tumor tissues by suppressing the expression of the AKT/NF-κB/MMP9 signaling axis. This may be related to inhibition of the expression of the AKT/NF-κB/MMP9 signaling axis in 4T1 tumor tissues after metabolism of 8-O-acetylharpagide to M5 and M3. Conclusion As a prodrug, 8-O-acetylharpagide is metabolized to M5, which may subsequently exert an anti-breast cancer effect through downregulation of the AKT/NF-κB/MMP9 signaling axis. This study provides a theoretical basis for the clinical application of Ajuga decumbens in breast cancer therapy.
Collapse
MESH Headings
- Animals
- Rats
- Female
- Rats, Sprague-Dawley
- Cell Proliferation/drug effects
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Antineoplastic Agents, Phytogenic/metabolism
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/administration & dosage
- Antineoplastic Agents, Phytogenic/chemistry
- Mice
- Drug Screening Assays, Antitumor
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/metabolism
- Humans
- Molecular Structure
- Dose-Response Relationship, Drug
- Molecular Docking Simulation
- Administration, Oral
Collapse
Affiliation(s)
- Xinyu Zhao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Sijia Su
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Jingna Zhou
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Junfeng Gao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Xu Tang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| | - Binyu Wen
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, People’s Republic of China
| |
Collapse
|
14
|
Jasim SA, Altalbawy FMA, Uthirapathy S, Bishoyi AK, Ballal S, Singh A, Devi A, Yumashev A, Mustafa YF, Abosaoda MK. Regulation of immune-mediated chemoresistance in cancer by lncRNAs: an in-depth review of signaling pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04081-3. [PMID: 40202675 DOI: 10.1007/s00210-025-04081-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Resistance to cancer therapies is increasingly recognized as being influenced by long non-coding RNAs (lncRNAs), which are pivotal in regulating cellular functions and gene expression. Elucidating the intricate relationship between lncRNAs and the mechanisms underlying drug resistance is critical for advancing effective therapeutic strategies. This study offers an in-depth review of the regulatory roles lncRNAs play in various signaling and immunological pathways implicated in cancer chemoresistance. lncRNA-mediated influence on drug resistance-related signaling pathways will be presented, including immune evasion mechanisms and other essential signaling cascades. Furthermore, the interplay between lncRNAs and the immune landscape will be dissected, illustrating their substantial impact on the development of chemoresistance. Overall, the potential of lncRNA-mediated signaling networks as a therapeutic strategy to combat cancer resistance has been highlighted. This review reiterates the fundamental role of lncRNAs in chemoresistance and proposes promising avenues for future research and the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq.
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Mosco, Russia
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
15
|
Hashem H, Abdelfattah S, Hassan HM, Al-Emam A, Alqarni M, Alotaibi G, Radwan IT, Kaur K, Rao DP, Bräse S, Alkhammash A. Discovery of a novel 4-pyridyl SLC-0111 analog targeting tumor-associated carbonic anhydrase isoform IX through tail-based design approach with potent anticancer activity. Front Chem 2025; 13:1571646. [PMID: 40255643 PMCID: PMC12006758 DOI: 10.3389/fchem.2025.1571646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/11/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction: Carbonic anhydrase IX (CA IX) is a tumor-associated enzyme involved in cancer progression and survival. Targeting CA IX with selective inhibitors like SLC-0111 has shown therapeutic potential. This study aimed to develop a novel 4-pyridyl analog (Pyr) of SLC-0111 with enhanced anticancer activity. Methods: Pyr was synthesized using a tail-based design and characterized by NMR. Its cytotoxicity was tested against cancer and normal cell lines. CA inhibition, cell cycle effects, apoptosis induction, and protein expression changes were evaluated. Molecular docking and ADMET predictions assessed binding and drug-like properties. Results and Discussion: Pyr showed selective cytotoxicity toward cancer cells and potent CA IX inhibition. It induced G0/G1 arrest, apoptosis, and modulated p53, Bax, and Bcl-2 levels. Docking confirmed strong CA IX binding, and ADMET analysis indicated good oral bioavailability. These results support Pyr as a promising anticancer candidate.
Collapse
Affiliation(s)
- Hamada Hashem
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Sohag University, Sohag, Egypt
| | - Shadwa Abdelfattah
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Merit University (MUE), Sohag, Egypt
| | - Hesham M. Hassan
- Department of Pathology, College of Medicine, King Khalid University, Asir, Saudi Arabia
| | - Ahmed Al-Emam
- Department of Pathology, College of Medicine, King Khalid University, Asir, Saudi Arabia
| | - Mohammed Alqarni
- Department of Pharmaceutical chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Ghallab Alotaibi
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
| | - Ibrahim Taha Radwan
- Supplementary General Sciences Department, Faculty of Oral and Dental Medicine, Future University in Egypt, Cairo, Egypt
| | - Kirandeep Kaur
- Department of Chemistry, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Devendra Pratap Rao
- Coordination Chemistry Laboratory, Department of Chemistry, Dayanand Anglo-Vedic (PG) College, Kanpur, Uttar Pradesh, India
| | - Stefan Bräse
- Institute of Biological and Chemical Systems, Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Abdullah Alkhammash
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
| |
Collapse
|
16
|
Fashe MM, Tiley JB, Lee CR. Mechanisms of altered hepatic drug disposition during pregnancy: small molecules. Expert Opin Drug Metab Toxicol 2025; 21:445-462. [PMID: 39992297 PMCID: PMC11961323 DOI: 10.1080/17425255.2025.2470792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/01/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Pregnancy alters the systemic exposure and clearance of many hepatically cleared drugs that are commonly used by obstetric patients. Understanding the molecular mechanisms underlying the changes in factors that affect hepatic drug clearance (blood flow, protein binding, and intrinsic clearance) is essential to more precisely predict systemic drug exposure and dose requirements in obstetric patients. AREAS COVERED This review (1) summarizes the anatomic, physiologic, and biochemical changes in maternal hepatic, cardiovascular, endocrine, and renal systems relevant to hepatic drug clearance and (2) reviews the molecular mechanisms underlying the altered hepatic metabolism and intrinsic clearance of drugs during pregnancy via a comprehensive PubMed search. It also identifies knowledge gaps in the molecular mechanisms and factors that modulate hepatic drug clearance during pregnancy. EXPERT OPINION Pharmacokinetic studies have shown that pregnancy alters systemic exposure, protein binding, and clearance of many drugs during gestation in part due to pregnancy-associated decreases in plasma albumin, increases in organ blood flow, and changes in the activity of drug-metabolizing enzymes (DMEs) and transporters. The changes in the activity of certain DMEs and transporters during pregnancy are likely driven by hormonal-changes that inhibit their activity or alter the expression of these proteins through activation of transcription factors.
Collapse
Affiliation(s)
- Muluneh M. Fashe
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Jacqueline B. Tiley
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Craig R. Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| |
Collapse
|
17
|
Yao L, Wang L, Zhang R, Soukas AA, Wu L. The direct targets of metformin in diabetes and beyond. Trends Endocrinol Metab 2025; 36:364-372. [PMID: 39227192 DOI: 10.1016/j.tem.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 09/05/2024]
Abstract
Metformin, an oral antihyperglycemic drug that has been in use for over 60 years, remains a first-line therapy for type 2 diabetes (T2D). Numerous studies have suggested that metformin promotes health benefits beyond T2D management, including weight loss, cancer prevention and treatment, and anti-aging, through several proposed mechanistic targets. Here we discuss the established effects of metformin and the progress made in identifying its direct targets. Additionally, we emphasize the importance of elucidating the structural bases of the drug and its direct targets. Ultimately, this review aims to highlight the current state of knowledge regarding metformin and its related emerging discoveries, while also outlining critical future research directions.
Collapse
Affiliation(s)
- Luxia Yao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Lei Wang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Runshuai Zhang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Alexander A Soukas
- Center for Genomic Medicine and Diabetes Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Lianfeng Wu
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
18
|
Coppinger C, Anderson PL. Considerations for drug-drug interactions between long-acting antiretrovirals and immunosuppressants for solid organ transplantation. Expert Opin Drug Metab Toxicol 2025; 21:343-346. [PMID: 39757464 DOI: 10.1080/17425255.2024.2448970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025]
Affiliation(s)
- Corwin Coppinger
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Peter L Anderson
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
19
|
Choi YJ, Saravanakumar K, Joo JH, Nam B, Park Y, Lee S, Park S, Li Z, Yao L, Kim Y, Irfan N, Cho N. Metabolomics and network pharmacology approach to identify potential bioactive compounds from Trichoderma sp. against oral squamous cell carcinoma. Comput Biol Chem 2025; 115:108348. [PMID: 39864356 DOI: 10.1016/j.compbiolchem.2025.108348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/21/2024] [Accepted: 01/05/2025] [Indexed: 01/28/2025]
Abstract
This study aimed to profile metabolites from five Trichoderma strains and assess their cytotoxic and pharmacological activities, particularly targeting oral squamous cell carcinoma (OSCC). UHPLC-TOF-MS analysis revealed the presence of 25 compounds, including heptelidic acid, viridiol isomers, and sorbicillinol from the different Trichoderma extracts. Pharmacokinetic analysis showed moderate permeability and low interaction with P-glycoprotein, suggesting good drug absorption with minimal interference in cellular uptake. ADME-Tox analysis indicated limited inhibition of cytochrome P450 enzymes, low renal clearance, which are favorable for maintaining therapeutic levels. Toxicity predictions revealed some compounds with potential mutagenicity, but low hepatotoxicity and skin sensitization risks. Network pharmacology identified MAPK1 as a key target for oral cancer, and molecular docking and induced fit docking studies demonstrated strong binding affinities of Trichoderma metabolites, including stachyose and harzianol, to MAPK1. In addition, molecular dynamics (MD) simulations confirmed stable interactions. In vitro studies on NIH3T3 and YD-10B cells showed significant cytotoxicity, particularly with extracts CNU-05-001 (IC50:10.15 µg/mL) and CNU-02-009 (10.00 µg/mL) against YD-10B cells. These findings underscore the potential of Trichoderma metabolites in drug discovery, particularly for cancer therapies.
Collapse
Affiliation(s)
- Young Ji Choi
- Division of bioresources bank, Honam National Institute of Biological Resources, 99, Gohadoan-gil, Mokpo-si, Jeollanam-do 58762, Republic of Korea.
| | - Kandasamy Saravanakumar
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Jae-Hyoung Joo
- Division of bioresources bank, Honam National Institute of Biological Resources, 99, Gohadoan-gil, Mokpo-si, Jeollanam-do 58762, Republic of Korea.
| | - Bomi Nam
- Division of bioresources bank, Honam National Institute of Biological Resources, 99, Gohadoan-gil, Mokpo-si, Jeollanam-do 58762, Republic of Korea.
| | - Yuna Park
- Division of bioresources bank, Honam National Institute of Biological Resources, 99, Gohadoan-gil, Mokpo-si, Jeollanam-do 58762, Republic of Korea.
| | - Soyeon Lee
- Division of bioresources bank, Honam National Institute of Biological Resources, 99, Gohadoan-gil, Mokpo-si, Jeollanam-do 58762, Republic of Korea.
| | - SeonJu Park
- Metropolitan Seoul Center, Korea Basic Science Institute (KBSI), Seoul 03759, Republic of Korea.
| | - Zijun Li
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Lulu Yao
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Yunyeong Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Navabshan Irfan
- Crescent School of Pharmacy, B.S Abdur Rahman Crescent Institute of Science and Technology, Chennai 600048, India.
| | - Namki Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea; Elicure, 12, Gyeongyeol-ro 17 beon-gil, Seo-gu, Gwangju, Republic of Korea.
| |
Collapse
|
20
|
Ngan DK, Sakamuru S, Zhao J, Xia M, Ferguson SS, Reif DM, Simeonov A, Huang R. Application of cytochrome P450 enzyme assays to predict p53 inducers and AChE inhibitors that require metabolic activation. Toxicol Appl Pharmacol 2025; 499:117315. [PMID: 40180188 DOI: 10.1016/j.taap.2025.117315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Metabolically active compounds can cause toxicity which would otherwise be undetected using traditional in vitro assays with limited proficiency for xenobiotic metabolism. Introduction of liver microsomes to assay systems enables enhanced identification of compounds that require biotransformation to induce toxicity. Previously, metabolically active compounds from the Tox21 10 K compound library were identified using assays probing two targets, p53 and acetylcholinesterase (AChE), in the presence and absence of human or rat liver microsomes, due to the established roles of cytochrome P450 (CYP) enzymes in human drug metabolism. To further explore the role of metabolic activation, the activities of the identified metabolically active compounds were evaluated against five CYP enzymes: CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4. CYP bioactivities were found to be highly predictive (>80 % accuracy) of compounds that required metabolic activation in these assays. Chemical features significantly enriched in metabolically active compounds, as well as chemical features that were specific for each of the five CYPs, were identified. Product use exposures of the metabolically active compounds were examined in this study, with "pesticides" appearing to be the largest category that may produce harmful metabolites. Additionally, the compound interactions with different CYPs were assessed and frequencies for both classes of compounds, drugs and environmental chemicals, were found to be proportionally similar across the five CYP isoforms.
Collapse
Affiliation(s)
- Deborah K Ngan
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Srilatha Sakamuru
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Jinghua Zhao
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Menghang Xia
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Stephen S Ferguson
- Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, NC, USA
| | - David M Reif
- Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Research Triangle Park, NC, USA
| | - Anton Simeonov
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Ruili Huang
- Division of Pre-clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD 20850, USA.
| |
Collapse
|
21
|
Makaro A, Kasprzak Z, Jaczynska M, Swierczynski M, Salaga M. Role of Cytochromes P450 in Intestinal Barrier Function: Possible Involvement in the Pathogenesis of Leaky Gut Syndrome. Dig Dis Sci 2025; 70:1293-1304. [PMID: 39971825 DOI: 10.1007/s10620-025-08873-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/14/2025] [Indexed: 02/21/2025]
Abstract
The intestinal barrier constitutes the largest surface of the human body communicating with the external environment. Alterations affecting elements of intestinal wall may lead to increased intestinal permeability and resulting translocation of bacteria or its components to the bloodstream in the form of the "leaky gut syndrome" (LGS). One of the most common causes of LGS is the disruption of tight junctions (TJ) maintained by tight junction proteins (TJP). LGS and associated alterations in TJP are observed in numerous gastrointestinal (GI) diseases, including inflammatory bowel diseases (IBD) such as Crohn's disease (CD) and ulcerative colitis (UC). Current literature indicates the key role of LGS in many pathological processes, further emphasizing the need for effective pharmacological approaches to treat this syndrome. One of the potential pharmacological targets in LGS treatment are members of the cytochrome P450 (CYP450) superfamily. By affecting intestinal permeability, they may lead to LGS development. It was found that the expression of CYP8B1 synthesizing cholic acid and CYP26 degrading all-trans retinoic acid indirectly influence TJs. CYP2E1 responsible for the metabolism of a wide variety of chemicals, including ethanol, plays a crucial role in the impairment of the intestinal wall. Contrarily, the overexpression of CYP27B1 has a protective effect on the intestinal integrity. CYP1A1, CYP2A6, CYP2J2 and CYP3A were also suggested to influence the GI tract, through their capability to metabolize serotonin, nicotine, endocannabinoids and gemcitabine, respectively. This review summarizes the findings on the role of CYP450 isoforms in intestinal hyperpermeability and their potential involvement in the pathophysiology of LGS.
Collapse
Affiliation(s)
- Adam Makaro
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Zuzanna Kasprzak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Maria Jaczynska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Mikolaj Swierczynski
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Maciej Salaga
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland.
| |
Collapse
|
22
|
Kamal MV, Prabhu K, Sharan K, Pai A, Chakrabarty S, Damerla RR, Shetty PS, Belle VS, Rao M, Kumar NAN. Investigation of the Molecular Mechanisms of Paraoxonase-2 Mediated Radiotherapy and Chemotherapy Resistance in Oral Squamous Cell Carcinoma. Clin Transl Sci 2025; 18:e70201. [PMID: 40134131 PMCID: PMC11936840 DOI: 10.1111/cts.70201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/27/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a common form of cancer, with 390,000 new cases estimated for 2022. OSCC has a poor prognosis, largely due to a high recurrence rate and resistance to therapy. Cancer cells develop resistance to standard therapy owing to various factors, such as genetic predispositions, alterations in the apoptotic pathway coupled with DNA repair pathways, drug efflux, and drug detoxification. This review is aimed at exploring the crucial role of paraoxonase 2 (PON2) in conferring resistance to chemotherapy and radiotherapy in OSCC cells. PON2, an antioxidant enzyme, protects cancer cells from the oxidative stress caused by these treatments. By influencing apoptotic pathways and DNA repair mechanisms, PON2 can reduce the effectiveness of therapy. This review is an attempt to explore the complex molecular mechanisms modulated by PON2, such as the mitigation of oxidative stress, enhancement of DNA repair, apoptosis regulation, drug efflux modulation, and drug detoxification, which decrease treatment efficacy.
Collapse
Affiliation(s)
- Mehta Vedant Kamal
- Department of Surgical OncologyManipal Comprehensive Cancer Care Center, Kasturba Medical College, Manipal, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Krishnananda Prabhu
- Department of BiochemistryKasturba Medical College, Manipal, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Krishna Sharan
- Department of Radiotherapy and OncologyKS Hegde Medical Academy, Nitte (Deemed to Be University)MangaluruKarnatakaIndia
| | - Ananth Pai
- Department of Medical OncologyManipal Comprehensive Cancer Care Center, Kasturba Medical College, Manipal, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Sanjiban Chakrabarty
- Department of Public Health and GenomicsManipal School of Life Sciences, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Rama Rao Damerla
- Department of Medical GeneticsKasturba Medical College, Manipal, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Preethi S. Shetty
- Department of Surgical OncologyManipal Comprehensive Cancer Care Center, Kasturba Medical College, Manipal, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Vijetha Shenoy Belle
- Department of BiochemistryKasturba Medical College, Manipal, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Mahadev Rao
- Department of Pharmacy PracticeCenter for Translational Research, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Naveena A. N. Kumar
- Department of Surgical OncologyManipal Comprehensive Cancer Care Center, Kasturba Medical College, Manipal, Manipal Academy of Higher EducationManipalKarnatakaIndia
| |
Collapse
|
23
|
Saini RS, Vaddamanu SK, Dermawan D, Bavabeedu SS, Khudaverdyan M, Mosaddad SA, Heboyan A. In Silico Docking of Medicinal Herbs Against P. gingivalis for Chronic Periodontitis Intervention. Int Dent J 2025; 75:1113-1135. [PMID: 39127518 PMCID: PMC11976485 DOI: 10.1016/j.identj.2024.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/25/2024] [Accepted: 06/29/2024] [Indexed: 08/12/2024] Open
Abstract
OBJECTIVE This study aimed to explore the therapeutic potential of medicinal herbs for chronic periodontitis by examining the molecular interactions between specific herbal compounds and the heme-binding protein of Porphyromonas gingivalis, a key pathogen involved in the disease. METHODS The crystal structure of heme-binding protein was obtained from the Protein Data Bank. Herbal compounds were identified through an extensive literature review. Molecular docking simulations were performed to predict binding affinities, followed by Absorption, Distribution, Metabolism, and Excretion (ADME) parameter prediction. Drug-likeness was assessed based on Lipinski's Rule of Five, and pharmacophore modeling was conducted to identify key molecular interactions. RESULTS The molecular docking simulations revealed that chelidonine, rotenone, and myricetin exhibited significant binding affinities to the heme-binding protein, with docking scores of -6.5 kcal/mol, -6.4 kcal/mol, and -6.1 kcal/mol, respectively. These compounds formed stable interactions with key amino acid residues within the binding pocket. ADME analysis indicated that all 3 compounds had favourable pharmacokinetic properties, with no violations of Lipinski's rules and minimal predicted toxicity. Pharmacophore modeling further elucidated the interaction profiles, highlighting specific hydrogen bonds and hydrophobic interactions critical for binding efficacy. CONCLUSIONS Chelidonine, rotenone, and myricetin emerged as promising therapeutic candidates for chronic periodontitis due to their strong binding affinities, favorable ADME profiles, and lack of significant toxicity. The detailed pharmacophore modeling provided insights into the molecular mechanisms underpinning their inhibitory effects on the heme-binding protein of P. gingivalis. These findings suggest that these compounds have the potential for further development as effective treatments for chronic periodontitis. Future research should focus on in vitro and in vivo validation of these findings to confirm the efficacy and safety of these compounds in biological systems.
Collapse
Affiliation(s)
- Ravinder S Saini
- Department of Dental Technology, COAMS, King Khalid University, Abha, Saudi Arabia
| | | | - Doni Dermawan
- Applied Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Shashit Shetty Bavabeedu
- Department of Restorative Dental Sciences, College of Dentistry, King Khalid University, Abha, Saudi Arabia
| | - Margarita Khudaverdyan
- The Center for Excellence in Dental Education, Yerevan State Medical University after Mkhitar Heratsi, Yerevan, Armenia
| | - Seyed Ali Mosaddad
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India; Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Conservative Dentistry and Bucofacial Prosthesis, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain.
| | - Artak Heboyan
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India; Department of Prosthodontics, Faculty of Stomatology, Yerevan State Medical University after Mkhitar Heratsi, Yerevan, Armenia; Department of Prosthodontics, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Bathaei P, Imenshahidi M, Vahdati-Mashhadian N, Hosseinzadeh H. Effects of Crocus sativus and its active constituents on cytochrome P450: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03525-6. [PMID: 40167627 DOI: 10.1007/s00210-024-03525-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 10/07/2024] [Indexed: 04/02/2025]
Abstract
Cytochrome P450 (CYP) enzymes play an important role in the biotransformation of drugs and endogenous substances. Clinical medications and herbal remedies can either enhance or inhibit the activity of CYP enzymes, leading to potential drug interactions between herbal supplements and prescribed medications. Such interactions can lead to serious consequences, especially for drugs with a narrow therapeutic index, such as digoxin, warfarin, and cyclosporine A. In this review article, we provide an updated review of the impact of saffron, and its active constituents, safranal and crocin, on the 12 major human CYP enzymes and possible drug interactions between saffron and prescription drugs. The available evidence indicates that saffron and its active constituents affect the expression or activity of some CYP isoforms, including the CYP1A1/2, CYP3A4, and CYP2E1 subfamily. Considering the important role of these CYPs in the biotransformation of frequently prescribed medications and the activation of procarcinogen into carcinogenic metabolites, it can be expected that the consumption of saffron and its active constituents may influence the pharmacokinetics and toxicity of several substances. In particular, given the critical role of CYP3A4 in drug metabolism, and saffron's inhibitory impact on this CYP enzyme, it appears that saffron's most significant interaction is linked to its inhibition of CYP3A4. In addition, the inhibitory effect of saffron on CYP1A1/2, and CYP2E1 expression can play a role in the chemopreventive effect of this herbal medicine. Additional research is crucial for evaluating the clinical significance of these interactions in patients who consume saffron along with prescription drugs and determining the dose that can lead to drug interactions.
Collapse
Affiliation(s)
- Pooneh Bathaei
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Imenshahidi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, P.O.Box: 1365-91775, Mashhad, Iran
| | - Nasser Vahdati-Mashhadian
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, P.O.Box: 1365-91775, Mashhad, Iran.
| |
Collapse
|
25
|
Vosála O, Krátký J, Matoušková P, Rychlá N, Štěrbová K, Raisová Stuchlíková L, Vokřál I, Skálová L. Biotransformation of anthelmintics in nematodes in relation to drug resistance. Int J Parasitol Drugs Drug Resist 2025; 27:100579. [PMID: 39827513 PMCID: PMC11787565 DOI: 10.1016/j.ijpddr.2025.100579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
In all organisms, the biotransformation of xenobiotics to less toxic and more hydrophilic compounds represents an effective defense strategy. In pathogens, the biotransformation of drugs (used for their elimination from the host) may provide undesirable protective effects that could potentially compromise the drug's efficacy. Accordingly, increased drug deactivation via accelerated biotransformation is now considered as one of the mechanisms of drug resistance. The present study summarizes the current knowledge regarding the biotransformation of anthelmintics, specifically drugs used to treat mainly nematodes, a group of parasites that are a significant health concern for humans and animals. The main biotransformation enzymes are introduced and their roles in anthelmintics metabolism in nematodes are discussed with a particular focus on their potential participation in drug resistance. Similarly, the inducibility of biotransformation enzymes with sublethal doses of anthelmintics is presented in view of its potential contribution to drug resistance development. In the conclusion, the main tasks awaiting scientists in this area are outlined.
Collapse
Affiliation(s)
- Ondřej Vosála
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Heyrovského 1203, Hradec Králové, CZ-500 05, Czech Republic
| | - Josef Krátký
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Heyrovského 1203, Hradec Králové, CZ-500 05, Czech Republic
| | - Petra Matoušková
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Heyrovského 1203, Hradec Králové, CZ-500 05, Czech Republic
| | - Nikola Rychlá
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Heyrovského 1203, Hradec Králové, CZ-500 05, Czech Republic
| | - Karolína Štěrbová
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Heyrovského 1203, Hradec Králové, CZ-500 05, Czech Republic
| | - Lucie Raisová Stuchlíková
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Heyrovského 1203, Hradec Králové, CZ-500 05, Czech Republic
| | - Ivan Vokřál
- Department of Pharmacology and Toxicology, Charles University in Prague, Faculty of Pharmacy, Heyrovského 1203, Hradec Králové, CZ-500 05, Czech Republic
| | - Lenka Skálová
- Department of Biochemical Sciences, Charles University in Prague, Faculty of Pharmacy, Heyrovského 1203, Hradec Králové, CZ-500 05, Czech Republic.
| |
Collapse
|
26
|
Liu J, Xu W, Liu Y, Zhang Q. Reproductive and developmental toxicology of nitrosamines. Toxicol Res (Camb) 2025; 14:tfaf051. [PMID: 40248818 PMCID: PMC12001771 DOI: 10.1093/toxres/tfaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 04/19/2025] Open
Abstract
With the development of science and technology and the acceleration of industrialization, environmental pollution is becoming more and more serious, and the global fertility rate is decreasing every year, which makes people pay more attention to reproductive health. Nitrosamines are a kind of easy to contact food pollutants, widely exist in pickled food (10.2-14.8 mg/kg) and contaminated water sources (10-150 ng/L), etc. They have been confirmed to be carcinogenic, but the reproductive and developmental toxic effects of nitrosamines have not been systematically reported. Based on relevant researches, the classification, distribution and metabolism kinetics of nitrosamines were summarized in this review. In addition, nitrosamines can inhibit testosterone synthesis (Leydig cells) and spermatogenesis (spermatogenic cells) in F0 male, and reduce ovary functions in F0 female, finally induce parental reproductive toxic effects. Meanwhile, the effects of parental (including maternal pregnancy, paternal) nitrosamine exposure on offspring development (such as cancer susceptibility) and related research deficiencies were summarized. To sum up, this paper systematically reviewed the reproductive and developmental toxic effects caused by exposure to nitrosamines, enabling people to fully understand the negative effects of nitrosamines on the body, so as to effectively avoid and reduce intake in daily life, and at the same time provide a theoretical and literature basis for guiding the healthy life and maintaining fertility.
Collapse
Affiliation(s)
- Juan Liu
- Department of Clinical Pharmacy, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, WuYang road, number 158, Enshi, Hubei 445000, China
| | - Wei Xu
- Department of Neurology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, WuYang road, number 157, Enshi, Hubei 445000, China
| | - Yi Liu
- School of basic medical science, Wuhan university DongHu road, number 115, 430070
| | - Qi Zhang
- Department of Clinical Pharmacy, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, WuYang road, number 158, Enshi, Hubei 445000, China
- Hubei Selenium and Human Health Institute, WuYang road, number 158, Enshi, Hubei 445000, China
| |
Collapse
|
27
|
Dhankhar S, Kumar J, Chauhan S, Zahoor I, Wani SN, Saini M, Borsha JA, Yasmin S, Ansari MY. Flavonoids and flavonoid-based nanoparticles for the treatment of arthritis. Inflammopharmacology 2025:10.1007/s10787-025-01722-0. [PMID: 40156677 DOI: 10.1007/s10787-025-01722-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/12/2025] [Indexed: 04/01/2025]
Abstract
Arthritis is an autoimmune disorder that predominantly causes inflammation and impacts peripheral joints. Even though immunosuppressive and NSAIDs or non-steroidal anti-inflammatory medicines are implemented for the management of this disorder, sbut they carry some severe side effects along with them. Therefore, society requires treatment with fewer side effects and powerful anti-arthritic properties, such as flavonoids. These are the most prevalent phenolic compounds found in nature that have potent antioxidant, and immunomodulatory activity and there are several bioactive flavonoids that carry potent anti-inflammatory properties. Nevertheless, only a handful has reached their clinical use. Still, in both clinical and preclinical models of arthritis, flavonoids found in the diet have been shown to reduce swelling in joints and arthritis symptoms. There are only a few scientific studies regarding their mechanisms of action in arthritis. However, the arthritic potential of dietary flavonoids is insufficient because of their limited solubility, absorption, and fast metabolism. Nanocarriers may enhance the bioavailability of flavonoids. This review examines the therapeutic effects of the most prevalent and abundant flavonoid groups on arthritis. Specifically, the modes of action of the most important flavonoids on the chemical messengers in the body that contribute to the signalling of joint inflammation-related indicators of arthritis are discussed in more detail.
Collapse
Affiliation(s)
- Sanchit Dhankhar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Jatin Kumar
- Chitkara University School of Pharmacy, Chitkara University, Solan, Himachal Pradesh, India
| | - Samrat Chauhan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Ishrat Zahoor
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, Haryana, India.
| | - Shahid Nazir Wani
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Monika Saini
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, Haryana, India
| | - Jamila Akter Borsha
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, Haryana, India
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Mohammad Yousuf Ansari
- M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, Haryana, India.
- Ibne Seena College of Pharmacy, Azmi Vidya Nagri, Anjhi Shahabad, Hardoi, 241124, Uttar Pradesh (U.P.), India.
| |
Collapse
|
28
|
Khdhiri E, Haffouz A, HadjKacem B, Dhouib I, Amor IB, Jerbi A, Gargouri J, Sahli E, Abid M, Allouche N, Gargouri A, Khemakhem B, Abid S, Ammar H. Novel Coumarin-Hydrazone Hybrids as Potential Antiplatelet Agents: Microwave-Assisted Synthesis, Characterization, In Vitro and In Silico Biological Evaluations and Toxicity Assessment. Chem Biodivers 2025:e202500101. [PMID: 40146071 DOI: 10.1002/cbdv.202500101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/04/2025] [Accepted: 03/25/2025] [Indexed: 03/28/2025]
Abstract
This work focuses on the synthesis and characterization of a new series of coumarin-hydrazone derivatives, as well as the evaluation of their inhibitory effects on platelet aggregation induced by adenosine diphosphate (ADP), arachidonic acid (AA), and collagen. Compounds 10 and 11 exhibit a significant inhibition of ADP-induced platelet aggregation. The AA-induced aggregation pathway was inhibited by compound 11 (45%), while none showed an inhibitory effect against collagen-induced aggregation. In addition, both compounds inhibited platelet binding to fibrinogen, CD62-P expression, and glycoprotein IIb/IIIa activation. The interaction of these compounds with their potential targets, P2Y12 and COX-1, was studied using a molecular docking approach. Furthermore, the ADMET properties of the compounds selected were evaluated in silico using Swiss ADME and ProTox-II tools. Selected active compounds demonstrated interesting pharmacokinetic and drug properties, indicating a favorable ADMET profile. In vitro and in silico results are in accordance. Toxicity on lymphocytes, erythrocytes, and platelets was evaluated for the two selected molecules and found to be safe. In summary, this study proposes two novel coumarin-hydrazone derivatives as new potential treatments to prevent cardiovascular events by acting on platelet adhesion, activation, and aggregation.
Collapse
Affiliation(s)
- Emna Khdhiri
- Laboratoire de Chimie Appliquée "Hétérocycles Corps Gras & Polymères", Faculté des Sciences, Université de Sfax, Sfax, Tunisia
| | - Asma Haffouz
- Laboratory of Molecular Biotechnology of Eucaryotes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Basma HadjKacem
- Laboratory of Molecular Biotechnology of Eucaryotes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
- Department of Life Sciences, Faculty of Sciences of Gafsa, University of Gafsa, Gafsa, Tunisia
| | - Ines Dhouib
- Laboratory of Plant Biotechnology, Faculty of Sciences of Sfax, BP 1171, University of Sfax, Sfax, Tunisia
| | - Ikram Ben Amor
- Laboratory of Hematology (LR19SP04), Medical Faculty of Sfax. University of Sfax, Sfax, Tunisia
| | - Amira Jerbi
- Laboratory of Hematology (LR19SP04), Medical Faculty of Sfax. University of Sfax, Sfax, Tunisia
| | - Jalel Gargouri
- Laboratory of Hematology (LR19SP04), Medical Faculty of Sfax. University of Sfax, Sfax, Tunisia
| | - Emna Sahli
- Analytical service provider unit, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Majdi Abid
- Department of Chemistry, College of Science, Jouf University, Sakaka, Kingdom of Saudi Arabia
| | - Noureddine Allouche
- Laboratory of Organic Chemistry LR17ES08 (Natural Substances Team), Faculty of Sciences of Sfax, University of Sfax, Sfax, Tunisia
| | - Ali Gargouri
- Laboratory of Molecular Biotechnology of Eucaryotes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Bassem Khemakhem
- Laboratory of Plant Biotechnology, Faculty of Sciences of Sfax, BP 1171, University of Sfax, Sfax, Tunisia
| | - Souhir Abid
- Department of Chemistry, College of Science, Jouf University, Sakaka, Kingdom of Saudi Arabia
| | - Houcine Ammar
- Laboratoire de Chimie Appliquée "Hétérocycles Corps Gras & Polymères", Faculté des Sciences, Université de Sfax, Sfax, Tunisia
| |
Collapse
|
29
|
Cai H, Xing X, Su Y, Yang C. Innovative applications and future perspectives of chromatography-mass spectrometry in drug research. Front Pharmacol 2025; 16:1529468. [PMID: 40206083 PMCID: PMC11979114 DOI: 10.3389/fphar.2025.1529468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
Chromatography coupled with mass spectrometry (MS) has emerged as a cornerstone analytical technique in drug research. Over the years, advancements in chromatography-MS have significantly enhanced its capabilities, leading to improved sensitivity, specificity, and throughput. This review explores the innovative applications of chromatography-MS in drug research, particularly focusing on its role in drug absorption, distribution, metabolism, excretion (ADME), toxicity evaluation, and personalized medicine. It also addresses the future perspectives of this powerful technique, including challenges and potential solutions, and highlights how emerging trends such as high spatial resolution imaging and multimodal integration could revolutionize drug discovery and development. Through these innovations, chromatography-MS promises to contribute substantially to the development of more effective, safer, and personalized therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Ying Su
- Department of Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Chunhui Yang
- Department of Clinical Laboratory, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
30
|
Bosch EL, Sommer IEC, Touw DJ. The influence of female sex and estrogens on drug pharmacokinetics: what is the evidence? Expert Opin Drug Metab Toxicol 2025:1-11. [PMID: 40109018 DOI: 10.1080/17425255.2025.2481891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION Pharmacological research has traditionally been skewed toward male subjects, leading to uniform treatment guidelines for both men and women that assume similar drug pharmacokinetics across sexes. This oversight contributes to women experiencing adverse drug reactions on average twice as often as men. More recent studies have revealed significant pharmacokinetic differences between the sexes, partly due to different sex hormone levels. Additionally, intraindividual differences in women have been observed due to fluctuating estrogen levels, impacting important aspects of drug pharmacokinetics. AREAS COVERED This review highlights key sex differences in drug pharmacokinetics, focusing on absorption, distribution, metabolism, and excretion. A particular emphasis is placed on the role of cytochrome P450 (CYP) and uridine diphosphate-glucuronosyltransferase (UGT) enzymes in drug metabolism, and on the role of P-glycoprotein (P-gp). The impact of estrogens is reviewed by exploring how drug pharmacokinetics change over the menstrual cycle, before and after menopause, and with estrogen-containing medications. EXPERT OPINION Personalized dosing based on sex and estrogen levels is important for improving treatment outcomes in female drug users. Clinical trials of drugs likely affected by these factors should incorporate pharmacokinetic studies that distinguish between sexes and evaluate the impact of estrogens, aiming to develop optimized dosing regimens.
Collapse
Affiliation(s)
- E L Bosch
- Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| | - I E C Sommer
- Department of Psychiatry and Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - D J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, Netherlands
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
| |
Collapse
|
31
|
Chevalley T, Dübi M, Fumeaux L, Merli MS, Sarre A, Schaer N, Simeoni U, Yzydorczyk C. Sexual Dimorphism in Cardiometabolic Diseases: From Development to Senescence and Therapeutic Approaches. Cells 2025; 14:467. [PMID: 40136716 PMCID: PMC11941476 DOI: 10.3390/cells14060467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
The global incidence and prevalence of cardiometabolic disorders have risen significantly in recent years. Although lifestyle choices in adulthood play a crucial role in the development of these conditions, it is well established that events occurring early in life can have an important effect. Recent research on cardiometabolic diseases has highlighted the influence of sexual dimorphism on risk factors, underlying mechanisms, and response to therapies. In this narrative review, we summarize the current understanding of sexual dimorphism in cardiovascular and metabolic diseases in the general population and within the framework of the Developmental Origins of Health and Disease (DOHaD) concept. We explore key risk factors and mechanisms, including the influence of genetic and epigenetic factors, placental and embryonic development, maternal nutrition, sex hormones, energy metabolism, microbiota, oxidative stress, cell death, inflammation, endothelial dysfunction, circadian rhythm, and lifestyle factors. Finally, we discuss some of the main therapeutic approaches, responses to which may be influenced by sexual dimorphism, such as antihypertensive and cardiovascular treatments, oxidative stress management, nutrition, cell therapies, and hormone replacement therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Catherine Yzydorczyk
- Developmental Origins of Health and Disease (DOHaD) Laboratory, Division of Pediatrics, Department Woman-Mother-Child, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; (T.C.); (M.D.); (L.F.); (M.S.M.); (A.S.); (N.S.)
| |
Collapse
|
32
|
da Fonsêca DV, Rocha JS, da Silva PR, de Sá Novaes Pereira HN, dos Santos LVN, de Santana MAD, Alves AF, Pontes AHO, de Souza Gomes J, Felipe CFB, de Sousa DP, Scotti MT, Scotti L. 4-Hydroxycoumarin Exhibits Antinociceptive and Anti-Inflammatory Effects Through Cytokine Modulation: An Integrated In Silico and In Vivo Study. Int J Mol Sci 2025; 26:2788. [PMID: 40141429 PMCID: PMC11943305 DOI: 10.3390/ijms26062788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/01/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Chronic pain significantly impacts quality of life and is often accompanied by inflammation, a natural bodily response that can become harmful when excessive. The orofacial region is commonly affected, making effective treatment crucial. However, current drugs often cause undesirable side effects, highlighting the need for new pharmacological alternatives. 4-hydroxycoumarin (4-HC), a natural compound, has shown promising antinociceptive and anti-inflammatory effects, but studies confirming its specific properties are limited. In silico analyses suggest that 4-HC exhibits favorable pharmacokinetic characteristics, not interacting with P-glycoprotein and successfully crossing the blood-brain barrier. Molecular docking studies indicate that its effects may be mediated through NMDAR or by inhibiting iNOS. Our study assessed the antinociceptive and anti-inflammatory effects of 4-HC in animal models at doses of 25, 50, and 75 mg/kg. 4-HC significantly reduced abdominal contortions induced by acetic acid and decreased nociceptive rubbing in orofacial pain models induced by formalin, glutamate, and capsaicin. Interactions with opioid receptors were not observed, suggesting that 4-HC's antinociceptive effect does not involve this pathway. Additionally, 4-HC reduced paw edema induced by carrageenan and significantly decreased leukocyte migration and TNF-α levels. These findings highlight the therapeutic potential of 4-HC and warrant further investigation into its mechanisms.
Collapse
Affiliation(s)
- Diogo Vilar da Fonsêca
- Postgraduate Program in Biosciences, Federal University of the São Francisco Valley (UNIVASF), Pernambuco 56304-917, PE, Brazil; (D.V.d.F.); (J.S.R.)
| | - Juliana Sousa Rocha
- Postgraduate Program in Biosciences, Federal University of the São Francisco Valley (UNIVASF), Pernambuco 56304-917, PE, Brazil; (D.V.d.F.); (J.S.R.)
| | - Pablo R. da Silva
- Postgraduate Program of Dentistry (PPGO), Health Sciences Center, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil;
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil;
- Postgraduate Program in Natural Synthetic and Bioactive Products, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | | | | | | | - Alan F. Alves
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.F.A.); (A.H.O.P.); (J.d.S.G.); (M.T.S.)
| | - Adiel H. O. Pontes
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.F.A.); (A.H.O.P.); (J.d.S.G.); (M.T.S.)
| | - Joás de Souza Gomes
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.F.A.); (A.H.O.P.); (J.d.S.G.); (M.T.S.)
| | - Cícero F. Bezerra Felipe
- Psychopharmacology Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil;
- Postgraduate Program in Natural Synthetic and Bioactive Products, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil
| | - Damião Pergentino de Sousa
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil;
| | - Marcus T. Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.F.A.); (A.H.O.P.); (J.d.S.G.); (M.T.S.)
| | - Luciana Scotti
- Cheminformatics Laboratory, Institute of Drugs and Medicines Research, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil; (A.F.A.); (A.H.O.P.); (J.d.S.G.); (M.T.S.)
| |
Collapse
|
33
|
Zhu Y, Zhuang W, Cheng H. Strategies to Enhance Protein Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:6457-6470. [PMID: 40052814 PMCID: PMC11924232 DOI: 10.1021/acs.langmuir.4c04636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Therapeutic proteins play a crucial role in modern healthcare. However, the rapid clearance of proteins in the circulation system poses a significant threat to their therapeutic efficacy. The generation of anti-drug antibodies expedites drug clearance, resulting in another challenge to overcome in protein delivery. Several methods to increase the circulation half-lives of these proteins and to minimize their immunogenicity have been developed. This Review discusses the causes of protein clearance in the body, evaluates the FDA-approved strategies to prolong protein circulation, and highlights recent progress in the field. Additionally, the strengths and drawbacks of these methods and our perspectives for advancing protein delivery are provided.
Collapse
Affiliation(s)
- Yucheng Zhu
- Department of Materials Science and Engineering, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Weisi Zhuang
- Department of Materials Science and Engineering, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Hao Cheng
- Department of Materials Science and Engineering, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
34
|
Durairaj P, Liu ZL. Brain Cytochrome P450: Navigating Neurological Health and Metabolic Regulation. J Xenobiot 2025; 15:44. [PMID: 40126262 PMCID: PMC11932283 DOI: 10.3390/jox15020044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
Human cytochrome P450 (CYP) enzymes in the brain represent a crucial frontier in neuroscience, with far-reaching implications for drug detoxification, cellular metabolism, and the progression of neurodegenerative diseases. The brain's complex architecture, composed of interconnected cell types and receptors, drives unique neuronal signaling pathways, modulates enzyme functions, and leads to distinct CYP gene expression and regulation patterns compared to the liver. Despite their relatively low levels of expression, brain CYPs exert significant influence on drug responses, neurotoxin susceptibility, behavior, and neurological disease risk. These enzymes are essential for maintaining brain homeostasis, mediating cholesterol turnover, and synthesizing and metabolizing neurochemicals, neurosteroids, and neurotransmitters. Moreover, they are key participants in oxidative stress responses, neuroprotection, and the regulation of inflammation. In addition to their roles in metabolizing psychotropic drugs, substances of abuse, and endogenous compounds, brain CYPs impact drug efficacy, safety, and resistance, underscoring their importance beyond traditional drug metabolism. Their involvement in critical physiological processes also links them to neuroprotection, with significant implications for the onset and progression of neurodegenerative diseases. Understanding the roles of cerebral CYP enzymes is vital for advancing neuroprotective strategies, personalizing treatments for brain disorders, and developing CNS-targeting therapeutics. This review explores the emerging roles of CYP enzymes, particularly those within the CYP1-3 and CYP46 families, highlighting their functional diversity and the pathological consequences of their dysregulation on neurological health. It also examines the potential of cerebral CYP-based biomarkers to improve the diagnosis and treatment of neurodegenerative disorders, offering new avenues for therapeutic innovation.
Collapse
Affiliation(s)
- Pradeepraj Durairaj
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL 32310, USA
- Department of Chemical and Biomedical Engineering, Florida A&M University, Tallahassee, FL 32310, USA
| | - Zixiang Leonardo Liu
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL 32310, USA
- Department of Chemical and Biomedical Engineering, Florida A&M University, Tallahassee, FL 32310, USA
- Institute for Successful Longevity, Florida State University, Tallahassee, FL 32310, USA
| |
Collapse
|
35
|
Ben Zichri- David S, Shkuri L, Ast T. Pulling back the mitochondria's iron curtain. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:6. [PMID: 40052109 PMCID: PMC11879881 DOI: 10.1038/s44324-024-00045-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/09/2024] [Indexed: 03/09/2025]
Abstract
Mitochondrial functionality and cellular iron homeostasis are closely intertwined. Mitochondria are biosynthetic hubs for essential iron cofactors such as iron-sulfur (Fe-S) clusters and heme. These cofactors, in turn, enable key mitochondrial pathways, such as energy and metabolite production. Mishandling of mitochondrial iron is associated with a spectrum of human pathologies ranging from rare genetic disorders to common conditions. Here, we review mitochondrial iron utilization and its intersection with disease.
Collapse
Affiliation(s)
| | - Liraz Shkuri
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001 Israel
| | - Tslil Ast
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001 Israel
| |
Collapse
|
36
|
Zhu J, Jiang C, Wang F, Tao MY, Wang HX, Sun Y, Hui HX. NOX4 Suppresses Ferroptosis Through Regulation of the Pentose Phosphate Pathway in Colorectal Cancer. Curr Med Sci 2025:10.1007/s11596-025-00013-7. [PMID: 40029499 DOI: 10.1007/s11596-025-00013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 11/23/2024] [Accepted: 11/28/2024] [Indexed: 03/05/2025]
Abstract
OBJECTIVE Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are known as major sources of reactive oxygen species (ROS), yet their role in regulating cellular antioxidative metabolism and ferroptosis is unclear. This study assessed the expression and clinical relevance of NOXs across pan-cancer and investigated the role of NOX4 in colorectal cancer progression METHODS: We analyzed transcriptomic and survival data from The Cancer Genome Atlas (TCGA) for NOXs across 22 types of solid tumors. A CRISPR library targeting NOXs was developed for potential therapeutic target screening in colorectal cancer cells (CRCs). Techniques such as CRISPR-knockout cell lines, 1,2-13C-glucose tracing, PI staining, BrdU assays, and coimmunoprecipitation were employed to elucidate the function of NOX4 in CRCs. RESULTS NOX4 emerged as a key therapeutic target for colorectal cancer from TCGA data. CRISPR screening highlighted its essential role in CRC survival, with functional experiments confirming that NOX4 upregulation promotes cell survival and proliferation. The interaction of NOX4 with glucose‑6‑phosphate dehydrogenase (G6PD) was found to enhance the pentose phosphate pathway (PPP), facilitating ROS clearance and protecting CRCs against ferroptosis. CONCLUSIONS This study identified NOX4 as a novel ferroptosis suppressor and a therapeutic target for the treatment of colorectal cancer. The findings suggest that a coupling between NADPH oxidase enzyme NOX4 and the PPP regulates ferroptosis and reveal an accompanying metabolic vulnerability for therapeutic targeting in colorectal cancer.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Chao Jiang
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Fan Wang
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Ming-Yue Tao
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Hai-Xiao Wang
- Department of General Surgery, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Yuan Sun
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Hong-Xia Hui
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China.
| |
Collapse
|
37
|
Gerlach S, Maruf AA, Shaheen SM, McCloud R, Heintz M, McAusland L, Arnold PD, Bousman CA. Prevalence Estimates of Cytochrome P450 Phenoconversion in Youth Receiving Pharmacotherapy for Mental Health Conditions. Clin Pharmacol Ther 2025; 117:670-675. [PMID: 39686785 PMCID: PMC11835427 DOI: 10.1002/cpt.3534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/08/2024] [Indexed: 12/18/2024]
Abstract
Pharmacogenetics-predicted drug metabolism may not match clinically observed metabolism due to a phenomenon known as phenoconversion. Phenoconversion can occur when an inhibitor or inducer of a drug-metabolizing enzyme is present. Although estimates of phenoconversion in adult populations are available, prevalence estimates in youth populations are limited. To address this gap, we estimated the prevalence of phenoconversion in 1281 youth (6-24 years) receiving pharmacotherapy for mental health conditions and who had pharmacogenetics testing completed for four genes (CYP2B6, CYP2C19, CYP2D6, CYP3A4). Self-reported medication and cannabidiol/cannabis use were collected at the time of pharmacogenetics testing. Nearly, half (46%) of the cohort was estimated to be phenoconverted for one of the four genes examined. Comparison of metabolizer phenotype frequencies before and after adjustment for phenoconversion showed significantly more youth had actionable phenotypes for CYP2C19 (60.3% vs. 69.1%; P =< 0.001), CYP2D6 (49.3% vs. 63.0%; P =< 0.001), and CYP3A4 (8.5% vs.12.2%; P = 0.003) after phenoconversion adjustment. Of youth who were phenoconverted, 24% had a change in their metabolizer phenotype that would lead to current pharmacogenetics-based prescribing guidelines recommending a change to standard prescribing (dose adjustment, alternative medication). Our findings indicate a high prevalence of cytochrome P450 phenoconversion among youth receiving pharmacotherapy for mental health conditions. Adjustment for phenoconversion should be considered when implementing pharmacogenetics testing in youth populations to improve the clinical utility of this testing in practice.
Collapse
Affiliation(s)
- Samuel Gerlach
- Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Abdullah Al Maruf
- College of Pharmacy, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
- Children's Hospital Research Institute of ManitobaWinnipegManitobaCanada
- The Mathison Centre for Mental Health Research & Education, Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of PsychiatryUniversity of CalgaryCalgaryAlbertaCanada
| | - Sarker M. Shaheen
- The Mathison Centre for Mental Health Research & Education, Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of PsychiatryUniversity of CalgaryCalgaryAlbertaCanada
| | - Ryden McCloud
- The Mathison Centre for Mental Health Research & Education, Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Madison Heintz
- The Mathison Centre for Mental Health Research & Education, Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of Medical GeneticsUniversity of CalgaryCalgaryAlbertaCanada
| | - Laina McAusland
- The Mathison Centre for Mental Health Research & Education, Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of PsychiatryUniversity of CalgaryCalgaryAlbertaCanada
- Department of Medical GeneticsUniversity of CalgaryCalgaryAlbertaCanada
| | - Paul D. Arnold
- The Mathison Centre for Mental Health Research & Education, Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of PsychiatryUniversity of CalgaryCalgaryAlbertaCanada
- Department of Medical GeneticsUniversity of CalgaryCalgaryAlbertaCanada
- Department of Physiology & PharmacologyUniversity of CalgaryCalgaryAlbertaCanada
- Alberta Children's Hospital Research InstituteUniversity of CalgaryCalgaryAlbertaCanada
| | - Chad A. Bousman
- The Mathison Centre for Mental Health Research & Education, Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Department of PsychiatryUniversity of CalgaryCalgaryAlbertaCanada
- Department of Medical GeneticsUniversity of CalgaryCalgaryAlbertaCanada
- Department of Physiology & PharmacologyUniversity of CalgaryCalgaryAlbertaCanada
- Alberta Children's Hospital Research InstituteUniversity of CalgaryCalgaryAlbertaCanada
- Department of Community Health SciencesUniversity of CalgaryCalgaryAlbertaCanada
| |
Collapse
|
38
|
Lykkesfeldt J, Carr AC, Tveden-Nyborg P. The pharmacology of vitamin C. Pharmacol Rev 2025; 77:100043. [PMID: 39986139 DOI: 10.1016/j.pharmr.2025.100043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 01/14/2025] [Indexed: 02/24/2025] Open
Abstract
Ascorbic acid, the reduced form of vitamin C, is a ubiquitous small carbohydrate. Despite decades of focused research, new metabolic functions of this universal electron donor are still being discovered and add to the complexity of our view of vitamin C in human health. Although praised as an unsurpassed water-soluble antioxidant in plasma and cells, the most interesting functions of vitamin C seem to be its roles as specific electron donor in numerous biological reactions ranging from the well-known hydroxylation of proline to cofactor for the epigenetic master regulators ten-eleven translocation enzymes and Jumonji domain-containing histone-lysine demethylases. Some of these functions may have important implications for disease prevention and treatment and have spiked renewed interest in, eg, vitamin C's potential in cancer therapy. Moreover, some fundamental pharmacokinetic properties of vitamin C remain to be established including if other mechanisms than passive diffusion governs the efflux of ascorbate anions from the cell. Taken together, there still seems to be much to learn about the pharmacology of vitamin C and its role in health and disease. This review explores new avenues of vitamin C and integrates our present knowledge of its pharmacology. SIGNIFICANCE STATEMENT: Vitamin C is involved in multiple biological reactions of which most are essential to human health. Hundreds of millions of people are considered deficient in vitamin C according to accepted guidelines, but little is known about the long-term consequences. Although the complexity of vitamin C's physiology and pharmacology has been widely disregarded in clinical studies for decades, it seems clear that a deeper understanding of particularly its pharmacology holds the key to unravel and possibly exploit the potential of vitamin C in disease prevention and therapy.
Collapse
Affiliation(s)
- Jens Lykkesfeldt
- Section of Biomedicine, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Anitra C Carr
- Nutrition in Medicine Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Pernille Tveden-Nyborg
- Section of Biomedicine, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
39
|
Yang X, Huang J, Wang J, Sun H, Li J, Li S, Tang YE, Wang Z, Song Q. Effect of glucose selenol on hepatic lipid metabolism disorder induced by heavy metal cadmium in male rats. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159589. [PMID: 39674492 DOI: 10.1016/j.bbalip.2024.159589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
This study used 24 male rats to determine the protective effects of a new selenium molecule (glucose selenol) on cadmium (Cd) induced hepatic toxicity. The rats were randomly divided into four groups: control group, Cd group, Cd + 0.15 Se group, and Cd + 0.4 Se group. The results showed that glucose selenol supplementation alleviated the adverse impact of Cd on lipid metabolism, including decreased serum triacylglycerol and cholesterol levels. Transcriptome analysis revealed that, compared to the control group, Cd changed the expression of 1379 genes - discernibly affecting lipid metabolism pathways. Proteomic analysis primarily indicated alterations in lipid metabolism-related pathways. In conclusion, glucose selenol restored lipid metabolism disorders induced by Cd, thus rescuing hepatic damage. This integrated analysis identified the influence of glucose selenol on Cd-induced hepatic toxicity and provided its potential application prospects in alleviating the impact of heavy metal pollution, such as Cd, on human health.
Collapse
Affiliation(s)
- Xinyi Yang
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China.
| | - Jinzhou Huang
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - Juan Wang
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - Huimin Sun
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - JinJin Li
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - Shunfeng Li
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - Yun-E Tang
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - Zhi Wang
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China.
| | - Qisheng Song
- Division of Plant Sciences and Technology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
40
|
Gust KA, Amar SK, Gut CP, Styles RM, Karna RR, James RA, Holtzapple DM, Stricker JL, McInturf SM, Phillips EA, Honnold C, Luo X, Mumy KL, Mattie DR, Chappell MA, Mayo ML. Multi-disciplinary investigation identifies increased potency of ethyl-parathion inhaled within a soil-dust matrix to cause acetylcholinesterase-dependent molecular impacts. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2025; 114:104615. [PMID: 39710123 DOI: 10.1016/j.etap.2024.104615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/13/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
Neurotoxicity investigations of inhaled organophosphorus pesticide (OP), ethyl-parathion (EP), were conducted in Sprague Dawley rats comparing exposures to EP volatilized at 0, 1, 10, and 20 mg/m3 versus EP incorporated into soil dust (5 mg/m3) at 0, 0.0095, 0.09, and 0.185 mg/mg3. All exposures were sublethal, caused no respiratory effects, and no effects on balance and coordination behavior. Both volatilized and dust-incorporated EP exposures significantly decreased acetylcholinesterase (AChE) activity in plasma and hippocampus tissue. Correspondingly, plasma and hippocampal dopamine levels spiked in these exposures suggesting compensatory cholinergic / dopaminergic signal balancing. The EP exposures significantly increased expression of pro-inflammatory genes, including MAPK-14, IL6, IL1β, and TNF-α, while global RNA-seq results identified significant enrichment of inflammation, oxidative stress, and apoptosis pathways. Remarkably, dust-incorporated EP impacted similar molecular endpoints as volatilized EP but at concentrations two orders of magnitude lower highlighting potentially increased potency of EP incorporated into soil dust.
Collapse
Affiliation(s)
- Kurt A Gust
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States.
| | - Saroj K Amar
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States; Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, United States
| | - Chet P Gut
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Renee M Styles
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Ranju R Karna
- Credere Associates LLC, Westbrook, ME located at US Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - R Arden James
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - David M Holtzapple
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Joshua L Stricker
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Shawn M McInturf
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Elizabeth A Phillips
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Cary Honnold
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - Xiao Luo
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Karen L Mumy
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, OH, United States
| | - David R Mattie
- Air Force Research Laboratory/711 HPW, Wright-Patterson Air Force Base, OH, United States
| | - Mark A Chappell
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| | - Michael L Mayo
- U S Army Engineer Research and Development Center, Vicksburg, MS, United States
| |
Collapse
|
41
|
Zhang M, Zhang L, Suo B, Wei Y, Xu Y, Jiang M, Dong J, Li X, Song Z, Liu D. Distribution Characteristics and Impacting Factors of Drug CYP Enzymes and Transporters in the Gastrointestinal Tract of Chinese Healthy Subjects. Clin Pharmacol Ther 2025; 117:676-689. [PMID: 39582241 DOI: 10.1002/cpt.3497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024]
Abstract
The abundance of drug metabolic enzymes (DMEs) and transporters (DTs) in the human gastrointestinal tract significantly affects xenobiotic exposure in the circulating system, the basis of these compounds acting on humans. However, accurately predicting individual exposure in healthy subjects remains challenging due to limited data on protein levels throughout the gastrointestinal tract within the same individuals and inadequate assessment of factors influencing these levels. Therefore, we conducted a clinical study to obtain biopsy samples from 8 different gastrointestinal segments in 24 healthy Chinese volunteers. Concurrently, blood and fecal samples were collected for genotypic analysis and fecal microbiota metagenomic sequencing. Using an optimized LC-MS/MS method, we quantified the absolute protein abundance of CYP2C9, CYP2C19, CYP2D6, CYP3A4, P-gp, and BCRP from the stomach to the colon. Our results revealed significant regional differences in protein expression: CYP3A4 was the most abundant in the small intestine, whereas CYP2C9 was predominantly found in the colon. CYP2D6 was primarily located in the ileum, while other DMEs/DTs showed higher concentrations in the jejunum. Meanwhile, the enzyme abundance in the small intestine and colon and the relative ratio of transporters in different regions to the jejunum were accurately calculated, providing valuable data for refining the physiological parameters in the virtual gastrointestinal tract of Chinese healthy population in PBBMs. Additionally, BMI, IBW, sex, age, genotype, and fecal microbiota were identified as critical factors influencing the protein levels of these DMEs/DTs throughout the gastrointestinal tract, with notable regional differences. Consequently, this study provides a unique foundation for understanding xenobiotic absorption in humans.
Collapse
Affiliation(s)
- Miao Zhang
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Lei Zhang
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
| | - Baojun Suo
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Yudong Wei
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Yue Xu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
- College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Muhan Jiang
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Jing Dong
- Shimadzu China Innovation Center, Beijing, China
| | - Xiaodong Li
- Shimadzu China Innovation Center, Beijing, China
| | - Zhiqiang Song
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
- Institute of Medical Innovation, Peking University Third Hospital, Beijing, China
| |
Collapse
|
42
|
Liu X, Chen G, Yang Y, Liu F, Wu G, An L, Tang T, Zhang J. Comprehensive multi-omics analysis reveals the mechanism of hepatotoxicity induced by Emilia sonchifolia (L.) DC. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119371. [PMID: 39826791 DOI: 10.1016/j.jep.2025.119371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Emilia sonchifolia is a very widely used traditional Chinese medicine, with the efficacy of heat-clearing, detoxicating, dissipating blood stasis, reducing swelling and relieving pain. As a widely used traditional miao herb, Emilia sonchifolia is often used to treat upper respiratory tract infections, oral ulcer, pneumonia, mastitis, enteritis, bacillum, urinary tract infection, sores, eczema, falls and injuries, etc. In fact, many cases of liver injury caused by Emilia sonchifolia have been reported clinically. However, the mechanisms underlying hepatotoxicity induced by Emilia sonchifolia remain poorly understood. AIM OF THE STUDY This study aimed to systematically evaluate the acute and chronic hepatotoxicity of water extract from Emilia sonchifolia, identify its hepatotoxic metabolites, and elucidate the potential mechanisms underlying Emilia sonchifolia-induced hepatotoxicity. MATERIAL AND METHOD The chemical components in the water extract of Emilia sonchifolia were identified using mass spectrometry. The acute toxicity study was conducted by orally administering a gradient dose of water extract of Emilia sonchifolia ranging from 0 to 37.6 g/kg. Mice were orally administered a water extract of Emilia sonchifolia at a dose of 13.72 g/kg/d for 14 days to induce liver injury. The hepatotoxicity was evaluated using hematoxylin and eosin staining as well as enzyme-linked immunosorbent assay (ELISA). The mechanisms of hepatotoxicity were explored through transcriptomics, proteomics, and metabolomics analysis. Meanwhile, the core pathways related to the hepatotoxicity of Emilia sonchifolia were analyzed and validated using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and ELISA. RESULT The present study demonstrates that the water extract of Emilia sonchifolia can induce hepatotoxicity in mice. We found that the water extract of Emilia sonchifolia contained hepatotoxic pyrrolizidine alkaloids, such as seneciphyllin, senecionine, rinderine, echimidine, retrorsine and echimidine N-oxide. A dose of 19.20 g/kg or higher of the water extract of Emilia sonchifolia caused acute liver failure and death in mice. A dose of 13.72 g/kg or lower of the water extract of Emilia sonchifolia produced dose-dependent acute hepatotoxicity. Meanwhile, a dose of 13.72 g/kg of the water extract from Emilia sonchifolia induced chronic hepatotoxicity in mice. Furthermore, the results of liver transcriptomics, proteomics, and metabolomics indicate that the mechanism of hepatotoxicity induced by the water extract of Emilia sonchifolia is associated with ferroptosis caused by abnormalities in bile acid accumulation, lipid and bilirubin accumulation, and glutathione metabolism. The validation experiment results demonstrate that in mice treated with the water extract of Emilia sonchifolia, the gene levels of Cyp2c29, Cyp3a41a and Ugt2b1 decreased while the gene level of Hsd3b3 increased. In mice treated with a water extract of Emilia sonchifolia, the levels of total bilirubin, direct bilirubin, total bile acids, alkaline phosphatase, and γ-glutamyl transferase were significantly elevated. Additionally, in mice treated with a water extract of Emilia sonchifolia, the levels of malondialdehyde increased while the levels of catalase and superoxide dismutase decreased. CONCLUSION In conclusion, our results suggest that the water extract of Emilia sonchifolia can cause hepatotoxicity in mice. The chronic hepatotoxicity of Emilia sonchifolia is associated with Cyp2c29, Cyp3a41a, Ugt2b1, and Hsd3b3-mediated cholestasis, oxidative stress, and ferroptosis.
Collapse
Affiliation(s)
- Xin Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Gongzhen Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China.
| | - Yuqi Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Feng Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Guangzhou Wu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Lili An
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Ting Tang
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China.
| | - Jinqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
43
|
Holliman AG, Mackay L, Biancardi VC, Tao YX, Foradori CD. Atrazine's effects on mammalian physiology. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2025:1-40. [PMID: 40016167 DOI: 10.1080/10937404.2025.2468212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Atrazine is a chlorotriazine herbicide that is one of the most widely used herbicides in the USA and the world. For over 60 years atrazine has been used on major crops including corn, sorghum, and sugarcane to control broadleaf and grassy weed emergence and growth. Atrazine has exerted a major economic and environmental impact over that time, resulting in reduced production costs and increased conservation tillage practices. However, widespread use and a long half-life led to a high prevalence of atrazine in the environment. Indeed, atrazine is the most frequent herbicide contaminant detected in water sources in the USA. Due to its almost ubiquitous presence and questions regarding its safety, atrazine has been well-studied. First reported to affect reproduction with potential disruptive effects which were later linked to the immune system, cancer, stress response, neurological disorders, and cardiovascular ailments in experimental models. Atrazine impact on multiple interwoven systems broadens the significance of atrazine exposure. The endeavor to uncover the mechanisms underlying atrazine-induced dysfunction in mammals is ongoing, with new genetic and pharmacological targets being reported. This review aims to summarize the prominent effects of atrazine on mammalian physiology, primarily focusing on empirical studies conducted in lab animal models and establish correlations with epidemiological human studies when relevant. In addition, current common patterns of toxicity and potential underlying mechanisms of atrazine action will be examined.
Collapse
Affiliation(s)
- Anna G Holliman
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Laci Mackay
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Vinicia C Biancardi
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Chad D Foradori
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| |
Collapse
|
44
|
Thankan RS, Thomas E, Weldemariam MM, Purushottamachar P, Huang W, Kane MA, Zhang Y, Ambulos N, Wang BD, Weber D, Njar VCO. Thermal proteome profiling and proteome analysis using high-definition mass spectrometry demonstrate modulation of cholesterol biosynthesis by next-generation galeterone analog VNPP433-3β in castration-resistant prostate cancer. Mol Oncol 2025. [PMID: 40007440 DOI: 10.1002/1878-0261.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 02/03/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Cholesterol (CHOL) homeostasis is significantly modulated in prostate cancer (PCa) suggesting an active role in PCa development and progression. Several studies indicate a strong correlation between elevated CHOL levels and increased PCa risk and severity. Inhibition of CHOL biosynthesis at different steps, including lanosterol synthase (LSS), has shown significant efficacy against both hormone-dependent and castration-resistant PCa. Earlier, we reported proteasomal degradation of androgen receptor (AR)/AR-Vs and Mnk1/2 as the primary mechanisms of action of VNPP433-3β in inhibiting PCa cell proliferation and tumor growth. Through thermal proteome profiling, comparative proteomics and cellular thermal shift assay, we identified VNPP433-3β's ancillary effect of lowering CHOL by binding to LSS and lanosterol 14-alpha demethylase, potentially inhibiting CHOL biosynthesis in PCa cells and tumors. Additionally, in conjunction with our previously reported transcriptome analysis, proteomics reveals that VNPP433-3β modulated upstream regulators and pathways critical for PCa stem cell maintenance and recurrence. The inhibition of CHOL biosynthesis by VNPP433-3β reinforces its multifaceted effects in PCa across all stages, highlighting its potential as a single-agent therapy. Achieving reduced CHOL levels aligns with better treatment outcomes, further substantiating VNPP433-3β's therapeutic potential.
Collapse
Affiliation(s)
- Retheesh S Thankan
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- Isoprene Pharmaceuticals, Inc., University of Maryland Baltimore BioPark, Baltimore, MD, USA
| | - Elizabeth Thomas
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- The Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mehari M Weldemariam
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Puranik Purushottamachar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- The Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Yuji Zhang
- Division of Biostatistics and Bioinformatics, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Nicholas Ambulos
- Department of Microbiology and Immunology, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Bi-Dar Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD, USA
| | - David Weber
- The Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vincent C O Njar
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- Isoprene Pharmaceuticals, Inc., University of Maryland Baltimore BioPark, Baltimore, MD, USA
- The Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
45
|
Zhao Y, Wu Z, Li J, Qi Y, Zhang X, Shen C. The key role of cytochrome P450s in the biosynthesis of plant derived natural products. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2025; 222:109695. [PMID: 40015195 DOI: 10.1016/j.plaphy.2025.109695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/05/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
Cytochrome P450 (CYP450 or CYP450, abbreviated as CYP450) represents a large family of self-oxidizable heme proteins, belonging to the class of monooxygenases, and is named because of the specific absorption peak at 450 nm in its ferrous/CO-bound complex. Cytochrome P450 has a wide spectrum of substrates and a rich variety of catalytic reactions, plays an important role in drug metabolism, natural product biosynthesis, and biocatalysis industry. In the biosynthesis of plant natural products, it plays an important role, especially in the downstream synthesis pathway and structural modification after skeleton synthesis. There are abundant natural products from plants, including terpenes, alkaloids, flavonoids, steroidal saponins, etc., which have great development value in the medical field. In the biosynthetic pathways of these natural products, cytochrome P450 enzymes often play an important role. They can serve as rate-limiting enzymes in the biosynthetic pathways or as modifying enzymes for the structural diversity of compounds. So, a deeper understanding of cytochrome P450 enzymes in the natural product synthesis pathway will enhance the development of natural products and advance the study of their synthetic biology. This review offers an overview of the biosynthesis of key medicinal natural products, with a particular focus on the critical role of cytochrome P450 enzymes in key catalytic steps. It also highlights recent advancements in the research of natural product biosynthesis and synthetic biology.
Collapse
Affiliation(s)
- Yawen Zhao
- College of Chemical & Pharmaceutical Engineering, Hebei University of Science & Technology, Shijiazhuang, China
| | - Zhenzhen Wu
- College of Chemical & Pharmaceutical Engineering, Hebei University of Science & Technology, Shijiazhuang, China
| | - Jiayao Li
- College of Chemical & Pharmaceutical Engineering, Hebei University of Science & Technology, Shijiazhuang, China
| | - Yaoxing Qi
- College of Chemical & Pharmaceutical Engineering, Hebei University of Science & Technology, Shijiazhuang, China
| | - Xiaoxiao Zhang
- College of Chemical & Pharmaceutical Engineering, Hebei University of Science & Technology, Shijiazhuang, China
| | - Chen Shen
- College of Chemical & Pharmaceutical Engineering, Hebei University of Science & Technology, Shijiazhuang, China; State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science & Technology, Shijiazhuang, China.
| |
Collapse
|
46
|
Oladeji SM, Conteh DN, Bello LA, Adegboyega AE, Shokunbi OS. Rational Design and Optimization of Novel PDE5 Inhibitors for Targeted Colorectal Cancer Therapy: An In Silico Approach. Int J Mol Sci 2025; 26:1937. [PMID: 40076563 PMCID: PMC11899891 DOI: 10.3390/ijms26051937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths globally. Current treatment options including chemotherapy and targeted therapies face challenges such as resistance and toxicity. Cyclic guanosine monophosphate (cGMP)-specific phosphodiesterase 5 (PDE5) has emerged as a promising target for CRC therapy due to its role in regulating cellular processes like proliferation and apoptosis. This study focuses on the in silico design of a novel PDE5 inhibitor MS01 derived from the lead compound exisulind which has shown apoptotic effects but failed due to hepatotoxicity. Using Schrödinger's Induced Fit Docking (IFD) and molecular dynamic simulations, MS01 was designed to enhance binding affinity and reduce toxicity. The docking studies showed that MS01 exhibits stronger interactions with key PDE5 residues, particularly Gln817 and Phe820. ADMET predictions indicate favorable pharmacokinetic profiles, with reduced risk of drug-drug interactions and improved bioavailability. Toxicity assessments revealed that MS01 and its analogs have moderate toxicity, with MS20 and MS21 demonstrating lower hepatotoxicity compared to exisulind. These findings suggest that MS01 has the potential to be a more effective and safer PDE5 inhibitor for CRC treatment pending further experimental validation.
Collapse
Affiliation(s)
| | - Deborah Ngozi Conteh
- Department of Medicine and Surgery, University of Ilorin, P.M.B, Ilorin 1515, Nigeria;
| | - Lukman Abidemi Bello
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA; (S.M.O.); (L.A.B.)
| | - Abayomi Emmanuel Adegboyega
- Department of Biological Sciences, Structural and Computational Biology, Purdue University, West Lafayette, IN 47907, USA;
- Jaris Computational Biology Centre, Bioinformatics Unit, Jos 930241, Nigeria
| | - Oluwatosin Sarah Shokunbi
- Department of Basic Sciences, Babcock University, School of Science and Technology, Babcock University, Ilishan-Remo 121003, Nigeria
| |
Collapse
|
47
|
Hoffmann M, Sauer J, Book M, Ermler TF, Fischer P, Gerlach S, Beltagi K, Morgenroth A, Alexa R, Kranz J, Saar M. Mechanism of Action and Interaction of Garlic Extract and Established Therapeutics in Prostate Cancer. Int J Mol Sci 2025; 26:1777. [PMID: 40004239 PMCID: PMC11855885 DOI: 10.3390/ijms26041777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
A detailed characterization of the mechanism of action of garlic extract (GE) on prostate cancer (PCa) cells is essential to ensure its safe use as a complementary therapy, particularly when combined with established treatments. A case report highlighted the potential benefits of GE in PCa management. A patient diagnosed with PCa, presenting an initial prostate-specific antigen (PSA) of 11.8 ng/mL, maintained PSA levels between 3.5 and 6 ng/mL for over 14 years with daily GE intake. To study GE's anti-proliferative effects and interactions with established therapeutics, healthy prostate epithelial cells (PNT2) and PCa cells (LNCaP, PC3, VCaP) were treated with GE. Proliferation, Integrin β1 pattern, DNA-damage, as well as androgen receptor (AR) and Cytochrome P450 (CYP450) expression were investigated. GE reduced the proliferation of LNCaP and PC3 cells compared to healthy PNT2 cells but had contrary effects on VCaP cells. The combination of GE with standard therapies, including chemotherapy, androgen deprivation therapy (ADT), and Poly-(ADP-ribose)-Polymerase inhibitors (PARPi), reduced the efficacy of these treatments in tumor cells, potentially due to the GE-induced upregulation of the metabolic enzyme CYP2C9 in PCa cell lines. These findings indicate that while GE has anti-proliferative effects, the use of highly concentrated natural extracts must be carefully assessed by expert physicians on a case-by-case basis, especially when combined with established therapies.
Collapse
Affiliation(s)
- Marco Hoffmann
- Department of Urology and Pediatric Urology, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.F.E.); (K.B.); (R.A.)
- Center for Integrated Oncology (CIO), University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Jana Sauer
- Department of Urology and Pediatric Urology, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.F.E.); (K.B.); (R.A.)
- Center for Integrated Oncology (CIO), University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Marie Book
- Department of Urology and Pediatric Urology, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.F.E.); (K.B.); (R.A.)
- Center for Integrated Oncology (CIO), University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Thomas Frank Ermler
- Department of Urology and Pediatric Urology, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.F.E.); (K.B.); (R.A.)
- Center for Integrated Oncology (CIO), University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Petra Fischer
- Center for Integrated Oncology (CIO), University Hospital RWTH Aachen, 52074 Aachen, Germany;
- Department of Otorhinolaryngology-Head and Neck Surgery, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Sven Gerlach
- Department of Urology and Pediatric Urology, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.F.E.); (K.B.); (R.A.)
- Center for Integrated Oncology (CIO), University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Kareem Beltagi
- Department of Urology and Pediatric Urology, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.F.E.); (K.B.); (R.A.)
- Center for Integrated Oncology (CIO), University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Radu Alexa
- Department of Urology and Pediatric Urology, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.F.E.); (K.B.); (R.A.)
- Center for Integrated Oncology (CIO), University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Jennifer Kranz
- Department of Urology and Pediatric Urology, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.F.E.); (K.B.); (R.A.)
- Center for Integrated Oncology (CIO), University Hospital RWTH Aachen, 52074 Aachen, Germany;
| | - Matthias Saar
- Department of Urology and Pediatric Urology, University Hospital RWTH Aachen, 52074 Aachen, Germany (T.F.E.); (K.B.); (R.A.)
- Center for Integrated Oncology (CIO), University Hospital RWTH Aachen, 52074 Aachen, Germany;
| |
Collapse
|
48
|
Pires CS, da Rocha MJ, Presa MH, Zuge NP, Besckow EM, Ledebuhr KNB, Kuntz NEB, Godoi B, Bortolatto CF, Brüning CA. Dopaminergic receptors involvement in the antidepressant-like effect of N-(3-((3-(trifluoromethyl)phenyl)selanyl)prop-2-yn-1-yl) benzamide in mice. Neurosci Lett 2025; 849:138144. [PMID: 39889880 DOI: 10.1016/j.neulet.2025.138144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/13/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
Major Depressive Disorder (MDD) directly impacts the lives of countless individuals worldwide, yet its causes remain incompletely understood. However, it is recognized that a deficiency in monoamines, including dopamine, may contribute to this disorder. N-(3-((3-(trifluoromethyl)phenyl)selenyl)prop-2-yn-1-yl) (CF3SePB) is an organoselenium compound that presented antidepressant-like effect in mice related to modulation of serotonergic, but not noradrenergic system. To expand the knowledge about CF3SePB mechanisms of action, this study aimed to evaluate the involvement of dopaminergic system in its antidepressant-like effect. Male Swiss mice were pre-treated with the haloperidol (0.05 mg/kg, i.p., a non-selective D2 receptor antagonist), SCH 23390 (0.01 mg/kg, s.c., a D1 receptor antagonist), and sulpiride (50 mg/kg, i.p., a D2 receptor antagonist) 15 min before CF3SePB (50 mg/kg, i.g.), and after 30 min of CF3SePB administration the forced swimming test (FST) was performed. CF3SePB presented an anti-immobility effect in the FST, demonstrated by increase in the latency to first episode of immobility and reduction of total immobility of mice, and the pre-treatment of mice with haloperidol, SCH 23390 and sulpiride prevented these effects, showing that the antidepressant-like effect of CF3SePB is related to the modulation of the dopaminergic system, specifically the D1 and D2 receptors. In addition, in silico pharmacokinetic profiling of CF3SePB predicted its low likelihood of inducing adverse effects and potential to cross the blood-brain barrier. These results expand the understanding of CF3SePB mechanisms for its antidepressant-like effect, reinforcing the potential of this organonoselenium compound for developing new antidepressants.
Collapse
Affiliation(s)
- Camila Simões Pires
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão do Leão Campus, Pelotas, RS 96010-900, Brazil
| | - Marcia Juciele da Rocha
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão do Leão Campus, Pelotas, RS 96010-900, Brazil
| | - Marcelo Heinemann Presa
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão do Leão Campus, Pelotas, RS 96010-900, Brazil
| | - Narryman Pinto Zuge
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão do Leão Campus, Pelotas, RS 96010-900, Brazil
| | - Evelyn Mianes Besckow
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão do Leão Campus, Pelotas, RS 96010-900, Brazil
| | - Kauane Nayara Bahr Ledebuhr
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão do Leão Campus, Pelotas, RS 96010-900, Brazil
| | - Natália Emanuele Biolosor Kuntz
- Nucleus of Synthesis and Application of Organic and Inorganic Compounds (NUSAACOI), Federal University of Fronteira Sul (UFFS), Campus Cerro Largo, Cerro Largo, RS, Brazil
| | - Benhur Godoi
- Nucleus of Synthesis and Application of Organic and Inorganic Compounds (NUSAACOI), Federal University of Fronteira Sul (UFFS), Campus Cerro Largo, Cerro Largo, RS, Brazil
| | - Cristiani Folharini Bortolatto
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão do Leão Campus, Pelotas, RS 96010-900, Brazil.
| | - César Augusto Brüning
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão do Leão Campus, Pelotas, RS 96010-900, Brazil.
| |
Collapse
|
49
|
Noh H, Choi S, Park KW, Lee S, Seok DW, Kim YE, Kwon HJ, Kim H, Park HJ, Kim TY, Kang D, Jeong JS. Amino Acid Hepatotoxicity Biomarkers in Human Hepatic Organoids: Promising Standardization of Drug Toxicity Evaluation. ACS Pharmacol Transl Sci 2025; 8:510-521. [PMID: 39974651 PMCID: PMC11833729 DOI: 10.1021/acsptsci.4c00612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 02/21/2025]
Abstract
Human hepatic organoids (hHOs) are regarded as physiologically relevant in vitro platforms to evaluate hepatotoxicity, a critical step in drug development, but their applications are currently limited by the lack of qualified and standardized evaluation markers. In this study, by leveraging the established reference measurement system of amino acids (AAs), we propose 12 new biomarkers for drug-induced hepatotoxicity evaluation in human induced pluripotent stem cell-derived hHOs. Two orthogonal analytical methods for AAs were developed and validated based on isotope dilution mass spectrometry. Four AAs (aspartic acid, arginine, glutamine, and phenylalanine) and eight ratios of two designated AAs in the media of hHOs showed reliable alteration by drug treatment, which was confirmed by differentiating between hepatotoxic and nonhepatotoxic drugs. The superiorities of AA-based toxicity evaluation using the media of hHOs are as follows: (i) ability to use media only, without direct damage to or consumption of the organoids, (ii) ability to measure and compare quantities of AAs through a standardized reference measurement system rather than nonstandardized cell viability indicators, and (iii) no requirement for further data normalization in the case of the AA ratios. The AA analysis-based results demonstrate the reliability and potential of the proposed biomarkers as not only straightforward indicators of drug-induced hepatotoxicity but also absolutely comparable measures as a step toward standardization based on the AA reference measurement system.
Collapse
Affiliation(s)
- Haneul Noh
- Department
of Predictive Toxicology, Korea Institute
of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Seohyun Choi
- Korea
Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
- University
of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Kyung Won Park
- Korea
Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Shinji Lee
- Korea
Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Dong Wook Seok
- Korea
Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
- University
of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Young Eun Kim
- Korea
Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
- School
of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Ha-Jeong Kwon
- Korea
Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Hyemin Kim
- Department
of Predictive Toxicology, Korea Institute
of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Han-Jin Park
- Department
of Predictive Toxicology, Korea Institute
of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Tae-Young Kim
- School
of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Dukjin Kang
- Korea
Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ji-Seon Jeong
- Korea
Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
- University
of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| |
Collapse
|
50
|
Sun H, Wang Y, Deng G, Gao R, Zhang M, Huang L, He W, Zhang Z, Yu D, Chen P, Lu F, Liu S. Cortex Dictamni-induced hepatotoxicity by enhanced oxidative phosphorylation: Insights from integrative transcriptomics, proteomics, and metabolomics analyses. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156511. [PMID: 39954621 DOI: 10.1016/j.phymed.2025.156511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Cortex Dictamni (CD) is a traditional Chinese medicine that is commonly used to treat various skin diseases. Recently, clinical reports have highlighted its potential to induce severe hepatotoxicity. However, the underlying mechanisms of toxicity remain inadequately explored. PURPOSE The aim of this study was to elucidate the intrinsic mechanisms of CD-induced hepatotoxicity. STUDY DESIGN Hepatotoxicity was assessed in SD rats, and human primary hepatocytes (HPHs) and differentiated HepaRG (dHepaRG) cells were used for in vitro testing. METHODS The major components of CD were determined using ultra-performance liquid chromatography (UPLC). Rats were randomly divided into control, CD-high (CD-H), CD-middle (CD-M), CD-low (CD-L), and isoniazid (INH) groups and administered oral gavage for four weeks. Serum biochemical indices, histopathological changes, apoptotic markers, and liver function were evaluated to assess hepatotoxicity. A comprehensive analysis of rat liver samples was performed using transcriptomic, proteomic, and metabolomic approaches to identify key pathways involved in CD-induced hepatotoxicity. In vitro toxicity validation of CD was performed using HPHs and dHepaRG cells. The key pathway was validated in vivo and in vitro. RESULTS CD primarily contained obacunone, fraxinellone, and dictamine. Administration of CD-H (9 times the maximum daily clinical dose in adults) and CD-M (3 times the maximum daily clinical dose in adults) for 4 weeks induced varying degrees of hepatotoxicity in rats. The CD-H group presented increased absolute and relative liver weights, reduced alanine aminotransferase (ALT) and bile acid transporter levels, and increased albumin (ALB) and cytochrome P450 (CYP) 3A4 levels, indicating significant hepatotoxicity in rats. Integrated multiomics analysis revealed that NADH dehydrogenase (ubiquinone) Fe-S protein 2 (Ndufs2) is a critical regulator of CD-induced hepatotoxicity involving oxidative phosphorylation (OXPHOS). CD inhibited the viability of HPHs and dHepaRG cells, demonstrating its significant cytotoxicity. Mechanistic validation revealed that CD upregulated Ndufs2, reactive oxygen species (ROS) and mitochondrial respiratory chain complex (MRCC) I, leading to nuclear factor erythroid 2-related factor 2 (Nrf2) pathway activation, apoptosis, mitochondrial dysfunction, and hepatotoxicity. CONCLUSION In summary, our study presents a comprehensive picture of the toxicity of CD in terms of dose and sex and reveals, for the first time, the central role of Ndufs2-regulated OXPHOS in CD-induced hepatotoxicity.
Collapse
Affiliation(s)
- Huijuan Sun
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Yu Wang
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Geyu Deng
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Rui Gao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Mengmeng Zhang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Lin Huang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Wenjie He
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Zhendong Zhang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Donghua Yu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Pingping Chen
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, PR China
| | - Fang Lu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, PR China.
| | - Shumin Liu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, PR China.
| |
Collapse
|