1
|
Danpanichkul P, Suparan K, Chaiyakunapruk N, Auttapracha T, Kongarin S, Wattanachayakul P, Ramadoss V, Suenghataiphorn T, Sukphutanan B, Pang Y, Lui RN, Yang JD, Noureddin M, Díaz LA, Liangpunsakul S, Arab JP, Wijarnpreecha K. Alcohol-related liver and extrahepatic malignancies: burden of disease and socioeconomic disparities in 2019. Eur J Gastroenterol Hepatol 2025; 37:198-206. [PMID: 39589794 DOI: 10.1097/meg.0000000000002882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
BACKGROUND Alcohol is linked to various cancers. While many studies have focused on developed countries, the burden of alcohol-related cancers in developing countries remains underexplored. METHODS We analyzed data from the Global Burden of Disease Study (2000-2019) to assess mortality and disability-adjusted life years (DALYs) from alcohol-related cancers in low and low-to-middle sociodemographic index (SDI) countries. RESULTS In 2019, there were 494 730 mortality from alcohol-related cancer. Low and low-middle SDI countries contributed over 15% of global mortality of alcohol-related cancer. Among multiple types of cancer, other pharyngeal cancers in these countries accounted for over 30% of global mortality of alcohol-related cancer. Primary liver cancer exhibited the highest mortality ( n = 16 090) in low and low-middle SDI countries. While deaths and DALYs rates from alcohol-related cancers decreased globally between 2000 and 2019, the related burden increased in low and low-middle SDI countries with a rise in all types of alcohol-related cancers, except for primary liver cancer. The most rapidly growing mortality rates in low SDI were from other pharyngeal cancers (+2.25%), whereas in low-middle SDI countries, colorectal cancer evidenced the highest increase (+2.76%). CONCLUSION The burden from alcohol-related cancer has risen in countries with low and low-to-middle SDI, especially other pharyngeal cancers and colorectal cancer. Policymakers should focus on improving alcohol-related policies as well as screening availability to tackle the associated burden of cancer in resource-constrained countries. However, the difficulty in isolating the impact of alcohol due to limited data on other confounders necessitates caution in interpreting these findings.
Collapse
Affiliation(s)
- Pojsakorn Danpanichkul
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Kanokphong Suparan
- Immunology Unit, Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nathorn Chaiyakunapruk
- Department of Pharmacotherapy, College of Pharmacy, University of Utah
- IDEAS Center, Veterans Affairs Salt Lake City Healthcare System, Salt Lake City, Utah, USA
| | | | | | | | - Vijay Ramadoss
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore
| | | | | | - Yanfang Pang
- Affiliated Hospital of Youjiang Medical University for Nationalities
- National Immunological Laboratory of Traditional Chinese Medicine
- Center for Medical Laboratory Science, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Rashid N Lui
- Department of Clinical Oncology, and Division of Gastroenterology and Hepatology, Department of Medicine and Therapeutics, Institute of Digestive Diseases, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Comprehensive Transplant Center, and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Mazen Noureddin
- Houston Research Institute and Houston Methodist Hospital, Houston, Texas, USA
| | - Luis Antonio Díaz
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile
- Observatorio Multicéntrico de Enfermedades Gastrointestinales, OMEGA, Santiago, Chile
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine
- Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| | - Juan Pablo Arab
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile
- Observatorio Multicéntrico de Enfermedades Gastrointestinales, OMEGA, Santiago, Chile
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Karn Wijarnpreecha
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Arizona College of Medicine
- Department of Internal Medicine, Banner University Medical Center
- BIO5 Institute, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
| |
Collapse
|
2
|
Wang J, Zhou T. Unveiling gut microbiota's role: Bidirectional regulation of drug transport for improved safety. Med Res Rev 2025; 45:311-343. [PMID: 39180410 DOI: 10.1002/med.22077] [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: 05/09/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/26/2024]
Abstract
Drug safety is a paramount concern in the field of drug development, with researchers increasingly focusing on the bidirectional regulation of gut microbiota in this context. The gut microbiota plays a crucial role in maintaining drug safety. It can influence drug transport processes in the body through various mechanisms, thereby modulating their efficacy and toxicity. The main mechanisms include: (1) The gut microbiota directly interacts with drugs, altering their chemical structure to reduce toxicity and enhance efficacy, thereby impacting drug transport mechanisms, drugs can also change the structure and abundance of gut bacteria; (2) bidirectional regulation of intestinal barrier permeability by gut microbiota, promoting the absorption of nontoxic drugs and inhibiting the absorption of toxic components; (3) bidirectional regulation of the expression and activity of transport proteins by gut microbiota, selectively promoting the absorption of effective components or inhibiting the absorption of toxic components. This bidirectional regulatory role enables the gut microbiota to play a key role in maintaining drug balance in the body and reducing adverse reactions. Understanding these regulatory mechanisms sheds light on novel approaches to minimize toxic side effects, enhance drug efficacy, and ultimately improve drug safety. This review systematically examines the bidirectional regulation of gut microbiota in drug transportation from the aforementioned aspects, emphasizing their significance in ensuring drug safety. Furthermore, it offers a prospective outlook from the standpoint of enhancing therapeutic efficacy and reducing drug toxicity, underscoring the importance of further exploration in this research domain. It aims to provide more effective strategies for drug development and treatment.
Collapse
Affiliation(s)
- Jinyi Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
3
|
Fang C, Yu Y, Di S, Wang X, Jin Y. Untargeted metabolomic analysis reveals a time-course hepatic metabolism disorder induced by short-term 6PPD exposure in rats. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:177071. [PMID: 39437917 DOI: 10.1016/j.scitotenv.2024.177071] [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: 08/27/2024] [Revised: 10/09/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
The tire antioxidant 6PPD has garnered extensive attention due to its widespread presence in the environment and the harmful effects of its transformation products on aquatic organisms. 6PPD has been detected in airborne dust, and it can enter mammals through inhalation exposure. While the toxic effects of 6PPD exposure have been reported in mammals, its effects on hepatic metabolism still remain poorly understood. Here, we collected the serum and liver samples at 1, 6, and 72 h following a single oral exposure of 100 mg/kg body weight (bw) 6PPD, respectively. We also investigated changes in serum and hepatic physiological indicators and metabolites, correspondingly. Results indicated that single time oral exposure a high dose of 6PPD did not significantly affect the physiological indexes of rats within a short time frame. However, untargeted metabolomics analysis of the metabolites in the liver at 1, 6, and 72 h revealed that the number of differential expression metabolites gradually increased over time and the most affected substances were lipids and lipid-like molecules. Interestingly, the KEGG pathway enrichment analysis indicated that 6PPD disrupted the riboflavin metabolism, leading to a significant decrease in FMN levels at all time points. In addition, the hepatic glucose metabolism was significantly affected at 6 and 72 h after oral administration. Taken together, short-term exposure to 6PPD disturbed lipid and riboflavin metabolism and gradually affected glucose metabolism in the liver of rats. These findings revealed the impacts of 6PPD on the hepatic metabolism in animals, and also offered some important insights into its toxicology and health risk.
Collapse
Affiliation(s)
- Chanlin Fang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yundong Yu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products/Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Shanshan Di
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products/Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Xinquan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products/Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China.
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
4
|
Liu L, Sun S, Li X. Physcion inhibition of CYP2C9, 2D6 and 3A4 in human liver microsomes. PHARMACEUTICAL BIOLOGY 2024; 62:207-213. [PMID: 38353248 PMCID: PMC10868446 DOI: 10.1080/13880209.2024.2314089] [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: 02/24/2023] [Accepted: 01/25/2024] [Indexed: 02/16/2024]
Abstract
CONTEXT The effect of the active ingredients in traditional Chinese medicines on the activity of cytochrome P450 enzymes (CYP450s) is a critical factor that should be considered in TCM prescriptions. Physcion, the major active ingredient of Rheum spp. (Polygonaceae), possesses wide pharmacological activities. OBJECTIVES The effect of physcion on CYP450 activity was investigated to provide a theoretical basis for use. MATERIALS AND METHODS The experiments were conducted in pooled human liver microsomes (HLMs). The activity of CYP450 isoforms was evaluated with corresponding substrates and probe reactions. Blank HLMs were set as negative controls, and typical inhibitors were employed as positive controls. The inhibition model was fitted with Lineweaver Burk plots. The concentration (0, 2.5, 5, 10, 25, 50 and 100 μM physcion) and time-dependent (0, 5, 10, 15 and 30 min) effects of physcion were also assessed. RESULTS Physcion suppressed CYP2C9, 2D6 and 3A4 in a concentration-dependent manner with IC50 values of 7.44, 17.84 and 13.50 μM, respectively. The inhibition of CYP2C9 and 2D6 was competitive with the Ki values of 3.69 and 8.66 μM, respectively. The inhibition of CYP3A4 was non-competitive with a Ki value of 6.70 μM. Additionally, only the inhibition of CYP3A4 was time-dependent with the KI and Kinact parameters of 3.10 μM-1 and 0.049 min-1, respectively. CONCLUSIONS The inhibition of CYP450s by physcion should be considered in its clinical prescription, and the study design can be employed to evaluate the interaction of CYP450s with other herbs.
Collapse
Affiliation(s)
- Lu Liu
- Department of Endocrine, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Sen Sun
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Shanghai, PR China
| | - Xiaohua Li
- Department of Endocrine, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| |
Collapse
|
5
|
Valladales-Restrepo LF, Ospina-Cano JA, Aristizábal-Carmona BS, Machado-Alba JE. Prescription Patterns of Inducers and Inhibitors of Cytochrome P450 and Their Potential Drug Interactions in the Real World: A Cross-Sectional Study. Drugs Real World Outcomes 2024; 11:617-626. [PMID: 39243339 PMCID: PMC11589024 DOI: 10.1007/s40801-024-00450-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2024] [Indexed: 09/09/2024] Open
Abstract
INTRODUCTION Both the induction and inhibition of cytochrome P450 are associated with multiple pharmacological interactions, which can lead to loss of efficacy or increase the risk of adverse drug reactions. OBJECTIVE The aim was to determine the prescription patterns of cytochrome P450-inducing and -inhibiting drugs and their contraindicated and major pharmacological interactions in a group of patients from Colombia. METHODS This cross-sectional observational study included patients who received drugs that induce or inhibit metabolism and examined their contraindicated and major pharmacological interactions. The patients were identified from a population-based database of drug dispensing. Patients were included between December 1 and December 31, 2021. Inhibitors and inducers of cytochrome P450 were classified based on FDA (Food and Drug Administration) guidelines. Drug interactions were identified using the Micromedex® database. Descriptive, bivariate and multivariable analysis was performed. RESULTS A total of 63,433 patients were analyzed. Antiseizure medications (35.9%) and antifungals (27.6%) were the most used inducers and inhibitors. A total of 30.1% of patients had potential contraindicated or greater interactions. The following factors were associated with a higher probability of presenting a potential pharmacological interaction: being male (OR 1.14; 95% CI 1.10-1.19), aged 18-39 years (OR 1.77; 95% CI 1.67-1.89) or 40-64 years (OR 1.64; 95% CI 1.56-1.72), having neurological diseases (OR 1.28; 95% CI 1.21-1.35), having psychiatric diseases (OR 3.84; 95% CI 3.58-4.13), having rheumatologic diseases (OR 1.32; 95% CI 1.23-1.41), receiving comedications with statins (OR 1.14; 95% CI 1.08-1.19), receiving comedications with analgesics (OR 1.33; 95% CI 1.27-1.38), receiving comedications with antiparasitics (OR 2.88; 95% CI 2.66-3.11) and an increase in the number of medications (OR 1.24; 95% CI 1.23-1.25). CONCLUSION Among the users of cytochrome P450 inhibitors and inducers, potential contraindications and greater interactions are very common, especially in men under 65 years of age with comorbidities and polypharmacy.
Collapse
Affiliation(s)
- Luis Fernando Valladales-Restrepo
- Grupo de Investigación en Farmacoepidemiología y Farmacovigilancia, Universidad Tecnológica de Pereira-Audifarma S.A, Calle 105 No. 14-140, 660003, Pereira, Risaralda, Colombia
- Grupo de Investigación Biomedicina, Facultad de Medicina, Fundación Universitaria Autónoma de las Américas, Pereira, Colombia
- Semillero de Investigación en Farmacología Geriátrica, Grupo de Investigación Biomedicina, Facultad de Medicina, Fundación Universitaria Autónoma de las Américas, Pereira, Colombia
| | - Juan Alberto Ospina-Cano
- Grupo de Investigación en Farmacoepidemiología y Farmacovigilancia, Universidad Tecnológica de Pereira-Audifarma S.A, Calle 105 No. 14-140, 660003, Pereira, Risaralda, Colombia
| | - Brayan Stiven Aristizábal-Carmona
- Semillero de Investigación en Farmacología Geriátrica, Grupo de Investigación Biomedicina, Facultad de Medicina, Fundación Universitaria Autónoma de las Américas, Pereira, Colombia
| | - Jorge Enrique Machado-Alba
- Grupo de Investigación en Farmacoepidemiología y Farmacovigilancia, Universidad Tecnológica de Pereira-Audifarma S.A, Calle 105 No. 14-140, 660003, Pereira, Risaralda, Colombia.
| |
Collapse
|
6
|
Chen J, Yao Y, Mao X, Chen Y, Ni F. Liver-targeted delivery based on prodrug: passive and active approaches. J Drug Target 2024; 32:1155-1168. [PMID: 39072411 DOI: 10.1080/1061186x.2024.2386416] [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: 03/20/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND The liver, a central organ in human metabolism, is often the primary target for drugs. However, conditions such as viral hepatitis, cirrhosis, non-alcoholic fatty liver disease (NAFLD), and hepatocellular carcinoma (HCC) present substantial health challenges worldwide. Existing treatments, which suffer from the non-specific distribution of drugs, frequently fail to achieve desired efficacy and safety, risking unnecessary liver harm and systemic side effects. PURPOSE The aim of this review is to synthesise the latest progress in the design of liver-targeted prodrugs, with a focus on passive and active targeting strategies, providing new insights into the development of liver-targeted therapeutic approaches. METHODS This study conducted an extensive literature search through databases like Google Scholar, PubMed, Web of Science, and China National Knowledge Infrastructure (CNKI), systematically collecting and selecting recent research on liver-targeted prodrugs. The focus was on targeting mechanisms, including the Enhanced Permeability and Retention (EPR) effect, the unique microenvironment of liver cancer, and active targeting through specific transporters and receptors. RESULTS Active targeting strategies achieve precise drug delivery by binding specific ligands to liver surface receptors. Passive targeting takes advantage of the EPR effect and tumour characteristics to enrich drugs in liver tumours. The review details successful cases of using small molecule ligands, peptides, antibodies and nanoparticles as drug carriers. CONCLUSION Liver-targeted prodrug strategies show great potential in enhancing the efficacy of drug treatment and reducing side effects for liver diseases. Future research should balance the advantages and limitations of both targeting strategies, focusing on optimising drug design and targeting efficiency, especially for clinical application. In-depth research on liver-specific receptors and the development of innovative targeting molecules are crucial for advancing the field of liver-targeted prodrugs.
Collapse
Affiliation(s)
- Jiaqi Chen
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingrui Yao
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoran Mao
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuzhou Chen
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Feng Ni
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| |
Collapse
|
7
|
Mokhosoev IM, Astakhov DV, Terentiev AA, Moldogazieva NT. Human Cytochrome P450 Cancer-Related Metabolic Activities and Gene Polymorphisms: A Review. Cells 2024; 13:1958. [PMID: 39682707 DOI: 10.3390/cells13231958] [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: 10/26/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Cytochromes P450 (CYPs) are heme-containing oxidoreductase enzymes with mono-oxygenase activity. Human CYPs catalyze the oxidation of a great variety of chemicals, including xenobiotics, steroid hormones, vitamins, bile acids, procarcinogens, and drugs. FINDINGS In our review article, we discuss recent data evidencing that the same CYP isoform can be involved in both bioactivation and detoxification reactions and convert the same substrate to different products. Conversely, different CYP isoforms can convert the same substrate, xenobiotic or procarcinogen, into either a more or less toxic product. These phenomena depend on the type of catalyzed reaction, substrate, tissue type, and biological species. Since the CYPs involved in bioactivation (CYP3A4, CYP1A1, CYP2D6, and CYP2C8) are primarily expressed in the liver, their metabolites can induce hepatotoxicity and hepatocarcinogenesis. Additionally, we discuss the role of drugs as CYP substrates, inducers, and inhibitors as well as the implication of nuclear receptors, efflux transporters, and drug-drug interactions in anticancer drug resistance. We highlight the molecular mechanisms underlying the development of hormone-sensitive cancers, including breast, ovarian, endometrial, and prostate cancers. Key players in these mechanisms are the 2,3- and 3,4-catechols of estrogens, which are formed by CYP1A1, CYP1A2, and CYP1B1. The catechols can also produce quinones, leading to the formation of toxic protein and DNA adducts that contribute to cancer progression. However, 2-hydroxy- and 4-hydroxy-estrogens and their O-methylated derivatives along with conjugated metabolites play cancer-protective roles. CYP17A1 and CYP11A1, which are involved in the biosynthesis of testosterone precursors, contribute to prostate cancer, whereas conversion of testosterone to 5α-dihydrotestosterone as well as sustained activation and mutation of the androgen receptor are implicated in metastatic castration-resistant prostate cancer (CRPC). CYP enzymatic activities are influenced by CYP gene polymorphisms, although a significant portion of them have no effects. However, CYP polymorphisms can determine poor, intermediate, rapid, and ultrarapid metabolizer genotypes, which can affect cancer and drug susceptibility. Despite limited statistically significant data, associations between CYP polymorphisms and cancer risk, tumor size, and metastatic status among various populations have been demonstrated. CONCLUSIONS The metabolic diversity and dual character of biological effects of CYPs underlie their implications in, preliminarily, hormone-sensitive cancers. Variations in CYP activities and CYP gene polymorphisms are implicated in the interindividual variability in cancer and drug susceptibility. The development of CYP inhibitors provides options for personalized anticancer therapy.
Collapse
Affiliation(s)
| | - Dmitry V Astakhov
- Department of Biochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Alexander A Terentiev
- Department of Biochemistry and Molecular Biology, N.I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | | |
Collapse
|
8
|
Subramani RM, Shamprasad BR, Viswanathan NSS, Nagarajan S, Sivasubramanian A. Therapeutic deep eutectic solvent with saponin, optimized through response surface methodology, exert potent in vivo antimicrobial effects against Pseudomonas aeruginosa. Sci Rep 2024; 14:28181. [PMID: 39548135 PMCID: PMC11567959 DOI: 10.1038/s41598-024-76993-8] [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: 03/23/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024] Open
Abstract
Therapeutic deep eutectic solvents (THEDES) are a type of deep eutectics that can be used as active pharmaceutical ingredients (APIs) by enhancing the drug bioavailability, by increasing the solubility of APIs in aqueous solutions or by increasing their permeability through biological barriers. In this study, we demonstrate the utility of designer THEDES, based on Quillaja Saponin (SAP), a triterpene natural product to unravel the antimicrobial potential of such THEDES. THEDES were prepared by gently mixing and heating five different α- hydroxy acids- malic, citric, tartaric, glycolic, lactic acids and SAP with water as a third component in the THEDES. The antimicrobial assays were performed with the as-prepared SAP-THEDES and among the panel of bacterial strains, saponin-malic acid THEDES [SMA-THEDES] displayed potent activity in disassembling PA biofilms, demonstrating that the SMA-THEDES can serve as therapeutic antibacterial biomaterials. To further optimize, a Box-Behnken- response surface methodology (RSM) experimental design was done and at the optimum conditions of SAP (1.014 m.mol), Malic acid (1.003 m.mol), Water (0.117 m.mol), SMA-THEDES exhibited maximum MIC of P. aeruginosa with the minimum absorbance at 590 nm of 0.105. The effectiveness of SMA-THEDES as antibiofilm agents on P. aeruginosa with the mechanism studies have been explored. The colony count from the in vivo infection zebrafish model in the treatment group showed a decline of > 2 fold for SMA-THEDES. Toxicity studies did not reveal any abnormality in liver and brain enzyme levels. Liver morphology images show no severe cytological alterations when treated with SMA-THEDES and were almost similar to the normal liver.
Collapse
Affiliation(s)
- Ramya M Subramani
- Department of Chemistry, School of Chemical & Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed University, Thanjavur, 613 401, Tamil Nadu, India
| | - Bhanuvalli R Shamprasad
- Department of Chemistry, School of Chemical & Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed University, Thanjavur, 613 401, Tamil Nadu, India
| | - Narayana Shri Sanjeev Viswanathan
- Department of Chemistry, School of Chemical & Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed University, Thanjavur, 613 401, Tamil Nadu, India
| | - Saisubramanian Nagarajan
- Department of Biotechnology, School of Chemical & Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed University, Thanjavur, 613 401, Tamil Nadu, India
- Centre for Research on Infectious Diseases, School of Chemical & Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed University, Thanjavur, 613 401, Tamil Nadu, India
| | - Arvind Sivasubramanian
- Department of Chemistry, School of Chemical & Biotechnology, Shanmugha Arts, Science, Technology & Research Academy (SASTRA) Deemed University, Thanjavur, 613 401, Tamil Nadu, India.
| |
Collapse
|
9
|
Wang MF, Sun T, Chen SY, Wang X, Li H, Wang JQ. Effects of Kalimeris indica on alcohol-induced liver injury through storing Nrf2/HO-1 pathway and gut microbiota. Front Pharmacol 2024; 15:1502096. [PMID: 39600370 PMCID: PMC11588484 DOI: 10.3389/fphar.2024.1502096] [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: 09/26/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Background Kalimeris indica (L.) Sch. Bip., (K. indica) is a plant classified under the genus Kalimeris within the Asteraceae family. The herb of K. indica has been historically utilized as a traditional medicine. The consumption of excessive amounts of alcohol represents a lifestyle choice that can induce tissue damage and contribute to the development of various health conditions. Method The HPLC-MS method was used to reveal the chemical composition of K. indica extract. HepG2 cells were used to test the in vitro oxidative stress. C57BL/6 mice were used to construct the in vivo alcohol-induced liver injury. H/E staining and serum ALT and AST levels were tested to assess the in vivo protective effect of ML (50 and 200 mg/kg). GSH, SOD, and CAT levels along with byproduct MDA levels were used to evaluate the in vivo oxidative stress. Immunohistochemical experiments were used to examine the in vivo Nrf2 and HO-1 levels. 16S rRNA gene-based profiling method was used to test the alteration in gut microbiota. Results 16 compounds were identified from K. indica extract. K. indica treatment reduced oxidative stress in HepG2 cells treated with 5% alcohol. H/E staining results showed that K. indica (50 and 200 mg/kg) alleviated liver injury caused by alcohol administration, eliciting a similar protective effect to the positive drug silymarin. Serum ALT and AST examination gave a consistent result, showing that ML could restore serum ALT and AST levels in mice treated with alcohol. Furthermore, K. indica could also restore GSH, SOD, CAT, and MDA levels in alcohol-treated mice, showing a potent effect on oxidative stress alleviation. Immunohistochemical experiments indicated that K. indica showed the liver protective effect through Nrf2/HO-1 pathway. 16S rRNA gene-based profiling revealed that alcohol treatment caused the alteration in gut microbiota, while K. indica treatment could result in a significantly richer variety of microbial communities compared to the alcohol group. Conclusion K. indica (ML) has a protective effect on liver injury caused by alcohol administration. The Nrf2/HO-1 pathway and gut microbiota regulation were involved in the ML-induced liver protection. All the results indicate that K. indica has a potential in the treatment of alcohol-induced liver injury.
Collapse
Affiliation(s)
- Mo-Fei Wang
- The Department of General Surgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, China
| | - Tong Sun
- The Department of General Surgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, China
| | - Shi-Yu Chen
- Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diesases, Liaoning University, Shenyang, China
| | - Xue Wang
- The Department of General Surgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, China
| | - Hao Li
- The Department of General Surgery, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, China
| | - Jia-Qi Wang
- Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diesases, Liaoning University, Shenyang, China
| |
Collapse
|
10
|
Dharmarathne G, Kazi S, King S, Jayasinghe TN. The Bidirectional Relationship Between Cardiovascular Medications and Oral and Gut Microbiome Health: A Comprehensive Review. Microorganisms 2024; 12:2246. [PMID: 39597635 PMCID: PMC11596509 DOI: 10.3390/microorganisms12112246] [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: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Cardiovascular diseases (CVDs) are a leading cause of widespread morbidity and mortality. It has been found that the gut and oral microbiomes differ in individuals with CVDs compared to healthy individuals. Patients with CVDs often require long-term pharmacological interventions. While these medications have been extensively studied for their cardiovascular benefits, emerging research indicates that they may also impact the diversity and composition of the oral and gut microbiomes. However, our understanding of how these factors influence the compositions of the oral and gut microbiomes in individuals remains limited. Studies have shown that statins and beta-blockers, in particular, cause gut and oral microbial dysbiosis, impacting the metabolism and absorption of these medications. These alterations can lead to variations in drug responses, highlighting the need for personalized treatment approaches. The microbiome's role in drug metabolism and the impact of CVD medications on the microbiome are crucial in understanding these variations. However, there are very few studies in this area, and not all medications have been studied, emphasizing the necessity for further research to conclusively establish cause-and-effect relationships and determine the clinical significance of these interactions. This review will provide evidence of how the oral and gut microbiomes in patients with cardiovascular diseases (CVDs) interact with specific drugs used in CVD treatment.
Collapse
Affiliation(s)
- Gangani Dharmarathne
- Australian Laboratory Services Global, Water and Hydrographic, Hume, ACT 2620, Australia
| | - Samia Kazi
- Westmead Applied Research Centre, The University of Sydney, Sydney, NSW 2145, Australia
- Department of Cardiology, Westmead Hospital, Sydney, NSW 2145, Australia
| | - Shalinie King
- Westmead Applied Research Centre, The University of Sydney, Sydney, NSW 2145, Australia
- The Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Thilini N. Jayasinghe
- The Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- The Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
11
|
Zhou M, Qian Y, Du M, Wang J, Li J, Wang W. Metabolite identification of emerging disinfection byproduct dibromo-benzoquinone in vivo and in vitro: Multi-strategy mass-spectrometry annotation and toxicity characterization. ENVIRONMENT INTERNATIONAL 2024; 193:109134. [PMID: 39522490 DOI: 10.1016/j.envint.2024.109134] [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: 08/30/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Halobenzoquinones (HBQs) are emerging disinfection byproducts (DBPs) of high toxicity and also are shared active toxic intermediates of multiple halogenated organic pollutants. Due to the strong oxidizing property and electrophilicity, HBQs exhibit extremely diverse metabolism pathways in organisms. The identification of toxic-decisive metabolites is pivotal, albeit challenging, for understanding the toxicity mechanisms of HBQs. We employed dibromo-benzoquinone (DBBQ) as a representative HBQ, and established a systematic analytical strategy using high-resolution mass spectrometry, which collectively coupled suspect screening (SS), mass defect filtering (MDF), product ion filtering (PIF), isotopic signature filtering (ISF), and molecular networking (MN). As a result, 20 biotransformation products of DBBQ were identified in vivo and in vitro, involving metabolism reactions such as hydroxylation, methylation, methoxylation, acetylation, sulfonation, glucuronidation, glutathionylation, dimerization, and conjugation with amino acids or fatty acids. Quantitative structure-activity relationship (QSAR) analysis and cytotoxicity experiments consistently demonstrated the significantly high toxicity of the fatty acid conjugate compared to the parent compound DBBQ and other metabolites, pinpointing the important role of the fatty acid conjugation in determining the metabolism and toxicity of HBQs. The research conducted a comprehensive evaluation of the metabolism of a typical HBQ with the combination of multiple analytical and toxicity characterization methods, therefore screen out the most important metabolism pathway of HBQs.
Collapse
Affiliation(s)
- Meijiao Zhou
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Hangzhou, Zhejiang 310058, China
| | - Yichao Qian
- Hangzhou Huihong Environmental Technology Co., Ltd., Hangzhou, Zhejiang 310058, China
| | - Mine Du
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Hangzhou, Zhejiang 310058, China
| | - Jun Wang
- Department of Health Toxicology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Jinhua Li
- Department of Health Toxicology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Wei Wang
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Organic Pollution Process and Control, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
12
|
Li Y, Guo N, Zhao Y, Chen J, Zhao J, Bian J, Guo J, Yang C, Zhang X, Huang L. IL-17A activates JAK/STAT signaling to affect drug metabolizing enzymes and transporters in HepaRG cells. Mol Immunol 2024; 175:55-62. [PMID: 39305848 DOI: 10.1016/j.molimm.2024.09.008] [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: 11/15/2023] [Revised: 08/13/2024] [Accepted: 09/15/2024] [Indexed: 11/11/2024]
Abstract
The founding family member, Interleukin (IL)-17A, is commonly known as IL-17 and has garnered increasingly attention for proinflammatory functions in autoimmune disorders. Although the effects of IL-17A on hepatic important drug-metabolizing enzymes and transporters (DMETs) expression still remain unclear, it is critical to ascertain owing to the well-established alterations of the drug disposition capacity of the liver occurring during immune imbalance. The present study was designed to explore the effects and mechanisms of IL-17A on DMETs mRNA and protein expression in HepaRG cells by real-time quantitative reverse transcription polymerase chain reaction and Western blot, respectively. It is discovered that IL-17A can inhibit most DMETs mRNA expression (drug-metabolizing enzymes of CYP1A2, CYP3A4, CYP2C9, CYP2C19, GSTA1 and UGT1A1 and transporters of NTCP, OCT1, OATP1B1, BCRP and MDR1) as well as the protein expression of CYP3A4 and CYP2C19, via the janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) signaling pathway. Thus, abnormal regulation of DMETs in IL-17A-mediated immune disorders such as psoriasis may cause alterations in pharmacokinetic processes and may occasionally result in unexpected drug-drug interactions (DDIs) in clinical practice.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Nan Guo
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yinyu Zhao
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jiali Chen
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinxia Zhao
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jialu Bian
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China; Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jing Guo
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China
| | - Changqing Yang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaohong Zhang
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China
| | - Lin Huang
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China.
| |
Collapse
|
13
|
Sethi N, Khokhar M, Mathur M, Batra Y, Mohandas A, Tomo S, Rao M, Banerjee M. Therapeutic Potential of Nutraceuticals against Drug-Induced Liver Injury. Semin Liver Dis 2024; 44:430-456. [PMID: 39393795 DOI: 10.1055/s-0044-1791559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Drug-induced liver injury (DILI) continues to be a major concern in clinical practice, thus necessitating a need for novel therapeutic approaches to alleviate its impact on hepatic function. This review investigates the therapeutic potential of nutraceuticals against DILI, focusing on examining the underlying molecular mechanisms and cellular pathways. In preclinical and clinical studies, nutraceuticals, such as silymarin, curcumin, and N-acetylcysteine, have demonstrated remarkable efficacy in attenuating liver injury induced by diverse pharmaceutical agents. The molecular mechanisms underlying these hepatoprotective effects involve modulation of oxidative stress, inflammation, and apoptotic pathways. Furthermore, this review examines cellular routes affected by these nutritional components focusing on their influence on hepatocytes, Kupffer cells, and stellate cells. Key evidence highlights that autophagy modulation as well as unfolded protein response are essential cellular processes through which nutraceuticals exert their cytoprotective functions. In conclusion, nutraceuticals are emerging as promising therapeutic agents for mitigating DILI, by targeting different molecular pathways along with cell processes involved in it concurrently.
Collapse
Affiliation(s)
- Namya Sethi
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Mitali Mathur
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Yashi Batra
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Amal Mohandas
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Sojit Tomo
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Karnataka, India
| | - Mithu Banerjee
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| |
Collapse
|
14
|
Mao K, Liu C, Tang Z, Rao Z, Wen J. Advances in drug resistance of osteosarcoma caused by pregnane X receptor. Drug Metab Rev 2024; 56:385-398. [PMID: 38872275 DOI: 10.1080/03602532.2024.2366948] [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: 01/17/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024]
Abstract
Osteosarcoma (OS) is a prevalent malignancy among adolescents, commonly manifesting during childhood and adolescence. It exhibits a high degree of malignancy, propensity for metastasis, rapid progression, and poses challenges in clinical management. Chemotherapy represents an efficacious therapeutic modality for OS treatment. However, chemotherapy resistance of OS is a major problem in clinical treatment. In order to treat OS effectively, it is particularly important to explore the mechanism of chemotherapy resistance in OS.The Pregnane X receptor (PXR) is a nuclear receptor primarily involved in the metabolism, transport, and elimination of xenobiotics, including chemotherapeutic agents. PXR involves three stages of drug metabolism: stage I: drug metabolism enzymes; stage II: drug binding enzyme; stage III: drug transporter.PXR has been confirmed to be involved in the process of chemotherapy resistance in malignant tumors. The expression of PXR is increased in OS, which may be related to drug resistance of OS. Therefore, wereviewed in detail the role of PXR in chemotherapy drug resistance in OS.
Collapse
Affiliation(s)
- Kunhong Mao
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Physiology, Hunan Normal University School of Medicine, Changsha, China
| | - Can Liu
- Department of Anatomy, Hunan Normal University school of Medicine, Changsha, China
| | - Zhongwen Tang
- Department of Pediatric Orthopedics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Zhouzhou Rao
- Key Laboratory of Translational Cancer Stem Cell Research, Department of Physiology, Hunan Normal University School of Medicine, Changsha, China
| | - Jie Wen
- Department of Anatomy, Hunan Normal University school of Medicine, Changsha, China
- Department of Pediatric Orthopedics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| |
Collapse
|
15
|
Popović L, Brankatschk B, Palladino G, Rossner MJ, Wehr MC. Polypharmacological profiling across protein target families and cellular pathways using the multiplexed cell-based assay platform safetyProfiler reveals efficacy, potency and side effects of drugs. Biomed Pharmacother 2024; 180:117523. [PMID: 39405910 DOI: 10.1016/j.biopha.2024.117523] [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: 08/16/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 11/14/2024] Open
Abstract
Selectivity profiling is key for assessing the pharmacological properties of multi-target drugs. We have developed a cell-based and barcoded assay encompassing ten druggable targets, including G protein-coupled receptors (GPCRs), receptor tyrosine kinases (RTKs), nuclear receptors, a protease as well as their key downstream pathways and profiled 17 drugs in living cells for efficacy, potency, and side effects. Notably, this multiplex assay, termed safetyProfiler assay, enabled the simultaneous assessment of multiple target and pathway activities, shedding light on the polypharmacological profile of compounds. For example, the neuroleptics clozapine, paliperidone, and risperidone potently inhibited primary targets DRD2 and HTR2A as well as cAMP and calcium pathways. However, while paliperidone and risperidone also potently inhibited the secondary target ADRA1A and mitogen-activated protein kinase (MAPK) downstream pathways, clozapine only exhibited mild antagonistic effects on ADRA1A and lacked MAPK inhibition downstream of DRD2 and HTR2A. Furthermore, we present data on the selectivity for bazedoxifene, an estrogen receptor antagonist currently undergoing clinical phase 2 trials for breast cancer, on MAPK signaling. Additionally, precise potency data for LY2452473, an androgen receptor antagonist, that completed a phase 2 clinical trial for prostate cancer, are presented. The non-selective kinase inhibitor staurosporine was observed to potently inactivate the two RTKs EGFR and ERBB4 as well as MAPK signaling, while eliciting stress-related cAMP responses. Our findings underscore the value of comprehensive profiling in elucidating the pharmacological properties of established and novel therapeutics, thereby facilitating the development of novel multi-target drugs with enhanced efficacy and selectivity.
Collapse
Affiliation(s)
- Lukša Popović
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstr. 7, Munich 80336, Germany; Systasy Bioscience GmbH, Fraunhoferstr. 8, Planegg-Martinsried 82152, Germany
| | - Ben Brankatschk
- Systasy Bioscience GmbH, Fraunhoferstr. 8, Planegg-Martinsried 82152, Germany
| | - Giulia Palladino
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstr. 7, Munich 80336, Germany; Systasy Bioscience GmbH, Fraunhoferstr. 8, Planegg-Martinsried 82152, Germany
| | - Moritz J Rossner
- Section of Molecular Neurobiology, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstr. 7, Munich 80336, Germany
| | - Michael C Wehr
- Research Group Cell Signalling, Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Nussbaumstr. 7, Munich 80336, Germany; Systasy Bioscience GmbH, Fraunhoferstr. 8, Planegg-Martinsried 82152, Germany.
| |
Collapse
|
16
|
Ali SA, Datusalia AK. Berberine attenuates ECM accumulation and the progression of acute liver failure through inhibition of NLRP3 inflammasome signalling. Toxicol Appl Pharmacol 2024; 492:117129. [PMID: 39428072 DOI: 10.1016/j.taap.2024.117129] [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: 06/19/2024] [Revised: 09/10/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Acute liver failure (ALF) is a life-threatening disease, characterized by upregulated extracellular matrix deposition and inflammatory signalling, with no effective treatment options and targets. The present study was designed to investigate the preventive and therapeutic effects of berberine (BBR) and its underlying mechanism in thioacetamide (TAA)-induced ALF. Male SD rats were administered with TAA 300 mg/kg, i.p., thrice to induce ALF and pre- or post-treated with BBR. To decipher the effects of BBR LFT markers, histopathological analysis of key fibrotic and inflammatory proteins was performed. In addition, the levels of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α were assessed by ELISA. Our work showed TAA-induced ALF animals were associated with increased ALT, AST, bilirubin (LFT markers) and histopathological alterations with profuse infiltration of inflammatory cells in the liver tissue. Treatment with BBR has significantly inhibited LFT markers and histological alterations triggered by TAA. In addition, TAA animals demonstrated increased collagen accumulation and upregulated expression of TGF-β1, vimentin, and α-SMA compared to control. The excessive accumulation of collagen, TGF-β1, vimentin, and α-SMA were significantly modulated with BBR treatment. Further, the fluorescence intensity of ROS an activator of NLRP3 including the NLRP3 inflammasome, and its downstream signalling ASC, cleaved IL-1β, and other pro-inflammatory cytokines like TNF-α and IL-6 stimulated by TAA were attenuated by BBR treatment. The current work indicated that BBR significantly ameliorated TAA-induced ALF by inhibiting the extracellular matrix accumulation associated with the NLRP3/IL-1β signalling pathway and could be a viable therapeutic option to treat ALF and other fibroinflammatory diseases.
Collapse
Affiliation(s)
- Syed Afroz Ali
- Laboratory of Molecular NeuroTherapeutics, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, Uttar Pradesh 226002, India
| | - Ashok Kumar Datusalia
- Laboratory of Molecular NeuroTherapeutics, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, Uttar Pradesh 226002, India; Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, Uttar Pradesh 226002, India.
| |
Collapse
|
17
|
Xie F, Gales T, Ringenberg MA, Wolf AI, Groseclose MR. Characterizing the Distribution of a Stimulator of Interferon Genes Agonist and Its Metabolites in Mouse Liver by Matrix-Assisted Laser Desorption/Ionization Imaging Mass Spectrometry. Drug Metab Dispos 2024; 52:1181-1186. [PMID: 37884391 DOI: 10.1124/dmd.122.001076] [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: 08/14/2023] [Revised: 10/06/2023] [Accepted: 10/19/2023] [Indexed: 10/28/2023] Open
Abstract
A STING (stimulator of interferon genes) agonist GSK3996915 under investigation in early discovery for hepatitis B was orally dosed to a mouse model for understanding the parent drug distribution in liver, the target organ. Matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) was used to quantify the distribution of GSK3996915 in liver collected from mice administered a single oral dose at 90 mg/kg. GSK3996915 was detected with a zonal distribution localized in the portal triad and highly concentrated in the main bile ducts, indicating clearance through biliary excretion. High spatial resolution imaging showed the distribution of the parent drug localized to the cellular populations in the sinusoids, including the Kupffer cells. Additionally, a series of drug-related metabolites were observed to be localized in the central zones of the liver. These results exemplify the potential of utilizing MALDI IMS for measuring not only quantitative drug distribution and target exposure but also drug metabolism and elimination in a single suite of experiments. SIGNIFICANCE STATEMENT: An integrated imaging approach utilizing matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI IMS) complemented with immunohistochemistry (IHC) and histology was used to address the question of target exposure at the cellular level. Localized quantification of the parent drug in the target organ and identification of potential metabolites in the context of tissue histology were also achieved in one experimental suite to support characterization of pharmacokinetic properties of the drug in the early discovery stage.
Collapse
Affiliation(s)
- Fang Xie
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - Tracy Gales
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - M A Ringenberg
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - Amaya I Wolf
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| | - M Reid Groseclose
- Bioimaging (F.X., T.G., M.R.G.), Nonclinical Safety (M.A.R.), and Immunology Research (A.I.W.), GlaxoSmithKline, Collegeville, Pennsylvania
| |
Collapse
|
18
|
Zhang Y, Chen Z, Xiao Y, Wu T, Yang H, Liu Y, Zhou R, Xiong Y, Xiong Y, Yang X, Zhou J, Zhou H, Zhang W, Shu Y, Li X, Guo F, Yin J, Liao S, Li Q, Zhu P. Effects of Compound Probiotics on Pharmacokinetics of Cytochrome 450 Probe Drugs in Rats. Drug Metab Dispos 2024; 52:1297-1312. [PMID: 39214665 DOI: 10.1124/dmd.124.001837] [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: 06/10/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Compound probiotics have been widely used and commonly coadministered with other drugs for treating various chronic illnesses, yet their effects on drug pharmacokinetics remain underexplored. This study elucidated the impact of VSL#3 on the metabolism of probe drugs for cytochrome P450 enzymes (P450s), specifically omeprazole, tolbutamide, midazolam, metoprolol, phenacetin, and chlorzoxazone. Male Wistar rats were administered drinking water containing VSL#3 or not for 14 days and then intragastrically administered a P450 probe cocktail; this was done to investigate the host P450's metabolic phenotype. Stool, liver/jejunum, and serum samples were collected for 16S ribosomal RNA sequencing, RNA sequencing, and bile acid profiling. The results indicated significant differences in both α and β diversity of intestinal microbial composition between the probiotic and vehicle groups in rats. In the probiotic group, the bioavailability of omeprazole increased by 269.9%, whereas those of tolbutamide and chlorpropamide decreased by 28.1% and 27.4%, respectively. The liver and jejunum exhibited 1417 and 4004 differentially expressed genes, respectively, between the two groups. In the probiotic group, most of P450 genes were upregulated in the liver but downregulated in the jejunum. The expression of genes encoding metabolic enzymes and drug transporters also changed. The serum-conjugated bile acids in the probiotic group were significantly reduced. Shorter duodenal villi and longer ileal villi were found in the probiotic group. In summary, VSL#3 administration altered the gut microbiota, host drug-processing gene expression, and intestinal structure in rats, which could be reasons for pharmacokinetic changes. SIGNIFICANCE STATEMENT: This study focused on the effects of the probiotic VSL#3 on the pharmacokinetic profile of cytochrome P450 probe drugs and the expression of host drug metabolism genes. Compared with previous studies, the present study provides a comprehensive explanation for the host drug metabolism profile modified by probiotics, combined here with the bile acid profile and histopathological analysis.
Collapse
Affiliation(s)
- Yanjuan Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Zhi Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yayi Xiao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Tianyuan Wu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Haijun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yujie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Rong Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yalan Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yanling Xiong
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Xuechun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Jian Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Yan Shu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Xiong Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Fugang Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Jianhui Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Shang Liao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| | - Peng Zhu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); National Clinical Research Center for Geriatric Disorders, Changsha, China (Y.Z., Y.X., T.W., H.Y., Y.L., R.Z., Yal.X., Yan.X., X.Y., J.Z., H.Z., W.Z., Q.L., P.Z.); Department of Hypertension, Xingsha Hospital, Changsha, China (Z.C., F.G., J.Y., S.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Maryland (Y.S.); and Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China (X.L.)
| |
Collapse
|
19
|
Chen K, Gao Z. Acacetin, a Natural Flavone with Potential in Improving Liver Disease Based on Its Anti-Inflammation, Anti-Cancer, Anti-Infection and Other Effects. Molecules 2024; 29:4872. [PMID: 39459239 PMCID: PMC11509893 DOI: 10.3390/molecules29204872] [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: 09/19/2024] [Revised: 10/07/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Liver disease is a global public problem, and the cost of its therapy is a large financial burden to governments. It is well known that drug therapy plays a critical role in the treatment of liver disease. However, present drugs are far from meeting clinical needs. Lots of efforts have been made to find novel agents to treat liver disease in the past several decades. Acacetin is a dihydroxy and monomethoxy flavone, named 5,7-dihydroxy-4'-methoxyflavone, which can be found in diverse plants. It has been reported that acacetin exhibits multiple pharmacological activities, including anti-cancer, anti-inflammation, anti-virus, anti-obesity, and anti-oxidation. These studies indicate the therapeutic potential of acacetin in liver disease. This review discussed the comprehensive information on the pathogenesis of liver disease (cirrhosis, viral hepatitis, drug-induced liver injury, and hepatocellular carcinoma), then introduced the biological source, structural features, and pharmacological properties of acacetin, and the possible application in preventing liver disease along with the pharmacokinetic and toxicity of acacetin, and future research directions. We systemically summarized the latest research progress on the potential therapeutic effect of acacetin on liver disease and existing problems. Based on the present published information, the natural flavone acacetin is an anticipated candidate agent for the treatment of liver disease.
Collapse
Affiliation(s)
- Kuihao Chen
- Department of Pharmacology, School of Medicine, Ningbo University, 818 Fenghua Rd., Ningbo 315211, China
| | - Zhe Gao
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd., Ningbo 315211, China
| |
Collapse
|
20
|
Gui X, Huang J, Ruan L, Wu Y, Guo X, Cao R, Zhou S, Tan F, Zhu H, Li M, Zhang G, Zhou H, Zhan L, Liu X, Tu S, Shao Z. zMAP toolset: model-based analysis of large-scale proteomic data via a variance stabilizing z-transformation. Genome Biol 2024; 25:267. [PMID: 39402594 PMCID: PMC11472442 DOI: 10.1186/s13059-024-03382-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
Isobaric labeling-based mass spectrometry (ILMS) has been widely used to quantify, on a proteome-wide scale, the relative protein abundance in different biological conditions. However, large-scale ILMS data sets typically involve multiple runs of mass spectrometry, bringing great computational difficulty to the integration of ILMS samples. We present zMAP, a toolset that makes ILMS intensities comparable across mass spectrometry runs by modeling the associated mean-variance dependence and accordingly applying a variance stabilizing z-transformation. The practical utility of zMAP is demonstrated in several case studies involving the dynamics of cell differentiation and the heterogeneity across cancer patients.
Collapse
Affiliation(s)
- Xiuqi Gui
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jing Huang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Linjie Ruan
- Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yanjun Wu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xuan Guo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ruifang Cao
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shuhan Zhou
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Fengxiang Tan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hongwen Zhu
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Mushan Li
- Department of Statistics, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Guoqing Zhang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hu Zhou
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lixing Zhan
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xin Liu
- Key Laboratory of Epigenetic Regulation and Intervention, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Shiqi Tu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Zhen Shao
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
21
|
Zhang Y, Gao J, Xu Y, Liu J, Huang S, Li G, Yao B, Sun Z, Wang X. Investigation of cytochrome P450 inhibitory properties of deoxyshikonin, a bioactive compound from Lithospermum erythrorhizon Sieb. et Zucc. Phytother Res 2024; 38:4855-4864. [PMID: 36317387 DOI: 10.1002/ptr.7664] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/26/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
Deoxyshikonin, a natural naphthoquinone compound extracted from Lithospermum erythrorhizon Sieb. et Zucc (Boraginaceae), has a wide range of pharmacological activities, including anti-tumor, anti-bacterial and wound healing effects. However, the inhibitory effect of deoxyshikonin on cytochrome P450 (CYP) remains unclear. This study investigated the potential inhibitory effects of deoxyshikonin on CYP1A2, 2B1/6, 2C9/11, 2D1/6, 2E1 and 3A2/4 enzymes in human and rat liver microsomes (HLMs and RLMs) by the cocktail approach in vitro. The single-point inactivation experiment showed that deoxyshikonin presented no time-dependent inhibition on CYP activities in HLMs and RLMs. Enzyme inhibition kinetics indicated that in HLMs, deoxyshikonin was not only a competitive inhibitor of CYP1A2 and 2E1, but also a mixed inhibitor of CYP2B6, 2C9, 2D6 and 3A4, with Ki of 2.21, 1.78, 1.68, 0.20, 4.08 and 0.44 μM, respectively. In RLMs, deoxyshikonin not only competitively inhibited CYP2B1 and 2E1, but also exhibited mixed inhibition on CYP1A2, 2C11, 2D1 and 3A2, with Ki values of no more than 18.66 μM. In conclusion, due to the low Ki values of deoxythiokonin on CYP enzymes in HLMs, this may lead to drug-drug interactions (DDI) and potential toxicity.
Collapse
Affiliation(s)
- Yuanjin Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| | - Jing Gao
- The College of Life Sciences, Northwest University, Xi'an, People's Republic of China
| | - Yuan Xu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| | - Jie Liu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| | - Shengbo Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| | - Guihong Li
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, People's Republic of China
| | - Bingyi Yao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| | - Zhenliang Sun
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, People's Republic of China
| | - Xin Wang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, People's Republic of China
| |
Collapse
|
22
|
Yao Q, Cheng S, Pan Q, Yu J, Cao G, Li L, Cao H. Organoids: development and applications in disease models, drug discovery, precision medicine, and regenerative medicine. MedComm (Beijing) 2024; 5:e735. [PMID: 39309690 PMCID: PMC11416091 DOI: 10.1002/mco2.735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Organoids are miniature, highly accurate representations of organs that capture the structure and unique functions of specific organs. Although the field of organoids has experienced exponential growth, driven by advances in artificial intelligence, gene editing, and bioinstrumentation, a comprehensive and accurate overview of organoid applications remains necessary. This review offers a detailed exploration of the historical origins and characteristics of various organoid types, their applications-including disease modeling, drug toxicity and efficacy assessments, precision medicine, and regenerative medicine-as well as the current challenges and future directions of organoid research. Organoids have proven instrumental in elucidating genetic cell fate in hereditary diseases, infectious diseases, metabolic disorders, and malignancies, as well as in the study of processes such as embryonic development, molecular mechanisms, and host-microbe interactions. Furthermore, the integration of organoid technology with artificial intelligence and microfluidics has significantly advanced large-scale, rapid, and cost-effective drug toxicity and efficacy assessments, thereby propelling progress in precision medicine. Finally, with the advent of high-performance materials, three-dimensional printing technology, and gene editing, organoids are also gaining prominence in the field of regenerative medicine. Our insights and predictions aim to provide valuable guidance to current researchers and to support the continued advancement of this rapidly developing field.
Collapse
Affiliation(s)
- Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Sheng Cheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Guoqiang Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic‐Chemical and Aging‐Related InjuriesHangzhouChina
| |
Collapse
|
23
|
Gunasaykaran SY, Chear NJY, Ismail S, Mohammad NA, Murugaiyah V, Ramanathan S. Drug-drug interactions of plant alkaloids derived from herbal medicines on the phase II UGT enzymes: an introductory review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03418-8. [PMID: 39325152 DOI: 10.1007/s00210-024-03418-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/28/2024] [Indexed: 09/27/2024]
Abstract
Herbal medicines are widely used as alternative or complementary therapies to treat and prevent chronic diseases. However, these can lead to drug-drug interactions (DDIs) that affect the glucuronidation reaction of UDP glucuronosyltransferases (UGTs), which convert drugs into metabolites. Plant extracts derived from medicinal herbs contain a diverse array of compounds categorized into different functional groups. While numerous studies have examined the inhibition of UGT enzymes by various herbal compounds, it remains unclear which group of compounds exerts the most significant impact on DDIs in the glucuronidation reaction. Recently, alkaloids derived from medicinal herbs, including kratom (Mitragyna speciosa), have gained attention due to their diverse pharmacological properties. This review primarily focuses on the DDIs of plant alkaloids from medicinal herbs, including kratom on the phase II UGT enzymes. Kratom is a new emerging herbal product in Western countries that is often used to self-treat chronic pain, opioid withdrawal, or as a replacement for prescription and non-prescription opioids. Kratom is well-known for its psychoactive alkaloids, which have a variety of psychopharmacological effects. However, the metabolism mechanism of kratom alkaloids, particularly on the phase II pathway, is still poorly understood. Simultaneously using kratom or other herbal products containing alkaloids with prescribed medicines may have an impact on the drug metabolism involving the phase II UGT enzymes. To ensure the safety and efficacy of treatments, gaining a better understanding of the DDIs when using herbal products with conventional medicine is crucial.
Collapse
Affiliation(s)
| | | | - Sabariah Ismail
- Centre for Drug Research, Universiti Sains Malaysia, 11800 USM, Pulau Pinang, Malaysia
| | | | - Vikneswaran Murugaiyah
- Centre for Drug Research, Universiti Sains Malaysia, 11800 USM, Pulau Pinang, Malaysia
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 USM, Pulau Pinang, Malaysia
| | - Surash Ramanathan
- Centre for Drug Research, Universiti Sains Malaysia, 11800 USM, Pulau Pinang, Malaysia.
| |
Collapse
|
24
|
Zhang X, Geng Q, Lin L, Zhang L, Shi C, Liu B, Yan L, Cao Z, Li L, Lu P, Tan Y, He X, Zhao N, Li L, Lu C. Insights gained into the injury mechanism of drug and herb induced liver injury in the hepatic microenvironment. Toxicology 2024; 507:153900. [PMID: 39079402 DOI: 10.1016/j.tox.2024.153900] [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: 06/08/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
Drug-Induced Liver Injury (DILI) and herb Induced Liver Injury (HILI) continues to pose a substantial challenge in both clinical practice and drug development, representing a grave threat to patient well-being. This comprehensive review introduces a novel perspective on DILI and HILI by thoroughly exploring the intricate microenvironment of the liver. The dynamic interplay among hepatocytes, sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, cholangiocytes, and the intricate vascular network assumes a central role in drug metabolism and detoxification. Significantly, this microenvironment is emerging as a critical determinant of susceptibility to DILI and HILI. The review delves into the multifaceted interactions within the liver microenvironment, providing valuable insights into the complex mechanisms that underlie DILI and HILI. Furthermore, we discuss potential strategies for mitigating drug-induced liver injury by targeting these influential factors, emphasizing their clinical relevance. By highlighting recent advances and future prospects, our aim is to shed light on the promising avenue of leveraging the liver microenvironment for the prevention and mitigation of DILI and HILI. This deeper understanding is crucial for advancing clinical practices and ensuring patient safety in the realm of DILI and HILI.
Collapse
Affiliation(s)
- Xiaomeng Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Geng
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lin Lin
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lulu Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Changqi Shi
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lan Yan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiwen Cao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peipei Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yong Tan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
25
|
Wang X, Yang W, Lv J, Liao X. Study on the uptake of Gastrodin in the liver. Heliyon 2024; 10:e36031. [PMID: 39229547 PMCID: PMC11369432 DOI: 10.1016/j.heliyon.2024.e36031] [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: 05/26/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/05/2024] Open
Abstract
Background Gastrodin is the active monomer of the Chinese herb Rhizoma Gastrodiae with the largest quantity of active components. Gastrodin is commonly used in the treatment of central nervous system disorders such as headaches and epilepsy due to its sedating and hypnotic properties. Its pharmacological mechanism and clinical application have been extensively explored due to its low toxicity. Methods To investigate the molecular mechanism of hepatic uptake of Gastrodin in rats, animals were randomly assigned to three groups: control group, rifampicin (RIF) group, and adrenalone (ADR) group. Blood samples were collected through the cardiac puncture 90, 180, and 300 min after injection, respectively. Rats were sacrificed 300 min after administration, and liver tissue was collected. Gastrodin concentration was determined by HPLC, and the Kp value was calculated. Results After administering the inhibitors of organic cation transporters (OCTs) and organic anion transporting polypeptides (OATPs), the KP values in the experimental groups were significantly lower compared to the blank control group (P < 0.05). Conclusions These findings imply that Gastrodin may be a substrate for both OCTs and OATPs.
Collapse
Affiliation(s)
- Xing Wang
- College of Medicine, Southwest Jiaotong University, No. 111, Chengdu North 2nd Ring Road, Chengdu, Sichuan, 610003, China
| | - Wenzhu Yang
- School of Life Science and Engineering, Southwest Jiaotong University, No. 111, Chengdu North 2nd Ring Road, Chengdu, Sichuan, 610003, China
| | - Jitong Lv
- School of Life Science and Engineering, Southwest Jiaotong University, No. 111, Chengdu North 2nd Ring Road, Chengdu, Sichuan, 610003, China
| | - Xinya Liao
- School of Life Science and Engineering, Southwest Jiaotong University, No. 111, Chengdu North 2nd Ring Road, Chengdu, Sichuan, 610003, China
| |
Collapse
|
26
|
Bathaei P, Imenshahidi M, Hosseinzadeh H. Effects of Berberis vulgaris, and its active constituent berberine on cytochrome P450: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03326-x. [PMID: 39141022 DOI: 10.1007/s00210-024-03326-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/22/2024] [Indexed: 08/15/2024]
Abstract
The cytochrome P450 (CYP450) family is crucial for metabolizing drugs and natural substances. Numerous compounds, such as pharmaceuticals and dietary items, can influence CYP activity by either enhancing or inhibiting these enzymes, potentially leading to interactions between drugs or between drugs and food. This research explores the impact of barberry and its primary component "berberine" on key human CYP450 enzymes. The text discusses the effects of this plant on the 12 primary human CYP450 enzymes, with summarized data presented in tables. Berberine exerts an influence on the function of various CYP450 isoforms, including CYP3A4/5, CYP2D6, CYP2C9, CYP2E1, CYP1A1/2, and most isoforms within the CYP2B subfamily. Given the significant role of these CYP450 isoforms in metabolizing commonly used drugs and endogenous substances, as well as activating procarcinogens into carcinogenic metabolites, the influence of barberry and its active constituent on these enzymes may impact the pharmacokinetics and toxicity profiles of various compounds. More specifically, regarding the crucial role of CYP2D6 and CYP3A4 in metabolizing clinically used drugs, and the inhibitory effects of berberine on these two CYP450 isoforms, it seems that the most important drug interaction of berberine that should be considered is related to its inhibitory effect on CYP2D6 and CYP3A4. In conclusion, due to the impact of barberry on multiple CYP450 isoforms, healthcare providers should conduct thorough consultations and investigations to ensure patient safety and prevent any potential adverse interactions before recommending the consumption of these herbs. Additional research, particularly clinical trials is crucial for preventing any potentially adverse interactions in patients who consume this herb.
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, 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, Mashhad, Iran.
| |
Collapse
|
27
|
Yin Z, Li P. Association of UGT1A6 gene polymorphisms with sodium valproate-induced tremor in patients with epilepsy. Seizure 2024; 120:56-60. [PMID: 38908142 DOI: 10.1016/j.seizure.2024.06.008] [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: 12/25/2023] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Individual susceptibility to sodium valproate (VPA)-induced tremors may be due to genetic polymorphisms in the gene encoding the uridine diphosphate glucuronosyltransferase (UGT) enzyme, which affec the drug's clinical efficacy and cause toxic side effects. This study aimed to investigate the association between UGT1A6 polymorphisms and VPA-induced tremors in patients with epilepsy. METHODS In total, 128 patients with epilepsy were enrolled. Patients with epilepsy who received VPA were divided into tremor and non-tremor groups. Polymerase chain reaction-restriction fragment length polymorphism was used to investigate the genotype of UGT1A6 polymorphisms. RESULTS Carriers of the UGT1A6 A541G mutant genotype conferred a higher risk of tremor than wild-type carriers (odds ratio 2.128, P = 0.045). Logistic regression analysis showed that the A541G mutant genotype was a significant genetic risk factor for VPA-induced tremors. This suggests that individual susceptibility to VPA-induced tremors may result, at least partially, from genetic variation in UGT1A6 A541G. CONCLUSIONS Patients with epilepsy carrying the UGT1A6 A541G mutant genotype may have VPA-induced tremors, and early detection of this genotype will help guide the clinical individualizsation of VPA treatment.
Collapse
Affiliation(s)
- Zheng Yin
- Qinghai University Graduate School, Xining, China
| | - Pei Li
- Department of Neurology, Qinghai Provincial People's Hospital, Xining, China.
| |
Collapse
|
28
|
Jia Z, Huang J, Yang Y, Yang Y, Lin W, Qu S, Sun N, Zhang W, Han L, Huang J. Establishing national reference materials for genetic testing of cytochrome P450. Pharmacogenet Genomics 2024; 34:175-183. [PMID: 38640061 PMCID: PMC11221791 DOI: 10.1097/fpc.0000000000000532] [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: 11/20/2023] [Accepted: 03/04/2024] [Indexed: 04/21/2024]
Abstract
OBJECTIVES Reference materials for in-vitro diagnostic reagents play a critical role in determining the quality of reagents and ensuring the accuracy of clinical test results. This study aimed to establish a national reference material (NRM) for detecting cytochrome P450 (CYP) genes related to drug metabolism by screening databases on the Chinese population to identify CYP gene polymorphism characteristics. METHODS To prepare the NRM, we used DNA extracted from healthy human immortalized B lymphoblastoid cell lines as the raw material. Samples of these cell lines were obtained from the Chinese Population PGx Gene Polymorphism Biobank. Further, we used Sanger sequencing, next-generation sequencing, and commercial assay kits to validate the polymorphic genotypes. RESULTS Among the CYP superfamily genes, we confirmed 24 riboswitch loci related to drug metabolism, with evidence levels of 1A, 2A, 3, and 4. We confirmed the polymorphic loci and validated their genotypes using various sequencing techniques. Our results were consistent with the polymorphism information of samples obtained from the biobank, thus demonstrating high precision and stability of the established NRM. CONCLUSION An NRM (360 056-202 201) for CYP genetic testing covering 24 loci related to drug metabolism was established and approved to assess in-vitro diagnostic reagents containing CYP family gene polymorphisms and perform clinical inter-room quality evaluations.
Collapse
Affiliation(s)
- Zheng Jia
- Department of In Vitro Diagnostic Reagent, National Institutes for Food and Drug Control, Beijing
| | - Junju Huang
- Department of Academic Collaboration, Research Institute, DAAN Gene Co., Ltd., Guangzhou
| | - Ying Yang
- Research and Development Department, Beijing Anngeen Technologies Co., Ltd., Beijing, China
| | - Yong Yang
- Department of Academic Collaboration, Research Institute, DAAN Gene Co., Ltd., Guangzhou
| | - Wei Lin
- Research and Development Department, Beijing Anngeen Technologies Co., Ltd., Beijing, China
| | - Shoufang Qu
- Department of In Vitro Diagnostic Reagent, National Institutes for Food and Drug Control, Beijing
| | - Nan Sun
- Department of In Vitro Diagnostic Reagent, National Institutes for Food and Drug Control, Beijing
| | - Wenxin Zhang
- Department of In Vitro Diagnostic Reagent, National Institutes for Food and Drug Control, Beijing
| | - Lulu Han
- Department of In Vitro Diagnostic Reagent, National Institutes for Food and Drug Control, Beijing
| | - Jie Huang
- Department of In Vitro Diagnostic Reagent, National Institutes for Food and Drug Control, Beijing
| |
Collapse
|
29
|
Lin J, Liu H, Huang X, Deng Y. Toxicological effects of Honokiol on zebrafish and its underlying mechanism. J Biochem Mol Toxicol 2024; 38:e23789. [PMID: 39097765 DOI: 10.1002/jbt.23789] [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: 02/28/2024] [Revised: 06/25/2024] [Accepted: 07/18/2024] [Indexed: 08/05/2024]
Abstract
The compound Honokiol, derived from the bark of Magnolia officinalis, possesses the ability to induce apoptosis and inhibit cellular damage caused by reactive oxygen species. The objective of this study was to investigate the toxicological and histopathological effects of Honokiol on zebrafish (Danio rerio) through conducting a semistatic acute toxicity test involving immersion in an Honokiol-containing solution. The results showed that the toxic effects of Honokiol on zebrafish were primarily manifested in the liver and gills. When exposed to 0.6 mg/L of Honokiol, it could lead to liver hemorrhage as well as swelling and necrosis of gill tissues, and high concentrations of Honokiol could trigger inflammatory responses. Additionally, research found that Honokiol could induce apoptosis in liver and gill tissues through the P53 pathway and possessed the ability to enhance antioxidation. The present findings significantly contribute to a more profound understanding of the toxic impact of Honokiol and its underlying mechanism, thereby providing a valuable reference for the future safe utilization of Honokiol and related pharmaceutical advancements.
Collapse
Affiliation(s)
- Jue Lin
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, Sichuan, China
| | - Hongli Liu
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaoli Huang
- Department of Aquaculture, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yongqiang Deng
- Fisheries Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, Sichuan, China
| |
Collapse
|
30
|
Guo Y, Long C, Ni J, Zeng J, Wang J, Dai Y, Zhao J. Glucuronidation dynamics of curcumin and tetrahydrocurcumin for differential structures and chemical reactivities in human liver microsome and uridine diphosphate glucuronosyltransferase 2B7. Food Chem 2024; 448:138929. [PMID: 38522299 DOI: 10.1016/j.foodchem.2024.138929] [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: 09/14/2023] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/26/2024]
Abstract
THC is the main metabolite of curcumin with better bioactivity. This study aimed to explore the factors that cause differences in the bioactivity of curcumin and THC. We analyzed the metabolic activities of curcumin and THC and the factors responsible for the differences in their activities by glucuronidation activity assay, LC-MS, HPLC, homologous sequence comparisons, and molecular docking. Curcumin has higher metabolic activity than THC in HLM and UGT2B7, while the keto-enol isomers of curcumin and THC were distinctly different under different pH, and their structural transformations were hypothesized. Furthermore, UGT1A and UGT2B are differential sequences of curcumin and THC in UGTs. The binding sites and patterns of curcumin and THC in UGT2B7 are markedly different. In summary, the difference in keto-enolic interconversion isomerism between curcumin and THC is the main factor causing the difference in their activities, which provides a scientific basis for the development of curcumin.
Collapse
Affiliation(s)
- Yanlei Guo
- West China School of Pharmacy, Sichuan University, 610041 Chengdu, China; Chongqing Academy of Chinese Materia Medica, 400065 Chongqing, China.
| | - Chengyan Long
- Chongqing Academy of Chinese Materia Medica, 400065 Chongqing, China
| | - Jimin Ni
- Chongqing Academy of Chinese Materia Medica, 400065 Chongqing, China
| | - Jin Zeng
- Key Laboratory of Biological Evaluation of Traditional Chinese Medicine Quality of National Administration of Traditional Chinese Medicine/ Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, 610041 Chengdu, China
| | - Jianbo Wang
- Key Laboratory of Biological Evaluation of Traditional Chinese Medicine Quality of National Administration of Traditional Chinese Medicine/ Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, 610041 Chengdu, China
| | - Ying Dai
- Key Laboratory of Biological Evaluation of Traditional Chinese Medicine Quality of National Administration of Traditional Chinese Medicine/ Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, 610041 Chengdu, China
| | - Junning Zhao
- West China School of Pharmacy, Sichuan University, 610041 Chengdu, China; National Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, National Medical Products Administration of China, 100037 Beijing, China; Key Laboratory of Biological Evaluation of Traditional Chinese Medicine Quality of National Administration of Traditional Chinese Medicine/ Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medical Sciences, 610041 Chengdu, China
| |
Collapse
|
31
|
Chen Y, Zhang Y, Jiang Q, Tang C, Wang Q, He C, Zuo Z, Yang C. Effects of whole life-cycle exposure to carbaryl on reproduction of female marine medaka (Oryzias melastigma) and their offspring. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 948:174789. [PMID: 39047820 DOI: 10.1016/j.scitotenv.2024.174789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/06/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
Carbaryl is widely used as a highly effective insecticide which harms the marine environment. This study aimed to assess the reproductive toxicity of chronic carbaryl exposure on female marine medaka and their female offspring. After a 180-day exposure from embryonic period to adulthood, females exhibited reduced attraction to males, decreased ovulation, increased gonadosomatic index and a higher proportion of mature and atretic follicles. These reproductive toxic effects of carbaryl may stem from changes in hormone levels and transcription levels of key genes along the HPG axis. Furthermore, maternal carbaryl exposure had detrimental effects on the offspring. F1 females showed the reproductive disorders similar to those observed in F0 females. The significant changes in the transcription levels of DNA methyltransferase and demethylase genes in the F0 and F1 generations of ovaries indicate changes in their DNA methylation levels. The changes in DNA methylation levels in F1 female marine medaka may lead to changes in the expression of certain reproductive key genes, such as an increase in the transcription level of cyp19a, which may be the reason for F1 reproductive toxicity. These findings indicate that maternal exposure may induce severe generational toxicity through alterations in DNA methylation levels. This study assesses the negative impacts of whole life-cycle carbaryl exposure on the reproductive and developmental processes of female marine medaka and its female offspring, while offering data to support the evaluation of the ecological risk posed by carbaryl in marine ecosystems.
Collapse
Affiliation(s)
- Yuxin Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Yuxuan Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Qun Jiang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Chen Tang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Qian Wang
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, Xiamen University, Xiamen, Fujian 361102, China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Chunyan Yang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China.
| |
Collapse
|
32
|
Ouyang J, Lin D, Chen X, Li Y, Liu Q, Li D, Quan H, Fu X, Wu Q, Wang X, Wu S, Li C, Feng Y, Mao W. Analysis of the chemical constituents and their metabolites in Orthosiphon stamineus Benth. via UHPLC-Q exactive orbitrap-HRMS and AFADESI-MSI techniques. PLoS One 2024; 19:e0304852. [PMID: 38917120 PMCID: PMC11198764 DOI: 10.1371/journal.pone.0304852] [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: 01/23/2024] [Accepted: 05/16/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Known for its strong diuretic properties, the perennial herbaceous plant Orthosiphon stamineus Benth. is believed to preserve the kidney disease. This study compared the boiling water extract with powdered Orthosiphon stamineus Benth. and used a highly sensitive and high resolution UHPLC-Q-Exactive-Orbitrap-HRMS technology to evaluate its chemical composition. RESULTS Furthermore, by monitoring the absorption of prototype components in rat plasma following oral treatment, the beneficial ingredients of the Orthosiphon stamineus Benth. decoction was discovered. Approximately 92 substances underwent a preliminary identification utilizing relevant databases, relevant literature, and reference standards. As the compound differences between the powdered Orthosiphon stamineus Benth. and its water decoction were analyzed, it was found that boiling produced additional compounds, 48 of which were new. 45 blood absorption prototype components and 49 OS metabolites were discovered from rat serum, and a kidney tissue homogenate revealed an additional 28 prototype components. Early differences in the distribution of ferulic acid, cis 4 coumaric acid, and rosmarinic acid were shown using spatial metabolomics. It was elucidated that the renal cortex region is where rosmarinic acid largely acts, offering a theoretical foundation for further studies on the application of OS in the prevention and treatment of illness as well as the preservation of kidney function. SIGNIFICANCE In this study, UHPLC-Q Exactive Orbitrap-HRMS was employed to discern OS's chemical composition, and a rapid, sensitive, and broad-coverage AFADESI-MSI method was developed to visualize the spatial distribution of compounds in tissues.
Collapse
Affiliation(s)
- Jianting Ouyang
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Danyao Lin
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Xuesheng Chen
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Yimeng Li
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Qin Liu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Delun Li
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Haohao Quan
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Xinwen Fu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Qiaoru Wu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Xiaowan Wang
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Shouhai Wu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Chuang Li
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Yi Feng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- Department of Pharmacokinetics of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Wei Mao
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| |
Collapse
|
33
|
Mizutani A, Kobayashi M, Nishi K, Fujita KI, Takahashi K, Muranaka Y, Sato K, Kitamura M, Suzuki C, Nishii R, Shikano N, Magata Y, Ishida Y, Kunishima M, Fukuchi K, Kawai K. Development of radioiodine-labeled mequitazine for evaluation of hepatic CYP2D activity. Front Pharmacol 2024; 15:1397288. [PMID: 38962307 PMCID: PMC11219936 DOI: 10.3389/fphar.2024.1397288] [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: 03/07/2024] [Accepted: 05/31/2024] [Indexed: 07/05/2024] Open
Abstract
Background: As drug-metabolizing enzyme activities are affected by a variety of factors, such as drug-drug interactions, a method to evaluate drug-metabolizing enzyme activities in real time is needed. In this study, we developed a novel SPECT imaging probe for evaluation of hepatic CYP2D activity. Methods: Iodine-123- and 125-labeled 4-iodobenzylmequitazine (123/125I-BMQ) was synthesized with high labeling and purity. CYP isozymes involved in the metabolism of 125I-BMQ in mouse liver microsomes were evaluated, and the utility of 123/125I-was assessed from biological distribution and SPECT imaging evaluation in normal and CYP2D-inhibited mice. Results: In vitro metabolite analysis using mouse liver microsomes showed that 125I-BMQ is specifically metabolized by CYP2D. Biological distribution and SPECT imaging of 123/125I-BMQ in normal mice showed that injection 123/125I-BMQ accumulated early in the liver and was excreted into the gallbladder and intestines. In CYP2D-inhibited mice, accumulation in the liver was increased, but accumulation in the gallbladder and intestines, the excretory organ, was delayed. Since only metabolites of 125I-BMQ are detected in bile, visualization and measuring of the accumulation of metabolites over time in the intestine, where bile is excreted, could predict the amount of metabolites produced in the body and evaluate CYP2D activity, which would be useful in determining the dosage of various drugs metabolized by CYP2D. Conclusion: 123/125I-BMQ is useful as a SPECT imaging probe for comprehensive and direct assessment of hepatic CYP2D activity in a minimally invasive and simple approach.
Collapse
Affiliation(s)
- Asuka Mizutani
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Masato Kobayashi
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kodai Nishi
- Department of Radioisotope Medicine, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Ken-ichi Fujita
- Division of Cancer Genome and Pharmacotherapy, Department of Clinical Pharmacy, Showa University School of Pharmacy, Tokyo, Japan
| | - Kotaro Takahashi
- Department of Radiologic Technology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yuka Muranaka
- Department of Radiological Technology, Faculty of Health Science, Juntendo University, Tokyo, Japan
| | - Kakeru Sato
- Division of Health Sciences, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Masanori Kitamura
- Faculty of Pharmaceutical Sciences, Matsuyama University, Matsuyama, Japan
| | - Chie Suzuki
- Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ryuichi Nishii
- Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Naoto Shikano
- Department of Radiological Sciences, Ibaraki Prefectural University of Health Sciences, Ibaraki, Japan
| | - Yasuhiro Magata
- Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yasushi Ishida
- Department of Psychiatry, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | | | - Kazuki Fukuchi
- Division of Medical Technology and Science, Department of Medical Physics and Engineering, Course of Health Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Keiichi Kawai
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
- Biomedical Imaging Research Center, University of Fukui, Fukui, Japan
| |
Collapse
|
34
|
Khalil SM, Qin X, Hakenjos JM, Wang J, Hu Z, Liu X, Wang J, Maletic-Savatic M, MacKenzie KR, Matzuk MM, Li F. MALDI Imaging Mass Spectrometry Visualizes the Distribution of Antidepressant Duloxetine and Its Major Metabolites in Mouse Brain, Liver, Kidney, and Spleen Tissues. Drug Metab Dispos 2024; 52:673-680. [PMID: 38658163 PMCID: PMC11185819 DOI: 10.1124/dmd.124.001719] [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: 03/11/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024] Open
Abstract
Imaging mass spectrometry (IMS) is a powerful tool for mapping the spatial distribution of unlabeled drugs and metabolites that may find application in assessing drug delivery, explaining drug efficacy, and identifying potential toxicity. This study focuses on determining the spatial distribution of the antidepressant duloxetine, which is widely prescribed despite common adverse effects (liver injury, constant headaches) whose mechanisms are not fully understood. We used high-resolution IMS with matrix-assisted laser desorption/ionization to examine the distribution of duloxetine and its major metabolites in four mouse organs where it may contribute to efficacy or toxicity: brain, liver, kidney, and spleen. In none of these tissues is duloxetine or its metabolites homogeneously distributed, which has implications for both efficacy and toxicity. We found duloxetine to be similarly distributed in spleen red pulp and white pulp but differentially distributed in different anatomic regions of the liver, kidney, and brain, with dose-dependent patterns. Comparison with hematoxylin and eosin staining of tissue sections reveals that the ion images of endogenous lipids help delineate anatomic regions in the brain and kidney, while heme ion images assist in differentiating regions within the spleen. These endogenous metabolites may serve as a valuable resource for examining the spatial distribution of other drugs in tissues when staining images are not available. These findings may facilitate future mechanistic studies of the therapeutic and adverse effects of duloxetine. In the current work, we did not perform absolute quantification of duloxetine, which will be reported in due course. SIGNIFICANCE STATEMENT: The study utilized imaging mass spectrometry to examine the spatial distribution of duloxetine and its primary metabolites in mouse brain, liver, kidney, and spleen. These results may pave the way for future investigations into the mechanisms behind duloxetine's therapeutic and adverse effects. Furthermore, the mass spectrometry images of specific endogenous metabolites such as heme could be valuable in analyzing the spatial distribution of other drugs within tissues in scenarios where histological staining images are unavailable.
Collapse
Affiliation(s)
- Saleh M Khalil
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Xuan Qin
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - John M Hakenjos
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Jian Wang
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Zhaoyong Hu
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Xinli Liu
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Jin Wang
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Mirjana Maletic-Savatic
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Kevin R MacKenzie
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Martin M Matzuk
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| | - Feng Li
- Center for Drug Discovery, Department of Pathology and Immunology (S.M.K., X.Q., J.M.H., Jia.W., M.M.-S., K.R.M., M.M.M., F.L.), NMR and Drug Metabolism Core, Advanced Technology Cores (X.Q., J.M.H., Jia.W., K.R.M., F.L.), Department of Biochemistry and Molecular Pharmacology (Jin.W., K.R.M., M.M.M., F.L.), Department of Pediatrics (S.M.K., M.M.-S.), and Nephrology Division, Department of Medicine (Z.H.), Baylor College of Medicine, Houston, Texas; Jan and Dan Duncan Neurologic Research Institute, Texas Children's Hospital, Houston, Texas (M.M.-S.); and Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (X.L.)
| |
Collapse
|
35
|
Cho S, Jo H, Hwang YJ, Kim C, Jo YH, Yun JW. Potential impact of underlying diseases influencing ADME in nonclinical safety assessment. Food Chem Toxicol 2024; 188:114636. [PMID: 38582343 DOI: 10.1016/j.fct.2024.114636] [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: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
Nonclinical studies involve in vitro, in silico, and in vivo experiments to assess the toxicokinetics, toxicology, and safety pharmacology of drugs according to regulatory requirements by a national or international authority. In this review, we summarize the potential effects of various underlying diseases governing the absorption, distribution, metabolism, and excretion (ADME) of drugs to consider the use of animal models of diseases in nonclinical trials. Obesity models showed alterations in hepatic metabolizing enzymes, transporters, and renal pathophysiology, which increase the risk of drug-induced toxicity. Diabetes models displayed changes in hepatic metabolizing enzymes, transporters, and glomerular filtration rates (GFR), leading to variability in drug responses and susceptibility to toxicity. Animal models of advanced age exhibited impairment of drug metabolism and kidney function, thereby reducing the drug-metabolizing capacity and clearance. Along with changes in hepatic metabolic enzymes, animal models of metabolic syndrome-related hypertension showed renal dysfunction, resulting in a reduced GFR and urinary excretion of drugs. Taken together, underlying diseases can induce dysfunction of organs involved in the ADME of drugs, ultimately affecting toxicity. Therefore, the use of animal models of representative underlying diseases in nonclinical toxicity studies can be considered to improve the predictability of drug side effects before clinical trials.
Collapse
Affiliation(s)
- Sumin Cho
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Harin Jo
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yeon Jeong Hwang
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Changuk Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Yong Hyeon Jo
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
36
|
Sun B, Liang Z, Wang Y, Yu Y, Zhou X, Geng X, Li B. A 3D spheroid model of quadruple cell co-culture with improved liver functions for hepatotoxicity prediction. Toxicology 2024; 505:153829. [PMID: 38740170 DOI: 10.1016/j.tox.2024.153829] [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: 02/22/2024] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
Drug-induced liver injury (DILI) is one of the major concerns during drug development. Wide acceptance of the 3 R principles and the innovation of in-vitro techniques have introduced various novel model options, among which the three-dimensional (3D) cell spheroid cultures have shown a promising prospect in DILI prediction. The present study developed a 3D quadruple cell co-culture liver spheroid model for DILI prediction via self-assembly. Induction by phorbol 12-myristate 13-acetate at the concentration of 15.42 ng/mL for 48 hours with a following 24-hour rest period was used for THP-1 cell differentiation, resulting in credible macrophagic phenotypes. HepG2 cells, PUMC-HUVEC-T1 cells, THP-1-originated macrophages, and human hepatic stellate cells were selected as the components, which exhibited adaptability in the designated spheroid culture conditions. Following establishment, the characterization demonstrated the competence of the model in long-term stability reflected by the maintenance of morphology, viability, cellular integration, and cell-cell junctions for at least six days, as well as the reliable liver-specific functions including superior albumin and urea secretion, improved drug metabolic enzyme expression and CYP3A4 activity, and the expression of MRP2, BSEP, and P-GP accompanied by the bile acid efflux transport function. In the comparative testing using 22 DILI-positive and 5 DILI-negative compounds among the novel 3D co-culture model, 3D HepG2 spheroids, and 2D HepG2 monolayers, the 3D culture method significantly enhanced the model sensitivity to compound cytotoxicity compared to the 2D form. The novel co-culture liver spheroid model exhibited higher overall predictive power with margin of safety as the classifying tool. In addition, the non-parenchymal cell components could amplify the toxicity of isoniazid in the 3D model, suggesting their potential mediating role in immune-mediated toxicity. The proof-of-concept experiments demonstrated the capability of the model in replicating drug-induced lipid dysregulation, bile acid efflux inhibition, and α-SMA upregulation, which are the key features of liver steatosis and phospholipidosis, cholestasis, and fibrosis, respectively. Overall, the novel 3D quadruple cell co-culture spheroid model is a reliable and readily available option for DILI prediction.
Collapse
Affiliation(s)
- Baiyang Sun
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China
| | - Zihe Liang
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China
| | - Yupeng Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China
| | - Yue Yu
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China
| | - Xiaobing Zhou
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China
| | - Xingchao Geng
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control, Beijing Key Laboratory for Nonclinical Safety Evaluation of Drugs, Beijing 100176, China.
| | - Bo Li
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; National Institutes for Food and Drug Control, Beijing 102629, China.
| |
Collapse
|
37
|
Yoshizaki M, Kuriya Y, Yamamoto M, Watanabe N, Araki M. Development of method using language processing techniques for extracting information on drug-health food product interactions. Br J Clin Pharmacol 2024; 90:1514-1524. [PMID: 38504605 DOI: 10.1111/bcp.16032] [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: 10/20/2023] [Revised: 12/25/2023] [Accepted: 01/22/2024] [Indexed: 03/21/2024] Open
Abstract
AIMS Health food products (HFPs) are foods and products related to maintaining and promoting health. HFPs may sometimes cause unforeseen adverse health effects by interacting with drugs. Considering the importance of information on the interactions between HFPs and drugs, this study aimed to establish a workflow to extract information on Drug-HFP Interactions (DHIs) from open resources. METHODS First, Information on drugs, enzymes, their interactions, and known DHIs was collected from multiple public databases and literature sources. Next, a network consisted of enzymes, HFP, and drugs was constructed, assuming enzymes as candidates for hubs in Drug-HFP interactions (Method 1). Furthermore, we developed methods to analyze the biomedical context of each drug and HFP to predict potential DHIs out of the DHIs obtained in Method 1 by applying BioWordVec, a widely used biomedical terminology quantifier (Method 2-1 and 2-2). RESULTS 44,965 DHIs (30% known) were identified in Method 1, including 38 metabolic enzymes, 157 HFPs, and 1256 drugs. Method 2-1 selected 7401 DHIs (17% known) from the DHIs of Method 1, while Method 2-2 chose 2819 DHIs (30% known). Based on the different assumptions in these methods where Method 2-1 specifically selects HFPs interacting with specific enzymes and Method 2-2 specifically selects HFPs with similar function with drugs, the propsed methods resulted in extracting a wide variety of DHIs. CONCLUSIONS By integrating the results of language processing techniques with those of the network analysis, a workflow to efficiently extract unknown and known DHIs was constructed.
Collapse
Affiliation(s)
- Mari Yoshizaki
- Biological Science and Technology, Life and Materials Systems Engineering, Graduate School of Advanced Technology and Science, Tokushima University, Tokushima City, Tokushima Prefecture, Japan
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Settsu City, Osaka Prefecture, Japan
| | - Yuki Kuriya
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Settsu City, Osaka Prefecture, Japan
| | - Masaki Yamamoto
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Settsu City, Osaka Prefecture, Japan
| | - Naoki Watanabe
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Settsu City, Osaka Prefecture, Japan
| | - Michihiro Araki
- Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition, Settsu City, Osaka Prefecture, Japan
| |
Collapse
|
38
|
Kandalgaonkar MR, Kumar V, Vijay‐Kumar M. Digestive dynamics: Unveiling interplay between the gut microbiota and the liver in macronutrient metabolism and hepatic metabolic health. Physiol Rep 2024; 12:e16114. [PMID: 38886098 PMCID: PMC11182692 DOI: 10.14814/phy2.16114] [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: 04/11/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Although the liver is the largest metabolic organ in the body, it is not alone in functionality and is assisted by "an organ inside an organ," the gut microbiota. This review attempts to shed light on the partnership between the liver and the gut microbiota in the metabolism of macronutrients (i.e., proteins, carbohydrates, and lipids). All nutrients absorbed by the small intestines are delivered to the liver for further metabolism. Undigested food that enters the colon is metabolized further by the gut microbiota that produces secondary metabolites, which are absorbed into portal circulation and reach the liver. These microbiota-derived metabolites and co-metabolites include ammonia, hydrogen sulfide, short-chain fatty acids, secondary bile acids, and trimethylamine N-oxide. Further, the liver produces several compounds, such as bile acids that can alter the gut microbial composition, which can in turn influence liver health. This review focuses on the metabolism of these microbiota metabolites and their influence on host physiology. Furthermore, the review briefly delineates the effect of the portosystemic shunt on the gut microbiota-liver axis, and current understanding of the treatments to target the gut microbiota-liver axis.
Collapse
Affiliation(s)
- Mrunmayee R. Kandalgaonkar
- Department of Physiology and PharmacologyUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
| | - Virender Kumar
- College of Pharmacy and Pharmaceutical SciencesUniversity of ToledoToledoOhioUSA
| | - Matam Vijay‐Kumar
- Department of Physiology and PharmacologyUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
| |
Collapse
|
39
|
Mahon E, Marsh O, Uriarte A, Stabile F. The effect of oral zonisamide treatment on serum phenobarbital concentrations in epileptic dogs. Front Vet Sci 2024; 11:1389615. [PMID: 38868500 PMCID: PMC11168201 DOI: 10.3389/fvets.2024.1389615] [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: 02/21/2024] [Accepted: 04/26/2024] [Indexed: 06/14/2024] Open
Abstract
Zonisamide is used in dogs for the treatment of epileptic seizures. It is predominantly metabolised by CYP450 hepatic enzymes. When used concurrently with phenobarbital (PB), zonisamide clearance is increased and its elimination half-life decreases. However, the effect that zonisamide may have on serum PB concentrations in dogs has not been previously described. Eight dogs diagnosed with idiopathic epilepsy and two dogs with structural epilepsy commenced zonisamide at 8.0 mg/kg/12 h [7.4-10 mg/kg/12 h] following an increase in the frequency of epileptic seizures. Nine dogs were receiving PB every 12 h (4.2 mg/kg/12 h [3.8-6 mg/kg/12 h]), and one dog was receiving PB every 8 h (6 mg/kg/8 h). Following the addition of zonisamide and despite no increase in PB dosage, an increase in phenobarbital serum PB concentration was observed in 9 out of 10 dogs in subsequent measurements. In five dogs, phenobarbital serum concentrations were raised to concentrations higher than the reported hepatotoxic concentrations (trough>35 mg/L). This required a reduction in daily doses of PB. This case series suggests that zonisamide affects the metabolism of PB and causes an increase in PB serum concentrations over time.
Collapse
Affiliation(s)
- Elizabeth Mahon
- Department of Neurology and Neurosurgery, Southfields Veterinary Specialists, Part of Linnaeus Ltd, Essex, United Kingdom
| | - Oliver Marsh
- Department of Neurology and Neurosurgery, Dick White Referrals, Part of Linnaeus Ltd, Newmarket, United Kingdom
| | - Ane Uriarte
- Department of Neurology and Neurosurgery, Southfields Veterinary Specialists, Part of Linnaeus Ltd, Essex, United Kingdom
| | - Fabio Stabile
- Department of Neurology and Neurosurgery, Wear Referrals, Part of Linnaeus Ltd, Bradbury, United Kingdom
| |
Collapse
|
40
|
Silva IMD, Vacario BGL, Okuyama NCM, Barcelos GRM, Fuganti PE, Guembarovski RL, Cólus IMDS, Serpeloni JM. Polymorphisms in drug-metabolizing genes and urinary bladder cancer susceptibility and prognosis: Possible impacts and future management. Gene 2024; 907:148252. [PMID: 38350514 DOI: 10.1016/j.gene.2024.148252] [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: 10/09/2023] [Revised: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 02/15/2024]
Abstract
Epidemiological studies have shown the association of genetic variants with risks of occupational and environmentally induced cancers, including bladder (BC). The current review summarizes the effects of variants in genes encoding phase I and II enzymes in well-designed studies to highlight their contribution to BC susceptibility and prognosis. Polymorphisms in genes codifying drug-metabolizing proteins are of particular interest because of their involvement in the metabolism of exogenous genotoxic compounds, such as tobacco and agrochemicals. The prognosis between muscle-invasive and non-muscle-invasive diseases is very different, and it is difficult to predict which will progress worse. Web of Science, PubMed, and Medline were searched to identify studies published between January 1, 2010, and February 2023. We included 73 eligible studies, more than 300 polymorphisms, and 46 genes/loci. The most studied candidate genes/loci of phase I metabolism were CYP1B1, CYP1A1, CYP1A2, CYP3A4, CYP2D6, CYP2A6, CYP3E1, and ALDH2, and those in phase II were GSTM1, GSTT1, NAT2, GSTP1, GSTA1, GSTO1, and UGT1A1. We used the 46 genes to construct a network of proteins and to evaluate their biological functions based on the Reactome and KEGG databases. Lastly, we assessed their expression in different tissues, including normal bladder and BC samples. The drug-metabolizing pathway plays a relevant role in BC, and our review discusses a list of genes that could provide clues for further exploration of susceptibility and prognostic biomarkers.
Collapse
Affiliation(s)
- Isabely Mayara da Silva
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil.
| | - Beatriz Geovana Leite Vacario
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil; Center of Health Sciences, State University of West Paraná (UNIOESTE), Francisco Beltrão-Paraná, 85605-010, Brazil.
| | - Nádia Calvo Martins Okuyama
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil.
| | - Gustavo Rafael Mazzaron Barcelos
- Department of Biosciences, Institute for Health and Society, Federal University of São Paulo (UNIFESP), Santos 11.060-001, Brazil.
| | | | - Roberta Losi Guembarovski
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil.
| | - Ilce Mara de Syllos Cólus
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil.
| | - Juliana Mara Serpeloni
- Department of General Biology, Center of Biological Sciences, State University of Londrina (UEL), Londrina 86057-970, Brazil.
| |
Collapse
|
41
|
Albadry M, Küttner J, Grzegorzewski J, Dirsch O, Kindler E, Klopfleisch R, Liska V, Moulisova V, Nickel S, Palek R, Rosendorf J, Saalfeld S, Settmacher U, Tautenhahn HM, König M, Dahmen U. Cross-species variability in lobular geometry and cytochrome P450 hepatic zonation: insights into CYP1A2, CYP2D6, CYP2E1 and CYP3A4. Front Pharmacol 2024; 15:1404938. [PMID: 38818378 PMCID: PMC11137285 DOI: 10.3389/fphar.2024.1404938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 04/29/2024] [Indexed: 06/01/2024] Open
Abstract
There is a lack of systematic research exploring cross-species variation in liver lobular geometry and zonation patterns of critical drug-metabolizing enzymes, a knowledge gap essential for translational studies. This study investigated the critical interplay between lobular geometry and key cytochrome P450 (CYP) zonation in four species: mouse, rat, pig, and human. We developed an automated pipeline based on whole slide images (WSI) of hematoxylin-eosin-stained liver sections and immunohistochemistry. This pipeline allows accurate quantification of both lobular geometry and zonation patterns of essential CYP proteins. Our analysis of CYP zonal expression shows that all CYP enzymes (besides CYP2D6 with panlobular expression) were observed in the pericentral region in all species, but with distinct differences. Comparison of normalized gradient intensity shows a high similarity between mice and humans, followed by rats. Specifically, CYP1A2 was expressed throughout the pericentral region in mice and humans, whereas it was restricted to a narrow pericentral rim in rats and showed a panlobular pattern in pigs. Similarly, CYP3A4 is present in the pericentral region, but its extent varies considerably in rats and appears panlobular in pigs. CYP2D6 zonal expression consistently shows a panlobular pattern in all species, although the intensity varies. CYP2E1 zonal expression covered the entire pericentral region with extension into the midzone in all four species, suggesting its potential for further cross-species analysis. Analysis of lobular geometry revealed an increase in lobular size with increasing species size, whereas lobular compactness was similar. Based on our results, zonated CYP expression in mice is most similar to humans. Therefore, mice appear to be the most appropriate species for drug metabolism studies unless larger species are required for other purposes, e.g., surgical reasons. CYP selection should be based on species, with CYP2E1 and CYP2D6 being the most preferable to compare four species. CYP1A2 could be considered as an additional CYP for rodent versus human comparisons, and CYP3A4 for mouse/human comparisons. In conclusion, our image analysis pipeline together with suggestions for species and CYP selection can serve to improve future cross-species and translational drug metabolism studies.
Collapse
Affiliation(s)
- Mohamed Albadry
- Department of General, Visceral and Vascular Surgery, Experimental Transplantation Surgery, Jena University Hospital, Jena, Germany
- Department of Pathology, Faculty of Veterinary Medicine, Menoufia University, Shebin Elkom, Menoufia, Egypt
| | - Jonas Küttner
- Department of General, Visceral and Vascular Surgery, Experimental Transplantation Surgery, Jena University Hospital, Jena, Germany
- Institute for Theoretical Biology, Institute für Biologie, Systems Medicine of the Liver, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jan Grzegorzewski
- Institute for Theoretical Biology, Institute für Biologie, Systems Medicine of the Liver, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Olaf Dirsch
- Institute for Pathology, BG Klinikum Unfallkrankenhaus Berlin, Berlin, Germany
| | - Eva Kindler
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany
| | - Robert Klopfleisch
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Vaclav Liska
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
- Department of Surgery, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Vladimira Moulisova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Sandra Nickel
- Clinic and Polyclinic for Visceral, Transplantation, Thoracic and Vascular Surgery, Leipzig University Hospital, Leipzig, Germany
| | - Richard Palek
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
- Department of Surgery, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Jachym Rosendorf
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
- Department of Surgery, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Sylvia Saalfeld
- Institute of Biomedical Engineering and Informatics, Ilmenau University of Technology, Ilmenau, Germany
| | - Utz Settmacher
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany
| | - Hans-Michael Tautenhahn
- Department of General, Visceral and Vascular Surgery, Experimental Transplantation Surgery, Jena University Hospital, Jena, Germany
- Clinic and Polyclinic for Visceral, Transplantation, Thoracic and Vascular Surgery, Leipzig University Hospital, Leipzig, Germany
| | - Matthias König
- Institute for Theoretical Biology, Institute für Biologie, Systems Medicine of the Liver, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Uta Dahmen
- Department of General, Visceral and Vascular Surgery, Experimental Transplantation Surgery, Jena University Hospital, Jena, Germany
| |
Collapse
|
42
|
Ishihara A. Hydroxylated polychlorinated biphenyls may affect the thyroid hormone-induced brain development during metamorphosis of Xenopus laevis by disturbing the expression of matrix metalloproteinases. Mol Biol Rep 2024; 51:624. [PMID: 38710963 DOI: 10.1007/s11033-024-09555-w] [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: 01/21/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Thyroid hormones are primarily responsible for the brain development in perinatal mammals. However, this process can be inhibited by external factors such as environmental chemicals. Perinatal mammals are viviparous, which makes direct fetal examination difficult. METHODS We used metamorphic amphibians, which exhibit many similarities to perinatal mammals, as an experimental system. Therefore, using metamorphic amphibians, we characterized the gene expression of matrix metalloproteinases, which play an important role in brain development. RESULTS The expression of many matrix metalloproteinases (mmps) was characteristically induced during metamorphosis. We also found that the expression of many mmps was induced by T3 and markedly inhibited by hydroxylated polychlorinated biphenyls (PCBs). CONCLUSION Overall, our findings suggest that hydroxylated PCBs disrupt normal brain development by disturbing the gene expression of mmps.
Collapse
Affiliation(s)
- Akinori Ishihara
- Department of Biological Science, Faculty of Science, Shizuoka University, Shizuoka, 422-8529, Japan.
| |
Collapse
|
43
|
Courville J, Roupe K, Arold G. Re-discover the value of protein binding assessments in hepatic and renal impairment studies and its contributions in drug labels and dose decisions. Clin Transl Sci 2024; 17:e13810. [PMID: 38716900 PMCID: PMC11077687 DOI: 10.1111/cts.13810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/05/2024] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
One of the key pharmacokinetic properties of most small molecule drugs is their ability to bind to serum proteins. Unbound or free drug is responsible for pharmacological activity while the balance between free and bound drug can impact drug distribution, elimination, and other safety parameters. In the hepatic impairment (HI) and renal impairment (RI) clinical studies, unbound drug concentration is often assessed; however, the relevance and impact of the protein binding (PB) results is largely limited. We analyzed published clinical safety and pharmacokinetic studies in subjects with HI or RI with PB assessment up to October 2022 and summarized the contribution of PB results on their label dose recommendations. Among drugs with HI publication, 32% (17/53) associated product labels include PB results in HI section. Of these, the majority (9/17, 53%) recommend dose adjustments consistent with observed PB change. Among drugs with RI publication, 27% (12/44) of associated product labels include PB results in RI section with the majority (7/12, 58%) recommending no dose adjustment, consistent with the reported absence of PB change. PB results were found to be consistent with a tailored dose recommendation in 53% and 58% of the approved labels for HI and RI section, respectively. We further discussed the interpretation challenges of PB results, explored treatment decision factors including total drug concentration, exposure-response relationships, and safety considerations in these case examples. Collectively, comprehending the alterations in free drug levels in HI and RI informs treatment decision through a risk-based approach.
Collapse
Affiliation(s)
- Jocelyn Courville
- Clinical Pharmacology—Drug Development SolutionICON plcBlue BellPennsylvaniaUSA
| | - Kathryn Roupe
- Clinical Pharmacology, PharmacokineticsWorldwide Clinical TrialsAustinTexasUSA
| | - Gerhard Arold
- Clinical Pharmacology—Drug Development SolutionICON plcLangenGermany
| |
Collapse
|
44
|
Kim R, Sung JH. Microfluidic gut-axis-on-a-chip models for pharmacokinetic-based disease models. BIOMICROFLUIDICS 2024; 18:031507. [PMID: 38947281 PMCID: PMC11210976 DOI: 10.1063/5.0206271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/05/2024] [Indexed: 07/02/2024]
Abstract
The low success rate of new drugs transitioning from animal testing to human clinical trials necessitates the development of more accurate and representative in vitro models. Recent advances in multi-organ-on-a-chip technology offer promising avenues for studying complex organ-organ interactions. Gut-liver-on-a-chip systems hold particular promise for mimicking the intricate interplay between the gut and liver, which play crucial roles in nutrient absorption, drug metabolism, detoxification, and immune response. Here, we discuss the key components of the gut-liver axis, including the gut epithelium, liver cells, gut microbiota, and their roles in the organ functions. We then explore the potential of gut-liver-on-a-chip models to replicate the intricate interactions between the two organs for pharmacokinetic studies and their expansion to more complicated multi-organ models. Finally, we provide perspectives and future directions for developing more physiologically relevant gut-liver-axis models for more efficient drug development, studying liver diseases, and personalizing treatment strategies.
Collapse
Affiliation(s)
- Raehyun Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul 04066, Republic of Korea
| |
Collapse
|
45
|
Rodríguez Castillo B, Cendrós M, Ciudad CJ, Sabater A. Comprehensive Analysis of Drug Utilization Patterns, Gender Disparities, Lifestyle Influences, and Genetic Factors: Insights from Elderly Cohort Using g-Nomic ® Software. Pharmaceuticals (Basel) 2024; 17:565. [PMID: 38794134 PMCID: PMC11123674 DOI: 10.3390/ph17050565] [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: 02/22/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Polypharmacy is a global healthcare concern, especially among the elderly, leading to drug interactions and adverse reactions, which are significant causes of death in developed nations. However, the integration of pharmacogenetics can help mitigate these risks. In this study, the data from 483 patients, primarily elderly and polymedicated, were analyzed using Eugenomic®'s personalized prescription software, g-Nomic®. The most prescribed drug classes included antihypertensives, platelet aggregation inhibitors, cholesterol-lowering drugs, and gastroprotective medications. Drug-lifestyle interactions primarily involved inhibitions but also included inductions. Interactions were analyzed considering gender. Significant genetic variants identified in the study encompassed ABCB1, SLCO1B1, CYP2C19, CYP2C9, CYP2D6, CYP3A4, ABCG2, NAT2, SLC22A1, and G6PD. To prevent adverse reactions and enhance medication effectiveness, it is strongly recommended to consider pharmacogenetics testing. This approach shows great promise in optimizing medication regimens and ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Bárbara Rodríguez Castillo
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universidad de Barcelona, 08007 Barcelona, Spain (C.J.C.)
| | - Marc Cendrós
- Technical Department, Eugenomic, 08029 Barcelona, Spain
| | - Carlos J. Ciudad
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universidad de Barcelona, 08007 Barcelona, Spain (C.J.C.)
| | - Ana Sabater
- Technical Department, Eugenomic, 08029 Barcelona, Spain
| |
Collapse
|
46
|
Song S, Gao Y, Feng S, Cheng Z, Huang H, Xue J, Zhang T, Sun H. Widespread occurrence of two typical N, N'-substituted p-phenylenediamines and their quinones in humans: Association with oxidative stress and liver damage. JOURNAL OF HAZARDOUS MATERIALS 2024; 468:133835. [PMID: 38394895 DOI: 10.1016/j.jhazmat.2024.133835] [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: 12/06/2023] [Revised: 02/11/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024]
Abstract
While N, N'-substituted p-phenylenediamines (PPDs) and their quinone derivatives (PPDQs) have been widely detected in the environment, there is currently limited data on their occurrence in humans. In this study, we conducted the first serum analysis of two PPDs and PPDQs in the healthy and secondary nonalcoholic fatty liver disease (S-NAFLD) cohorts in South China. The concentrations of four oxidative stress biomarkers (OSBs), namely, 8-iso-prostaglandin F2α (8-PGF2α), 11β-prostaglandin F2α (11-PGF2α), 15(R)-prostaglandin F2α (15-PGF2α), and 8-hydroxy-2'-deoxyguanosine in serum samples were also measured. Results showed that N-(1,3-dimethybutyl)-N'-phenyl-p-phenylenediamine (6PPD) quinone was the predominant target analytes both in the healthy and S-NAFLD cohorts, with the median concentrations of 0.13 and 0.20 ng/mL, respectively. Significant (p < 0.05) and positive correlations were found between 6PPD concentration and 8-PGF2α, 11-PGF2α, and 15-PGF2α in both the healthy and S-NAFLD cohorts, indicating that 6PPD may be associated with lipid oxidative damage. In addition, concentrations of 6PPD in serum were associated significantly linked with total bilirubin (β = 0.180 μmol/L, 95%CI: 0.036-0.396) and direct bilirubin (DBIL, β = 0.321 μmol/L, 95%CI: 0.035-0.677) related to hepatotoxicity. Furthermore, 8-PGF2α, 11-PGF2α, and 15-PGF2α mediated 17.1%, 24.5%, and 16.6% of 6PPD-associated DBIL elevations, respectively. Conclusively, this study provides novel insights into human exposure to and hepatotoxicity assessment of PPDs and PPDQs.
Collapse
Affiliation(s)
- Shiming Song
- School of Chemistry and Environment, Jiaying University, Mei Zhou 514015, China; School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yanxia Gao
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Shuai Feng
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Zhipeng Cheng
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Haibao Huang
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China
| | - Jingchuan Xue
- Guangdong Provincial Key Laboratory of Water Quality Improvement and Ecological Restoration for Watersheds, Institute of Environmental and Ecological Engineering, Guangdong University of Technology, Guangzhou 510006, China
| | - Tao Zhang
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Hongwen Sun
- MOE Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| |
Collapse
|
47
|
Zhang H, Lu C, Yao Q, Jiao Q. In silico study to identify novel NEK7 inhibitors from natural sources by a combination strategy. Mol Divers 2024:10.1007/s11030-024-10838-4. [PMID: 38598164 DOI: 10.1007/s11030-024-10838-4] [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: 09/02/2023] [Accepted: 03/06/2024] [Indexed: 04/11/2024]
Abstract
Cancer poses a significant global health challenge and significantly contributes to mortality. NEK7, related to the NIMA protein kinase family, plays a crucial role in spindle assembly and cell division. The dysregulation of NEK7 is closely linked to the onset and progression of various cancers, especially colon and breast cancer, making it a promising target for cancer therapy. Nevertheless, the shortage of high-quality NEK7 inhibitors highlights the need for new therapeutic strategies. In this study, we utilized a multidisciplinary approach, including virtual screening, molecular docking, pharmacokinetics, molecular dynamics simulations (MDs), and MM/PBSA calculations, to evaluate natural compounds as NEK7 inhibitors comprehensively. Through various docking strategies, we identified three natural compounds: (-)-balanol, digallic acid, and scutellarin. Molecular docking revealed significant interactions at residues such as GLU112 and ALA114, with docking scores of -15.054, -13.059, and -11.547 kcal/mol, respectively, highlighting their potential as NEK7 inhibitors. MDs confirmed the stability of these compounds at the NEK7-binding site. Hydrogen bond analysis during simulations revealed consistent interactions, supporting their strong binding capacity. MM/PBSA analysis identified other crucial amino acids contributing to binding affinity, including ILE20, VAL28, ILE75, LEU93, ALA94, LYS143, PHE148, LEU160, and THR161, crucial for stabilizing the complex. This research demonstrated that these compounds exceeded dabrafenib in binding energy, according to MM/PBSA calculations, underscoring their effectiveness as NEK7 inhibitors. ADME/T predictions showed lower oral toxicity for these compounds, suggesting their potential for further development. This study highlights the promise of these natural compounds as bases for creating more potent derivatives with significant biological activities, paving the way for future experimental validation.
Collapse
Affiliation(s)
- Heng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Chenhong Lu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Qilong Yao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Qingcai Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
48
|
Balloni A, Tini A, Prospero E, Busardò FP, Huestis MA, Lo Faro AF. Exposure to Synthetic Psychoactive Substances: A Potential Cause for Increased Human Hepatotoxicity Markers. Clin Chem 2024; 70:597-628. [PMID: 38427953 DOI: 10.1093/clinchem/hvad210] [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: 08/16/2023] [Accepted: 10/20/2023] [Indexed: 03/03/2024]
Abstract
BACKGROUND Approximately 30 million people worldwide consume new psychoactive substances (NPS), creating a serious public health issue due to their toxicity and potency. Drug-induced liver injury is the leading cause of liver disease, responsible for 4% of global deaths each year. CONTENT A systematic literature search revealed 64 case reports, in vitro and in vivo studies on NPS hepatotoxicity. Maximum elevated concentrations of aspartate aminotransferase (136 to 15 632 U/L), alanine transaminase (121.5 to 9162 U/L), total bilirubin (0.7 to 702 mg/dL; 0.04 to 39.03 mmol/L), direct (0.2-15.1 mg/dL; 0.01-0.84 mmol/L) and indirect (5.3 mg/dL; 0.29 mmol/L) bilirubin, alkaline phosphatase (79-260 U/L), and gamma-glutamyltransferase (260 U/L) were observed as biochemical markers of liver damage, with acute and fulminant liver failure the major toxic effects described in the NPS case reports. In vitro laboratory studies and subsequent in vivo NPS exposure studies on rats and mice provide data on potential mechanisms of toxicity. Oxidative stress, plasma membrane stability, and cellular energy changes led to apoptosis and cell death. Experimental studies of human liver microsome incubation with synthetic NPS, with and without specific cytochrome P450 inhibitors, highlighted specific enzyme inhibitions and potential drug-drug interactions leading to hepatotoxicity. SUMMARY Mild to severe hepatotoxic effects following synthetic NPS exposure were described in case reports. In diagnosing the etiology of liver damage, synthetic NPS exposure should be considered as part of the differential diagnosis. Identification of NPS toxicity is important for educating patients on the dangers of NPS consumption and to suggest promising treatments for observed hepatotoxicity.
Collapse
Affiliation(s)
- Aurora Balloni
- Department of Excellence-Biomedical Sciences and Public Health, Section of Legal Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Anastasio Tini
- Department of Excellence-Biomedical Sciences and Public Health, Section of Legal Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Emilia Prospero
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine, and Public Health, Università Politecnica delle Marche, Ancona, Italy
- School of Nursing Science, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Paolo Busardò
- Department of Excellence-Biomedical Sciences and Public Health, Section of Legal Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Marilyn Ann Huestis
- Institute of Emerging Health Professions, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alfredo Fabrizio Lo Faro
- Department of Excellence-Biomedical Sciences and Public Health, Section of Legal Medicine, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
49
|
Iqbal H, Ilyas K, Akash MSH, Rehman K, Hussain A, Iqbal J. Real-time fluorescent monitoring of phase I xenobiotic-metabolizing enzymes. RSC Adv 2024; 14:8837-8870. [PMID: 38495994 PMCID: PMC10941266 DOI: 10.1039/d4ra00127c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024] Open
Abstract
This article explores the intricate landscape of advanced fluorescent probes crafted for the detection and real-time monitoring of phase I xenobiotic-metabolizing enzymes. Employing state-of-the-art technologies, such as fluorescence resonance energy transfer, intramolecular charge transfer, and solid-state luminescence enhancement, this article unfolds a multifaceted approach to unraveling the dynamics of enzymatic processes within living systems. This encompassing study involves the development and application of a diverse range of fluorescent probes, each intricately designed with tailored mechanisms to heighten sensitivity, providing dynamic insights into phase I xenobiotic-metabolizing enzymes. Understanding the role of phase I xenobiotic-metabolizing enzymes in these pathophysiological processes, is essential for both medical research and clinical practice. This knowledge can guide the development of approaches to prevent, diagnose, and treat a broad spectrum of diseases and conditions. This adaptability underscores their potential clinical applications in cancer diagnosis and personalized medicine. Noteworthy are the trifunctional fluorogenic probes, uniquely designed not only for fluorescence-based cellular imaging but also for the isolation of cellular glycosidases. This innovative feature opens novel avenues for comprehensive studies in enzyme biology, paving the way for potential therapeutic interventions. The research accentuates the selectivity and specificity of the probes, showcasing their proficiency in distinguishing various enzymes and their isoforms. The sophisticated design and successful deployment of these fluorescent probes mark significant advancements in enzymology, providing powerful tools for both researchers and clinicians. Beyond their immediate applications, these probes offer illuminating insights into disease mechanisms, facilitating early detection, and catalyzing the development of targeted therapeutic interventions. This work represents a substantial leap forward in the field, promising transformative implications for understanding and addressing complex biological processes. In essence, this research heralds a new era in the development of fluorescent probes, presenting a comprehensive and innovative approach that not only expands the understanding of cellular enzyme activities but also holds great promise for practical applications in clinical settings and therapeutic endeavors.
Collapse
Affiliation(s)
- Hajra Iqbal
- Department of Pharmaceutical Chemistry, Government College University Faisalabad Pakistan
| | - Kainat Ilyas
- Department of Pharmaceutical Chemistry, Government College University Faisalabad Pakistan
| | | | - Kanwal Rehman
- Department of Pharmacy, The Women University Multan Pakistan
| | - Amjad Hussain
- Institute of Chemistry, University of Okara Okara Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus Abbottabad 22044 Pakistan
| |
Collapse
|
50
|
Xiang Z, Guan H, Zhao X, Xie Q, Xie Z, Cai F, Dang R, Li M, Wang C. Dietary gallic acid as an antioxidant: A review of its food industry applications, health benefits, bioavailability, nano-delivery systems, and drug interactions. Food Res Int 2024; 180:114068. [PMID: 38395544 DOI: 10.1016/j.foodres.2024.114068] [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: 10/12/2023] [Revised: 01/12/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024]
Abstract
Gallic acid (GA), a dietary phenolic acid with potent antioxidant activity, is widely distributed in edible plants. GA has been applied in the food industry as an antimicrobial agent, food fresh-keeping agent, oil stabilizer, active food wrap material, and food processing stabilizer. GA is a potential dietary supplement due to its health benefits on various functional disorders associated with oxidative stress, including renal, neurological, hepatic, pulmonary, reproductive, and cardiovascular diseases. GA is rapidly absorbed and metabolized after oral administration, resulting in low bioavailability, which is susceptible to various factors, such as intestinal microbiota, transporters, and metabolism of galloyl derivatives. GA exhibits a tendency to distribute primarily to the kidney, liver, heart, and brain. A total of 37 metabolites of GA has been identified, and decarboxylation and dihydroxylation in phase I metabolism and sulfation, glucuronidation, and methylation in phase Ⅱ metabolism are considered the main in vivo biotransformation pathways of GA. Different types of nanocarriers, such as polymeric nanoparticles, dendrimers, and nanodots, have been successfully developed to enhance the health-promoting function of GA by increasing bioavailability. GA may induce drug interactions with conventional drugs, such as hydroxyurea, linagliptin, and diltiazem, due to its inhibitory effects on metabolic enzymes, including cytochrome P450 3A4 and 2D6, and transporters, including P-glycoprotein, breast cancer resistance protein, and organic anion-transporting polypeptide 1B3. In conclusion, in-depth studies of GA on food industry applications, health benefits, bioavailability, nano-delivery systems, and drug interactions have laid the foundation for its comprehensive application as a food additive and dietary supplement.
Collapse
Affiliation(s)
- Zedong Xiang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Huida Guan
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Xiang Zhao
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Qi Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Zhejun Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Fujie Cai
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Rui Dang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China
| | - Manlin Li
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China.
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, 201203, China.
| |
Collapse
|