1
|
Huang W, Tang P, Mei J, Zhang Z, Lu G. Identification of molecular subtypes and a prognostic signature based on machine learning and purine metabolism-related genes in breast cancer. Medicine (Baltimore) 2025; 104:e42288. [PMID: 40419914 DOI: 10.1097/md.0000000000042288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
Breast cancer (BC), one of the most prevalent malignant tumors worldwide, lacks efficacious diagnostic biomarkers and therapeutic targets. This study harnesses multi-omics data to identify novel purine metabolism-related genes (PMRG) as potential biomarkers and risk signatures. Univariate Cox regression was employed to assess the correlation between PMRGs and BC patient prognosis, while a Lasso Cox model was constructed to derive a prognostic signature. Gene set enrichment analysis was utilized to investigate functional differences. Kaplan-Meier survival curves were plotted to examine overall survival disparities between these 2 risk groups, with further exploration into the relationship between the prognostic signature, immune landscape, and drug sensitivity. Ultimately, a predictive nomogram was developed based on these findings. BC patients were stratified into 2 distinct molecular subtypes with significantly different prognoses using the identified PMRG-based signature, which comprised 17 PMRGs. This signature emerged as an independent prognosticator for BC and was integrated into a nomogram along with age, chemotherapy/radiotherapy treatment history, and clinical staging to accurately predict patient outcomes. Moreover, the signature showed associations with the tumor immune microenvironment and drug responsiveness, where lower-risk patients exhibited increased chemotherapeutic sensitivity, immune scores, and decreased tumor purity. Gene set enrichment analysis highlighted significant activation in pathways such as the complement and coagulation cascades, ribosome biogenesis, MAPK signaling, cAMP signaling, and drug metabolism pathways in the low-risk group. The derived PMRG-based signature holds promise for predicting the prognosis of BC patients and guiding their clinical management, including immunotherapy interventions.
Collapse
Affiliation(s)
- Wei Huang
- Department of Surgery, Ninghai Maternal and Child Health Hospital, Ningbo, Zhejiang, China
| | | | | | | | | |
Collapse
|
2
|
Chiodi D, Ishihara Y. Tertiary Alcohol: Reaping the Benefits but Minimizing the Drawbacks of Hydroxy Groups in Drug Discovery. J Med Chem 2025; 68:7889-7913. [PMID: 40231785 DOI: 10.1021/acs.jmedchem.4c03078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Among the smaller substituents in the medicinal chemist's toolbox, the hydroxy (OH) group can bestow one of the largest impacts in the drug-like properties of a molecule. A previous study showed that an H-to-OH structural modification effectively decreases lipophilicity, increases solubility, and decreases hERG inhibition. Despite these benefits, an OH group is not always recommended in drug molecules because it presents a metabolic "soft spot" for oxidation and glucuronidation in primary and secondary alcohols. Furthermore, the OH group presents challenges in permeability. In contrast, tertiary alcohols (3° ROH) often display an improved metabolic profile because oxidation at the 3° ROH is not possible, and the geminal alkyl groups could sterically shield the OH group from glucuronidation and permeability challenges. Through a series of matched molecular pairs, this Perspective highlights the 3° ROH as a motif that can reap the benefits but minimize the drawbacks of hydroxy groups in drug discovery.
Collapse
Affiliation(s)
- Debora Chiodi
- Department of Chemistry, Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Yoshihiro Ishihara
- Department of Chemistry, Genesis Therapeutics, 11568 Sorrento Valley Road Suite 8, San Diego, California 92121, United States
| |
Collapse
|
3
|
Zhou Y, Aliagas I, Wang S, Li CS, Liu Z, Bowman CM, Burdick DJ, Clark KR, Dening TJ, Flygare J, Ganti A, Girgis HS, Hanan EJ, Harris SF, Hu C, Kapadia SB, Koehler MFT, Lai T, Liang J, Liu X, Ma F, Mao J, Nicolai J, Sims J, Unhayaker S, Wai J, Wang X, Wu P, Xu Y, Yen CW, Zhang R, Elfert TF, Tan MW, Kofoed EM, Crawford TD. Discovery of potent dihydro-oxazinoquinolinone inhibitors of GuaB for the treatment of tuberculosis. Bioorg Med Chem Lett 2025; 117:130026. [PMID: 39536836 DOI: 10.1016/j.bmcl.2024.130026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Tuberculosis is the leading cause of death from an infectious disease, and is caused by Mycobacterium tuberculosis (M.tb). More than 1 billion people worldwide are thought to harbor an M.tb infection. The multidrug therapy that represents the current standard of care requires a minimum of four months of dosing and drug resistant Mycobacterium tuberculosis treatment regimens are significantly longer. Inosine-5'-monophosphate dehydrogenase (GuaB) is the enzyme that performs the rate-limiting step in de novo guanine nucleotide biosynthesis that is critical for growth and viability of bacteria including M.tb. The development of a novel antibiotic that inhibits GuaB could combine with existing therapies in novel ways and thereby contribute to effective therapeutic regimens for the treatment of tuberculosis. Here we describe the discovery of structurally distinct small molecule GuaB inhibitors that are potent against M.tb H37Ra and H37Rv strains and have desirable safety and ADME profiles.
Collapse
Affiliation(s)
- Yuebiao Zhou
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| | - Ignacio Aliagas
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Shumei Wang
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Chun Sing Li
- Wuxi Apptec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | - Zhiguo Liu
- Wuxi Apptec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | | | - Daniel J Burdick
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Kevin R Clark
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Tahnee J Dening
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - John Flygare
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Anjani Ganti
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Hany S Girgis
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Emily J Hanan
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Seth F Harris
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Chloe Hu
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | | | | | - Tommy Lai
- Wuxi Apptec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | - Jun Liang
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Xingrong Liu
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Fang Ma
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Jialin Mao
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Jeremy Nicolai
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Jessica Sims
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Savita Unhayaker
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - John Wai
- Wuxi Apptec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | - Xiaojing Wang
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Ping Wu
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Yiming Xu
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Chun-Wan Yen
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Renwei Zhang
- Wuxi Apptec Co., Ltd., 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | - Torben F Elfert
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Man-Wah Tan
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Eric M Kofoed
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Terry D Crawford
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, United States.
| |
Collapse
|
4
|
Olafuyi O, Michelet R, Garle M, Allegaert K. Exploring the Impact of Developmental Clearance Saturation on Propylene Glycol Exposure in Adults and Term Neonates Using Physiologically Based Pharmacokinetic Modeling. J Clin Pharmacol 2025; 65:272-284. [PMID: 39404076 PMCID: PMC11867916 DOI: 10.1002/jcph.6150] [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: 08/08/2024] [Accepted: 09/25/2024] [Indexed: 03/01/2025]
Abstract
Propylene glycol (PG) is a pharmaceutical excipient which is generally regarded as safe (GRAS), though clinical toxicity has been reported. PG toxicity has been attributed to accumulation due to saturation of the alcohol dehydrogenase (ADH)-mediated clearance pathway. This study aims to explore the impact of the saturation of ADH-mediated PG metabolism on its developmental clearance in adults and neonates and assess the impact of a range of doses on PG clearance saturation and toxicity. Physiologically based pharmacokinetic (PBPK) models for PG in adults and term neonates were developed using maximum velocity (Vmax) and Michaelis-Menten's constant (Km) of ADH-mediated metabolism determined in vitro in human liver cytosol, published physicochemical, drug-related and ADH ontogeny parameters. The models were validated and used to determine the impact of dosing regimen on PG clearance saturation and toxicity in adults and neonates. The Vmax and Km of PG in human liver cytosol were 1.57 nmol/min/mg protein and 25.1 mM, respectively. The PG PBPK model adequately described PG PK profiles in adults and neonates. The PG dosing regimens associated with saturation and toxicity were dependent on both dose amount and cumulative in standard dosing frequencies. Doses resulting in saturation were higher than those associated with clinically observed toxicity. In individuals without impaired clearance or when PG exposure is through formulations that contain excipients with possible interaction with PG, a total daily dose of 100-200 mg/kg/day in adults and 25-50 mg/kg/day in neonates is unlikely to result in toxic PG levels or PG clearance saturation.
Collapse
Affiliation(s)
- Olusola Olafuyi
- Division of Physiology, Pharmacology and Neuroscience, School of Life SciencesUniversity of NottinghamNottinghamUK
| | - Robin Michelet
- Department of Clinical Pharmacy and BiochemistryInstitute of PharmacyFreie Universität BerlinBerlinGermany
- qharmetra LLCBerlinGermany
| | - Michael Garle
- Division of Physiology, Pharmacology and Neuroscience, School of Life SciencesUniversity of NottinghamNottinghamUK
| | - Karel Allegaert
- Department of Development and RegenerationKU LeuvenLeuvenBelgium
- Department of Pharmaceutical and Pharmacological SciencesKU LeuvenLeuvenBelgium
- Department of Hospital PharmacyErasmus Medical CenterRotterdamThe Netherlands
| |
Collapse
|
5
|
McGinnis CD, Harris PS, Graham BIM, Marentette JO, Michel CR, Saba LM, Reisdorph R, Roede JR, Fritz KS. Acetylation of proximal cysteine-lysine pairs by alcohol metabolism. Redox Biol 2025; 79:103462. [PMID: 39729908 PMCID: PMC11732177 DOI: 10.1016/j.redox.2024.103462] [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: 09/27/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/29/2024] Open
Abstract
Alcohol consumption induces hepatocyte damage through complex processes involving oxidative stress and disrupted metabolism. These factors alter proteomic and epigenetic marks, including alcohol-induced protein acetylation, which is a key post-translational modification (PTM) that regulates hepatic metabolism and is associated with the pathogenesis of alcohol-associated liver disease (ALD). Recent evidence suggests lysine acetylation occurs when a proximal cysteine residue is within ∼15 Å of a lysine residue, referred to as a cysteine-lysine (Cys-Lys) pair. Here, acetylation can occur through the transfer of an acetyl moiety via an S → N transfer reaction. Alcohol-mediated redox stress is known to occur coincidentally with lysine acetylation, yet the biochemical mechanisms related to cysteine and lysine crosstalk within ALD remain unexplored. A murine model of ALD was employed to quantify hepatic cysteine redox changes and lysine acetylation, revealing that alcohol metabolism significantly reduced the cysteine thiol proteome and increased protein acetylation. Interrogating both cysteine redox and lysine acetylation datasets, 1280 protein structures generated by AlphaFold2 represented by a 3D spatial matrix were used to quantify the distances between 557,815 cysteine and lysine residues. Our analysis revealed that alcohol metabolism induces redox changes and acetylation selectively on proximal Cys-Lys pairs with an odds ratio of 1.88 (p < 0.0001). Key Cys-Lys redox signaling hubs were impacted in metabolic pathways associated with ALD, including lipid metabolism and the electron transport chain. Proximal Cys-Lys pairs exist as sets with four major motifs represented by the number of Cys and Lys residues that are pairing (Cys1:Lys1, Cysx:Lys1, Cys1:Lysx and Cysx:Lysx) each with a unique microenvironment. The motifs are composed of functionally relevant Cys-Ly altered within ALD, identifying potential therapeutic targets. Furthermore, these unique Cys-Lys redox signatures are translationally relevant as revealed by orthologous comparison with severe alcohol-associated hepatitis (SAH) explants, revealing numerous pathogenic thiol redox signals in these patients.
Collapse
Affiliation(s)
- Courtney D McGinnis
- Graduate Program in Toxicology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Peter S Harris
- Graduate Program in Toxicology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brenton I M Graham
- Graduate Program in Toxicology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - John O Marentette
- Graduate Program in Toxicology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Cole R Michel
- Graduate Program in Toxicology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Laura M Saba
- Graduate Program in Toxicology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Richard Reisdorph
- Graduate Program in Toxicology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - James R Roede
- Graduate Program in Toxicology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristofer S Fritz
- Graduate Program in Toxicology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
6
|
Schröder S, Massarou C, Pfister T, Bleich S, Proskynitopoulos PJ, Heck J, Schulze Westhoff M, Glahn A. Drug interactions in patients with alcohol use disorder: results from a real-world study on an addiction-specific ward. Ther Adv Drug Saf 2025; 16:20420986241311214. [PMID: 39830585 PMCID: PMC11742168 DOI: 10.1177/20420986241311214] [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: 05/07/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
Background The majority of patients with alcohol use disorder (AUD) regularly take medication. Alcohol interacts negatively with many commonly prescribed drugs. However, little is known about the characteristics and frequency of potential alcohol-medication and drug-drug interactions in patients with AUD. Objectives This study aimed to determine the prevalence and characteristics of drug interactions in patients with AUD during withdrawal therapy on an addiction-specific ward. Design Retrospective cohort study. Methods Medication charts were analyzed and screened for potential alcohol-medication and drug-drug interactions. For the screening of potential alcohol-medication interactions, the drugs.com classification was utilized and potential drug-drug interactions were identified using the mediQ electronic interaction program. Results In our study, almost two-thirds (66.3%; 1089/1643) of all patient cases were prescribed at least one drug that could potentially interact with alcohol. Four percent of all alcohol-medication interactions were classified as severe, 91.8% as moderate, and 4.3% as mild. Drug classes commonly involved in serious interactions with alcohol were analgesics and drugs used in diabetes. A total of 811 potential drug-drug interactions were identified, of which 3.3% were classified as severe and 96.5% as moderate. Psychoanaleptics (ATC N06) and psycholeptics (ATC N05) were most frequently associated with moderate to severe interactions. Conclusion Potential alcohol-medication and drug-drug interactions are common in hospitalized patients with AUD. Improvements in the quality of prescribing should focus on the use of psychotropic drugs.
Collapse
Affiliation(s)
- Sebastian Schröder
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Christina Massarou
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Tabea Pfister
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Stefan Bleich
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | | | - Johannes Heck
- Institute for Clinical Pharmacology, Hannover Medical School, Hannover, Germany
| | - Martin Schulze Westhoff
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Alexander Glahn
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| |
Collapse
|
7
|
Sato R, Fukami T, Shimomura K, Zhang Y, Nakano M, Nakajima M. Characterization of human alcohol dehydrogenase 4 and aldehyde dehydrogenase 2 as enzymes involved in the formation of 5-carboxylpirfenidone, a major metabolite of pirfenidone. Drug Metab Dispos 2025; 53:100010. [PMID: 39884816 DOI: 10.1124/dmd.124.001917] [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: 08/27/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 01/22/2025] Open
Abstract
Pirfenidone (PIR) is used in the treatment of idiopathic pulmonary fibrosis. After oral administration, it is metabolized by cytochrome P450 1A2 to 5-hydroxylpirfenidone (5-OH PIR) and further oxidized to 5-carboxylpirfenidone (5-COOH PIR), a major metabolite excreted in the urine (90% of the dose). This study aimed to identify enzymes that catalyze the formation of 5-COOH PIR from 5-OH PIR in the human liver. 5-COOH PIR was formed from 5-OH PIR in the presence of NAD+ by human liver microsomes (HLMs) more than by human liver cytosol (HLC), with the concomitant formation of the aldehyde form (5-CHO PIR) as an intermediate metabolite. By purifying enzymes from HLMs, alcohol dehydrogenases (ADHs) were identified as candidate enzymes catalyzing 5-CHO PIR formation, although ADHs are localized in the cytoplasm. Among constructed recombinant ADH1-5 expressed in HEK293T cells, only ADH4 efficiently catalyzed 5-CHO PIR formation from 5-OH PIR with a Km value (29.0 ± 4.9 μM), which was close to that by HLMs (59.1 ± 4.6 μM). In contrast to commercially available HLC, HLC prepared in-house clearly showed substantial 5-CHO PIR formation, and ADH4 protein levels were significantly (rs = 0.772, P < .0001) correlated with 5-CHO PIR formation in 25 HLC samples prepared in-house. Some components of the commercially available HLC may inhibit ADH4 activity. Disulfiram, an inhibitor of aldehyde dehydrogenases (ALDH), decreased 5-COOH PIR formation and increased 5-CHO PIR formation from 5-OH PIR in HLMs. ALDH2 knockdown in HepG2 cells by siRNA decreased 5-COOH PIR formation by 61%. SIGNIFICANCE STATEMENT: This study clarified that 5-carboxylpirfenidone formation from 5-hydroxylpirfenidone proceeds via a 2-step oxidation reaction catalyzed by ADH4 and disulfiram-sensitive enzymes, including ALDH2. Interindividual differences in the expression levels or functions of these enzymes could cause variations in the pharmacokinetics of pirfenidone.
Collapse
Affiliation(s)
- Rei Sato
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa, Japan.
| | - Kazuya Shimomura
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan
| | - Yongjie Zhang
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa, Japan; Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Masataka Nakano
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa, Japan
| |
Collapse
|
8
|
V U P, T I M, K K M. An integrative analysis to identify pancancer epigenetic biomarkers. Comput Biol Chem 2024; 113:108260. [PMID: 39467487 DOI: 10.1016/j.compbiolchem.2024.108260] [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: 06/25/2024] [Revised: 09/13/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024]
Abstract
Integrating and analyzing the pancancer data collected from different experiments is crucial for gaining insights into the common mechanisms in the molecular level underlying the development and progression of cancers. Epigenetic study of the pancancer data can provide promising results in biomarker discovery. The genes that are epigenetically dysregulated in different cancers are powerful biomarkers for drug-related studies. This paper identifies the genes having altered expression due to aberrant methylation patterns using differential analysis of TCGA pancancer data of 12 different cancers. We identified a comprehensive set of 115 epigenetic biomarker genes out of which 106 genes having pancancer properties. The correlation analysis, gene set enrichment, protein-protein interaction analysis, pancancer characteristics analysis, and diagnostic modeling were performed on these biomarkers to illustrate the power of this signature and found to be important in different molecular operations related to cancer. An accuracy of 97.56% was obtained on TCGA pancancer gene expression dataset for predicting the binary class tumor or normal. The source code and dataset of this work are available at https://github.com/panchamisuneeth/EpiPanCan.git.
Collapse
Affiliation(s)
- Panchami V U
- Adi Shankara Institute of Engineering and Technology, Ernakulam, 683574, Kerala, India; Government Engineering College Thrissur, 680009, Kerala, India; APJ Abdul Kalam Technological University, 695016, Kerala, India.
| | - Manish T I
- SCMS School of Engineering and Technology, Ernakulam, 683576, Kerala, India; APJ Abdul Kalam Technological University, 695016, Kerala, India
| | - Manesh K K
- Government Engineering College Thrissur, 680009, Kerala, India; APJ Abdul Kalam Technological University, 695016, Kerala, India
| |
Collapse
|
9
|
Ji K, Zhang Y, Zhang T, Li D, Yuan Y, Wang L, Huang Q, Chen W. sll1019 and slr1259 encoding glyoxalase II improve tolerance of Synechocystis sp. PCC 6803 to methylglyoxal- and ethanol- induced oxidative stress by glyoxalase pathway. Appl Environ Microbiol 2024; 90:e0056424. [PMID: 39431850 PMCID: PMC11577758 DOI: 10.1128/aem.00564-24] [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: 04/02/2024] [Accepted: 09/20/2024] [Indexed: 10/22/2024] Open
Abstract
The glyoxalase pathway is the primary detoxification mechanism for methylglyoxal (MG), a ubiquitous toxic metabolite that disrupts redox homeostasis. In the glyoxalase pathway, glyoxalase II (GlyII) can completely detoxify MG. Increasing the activity of the glyoxalase system can enhance the resistance of plants or organisms to abiotic stress, but the relevant mechanism remains largely unknown. In this study, we investigated the physiological functions of GlyII genes (sll1019 and slr1259) in Synechocystis sp. PCC 6803 under MG or ethanol stress based on transcriptome and metabolome data. High-performance liquid chromatography (HPLC) results showed that proteins Sll1019 and Slr1259 had GlyII activity. Under stress conditions, sll1019 and slr1259 protected the strain against oxidative stress by enhancing the activity of the glyoxalase pathway and raising the contents of antioxidants such as glutathione and superoxide dismutase. In the photosynthetic system, sll1019 and slr1259 indirectly affected the light energy absorption by strains, synthesis of photosynthetic pigments, and activities of photosystem I and photosystem II, which was crucial for the growth of the strain under stress conditions. In addition, sll1019 and slr1259 enhanced the tolerance of strain to oxidative stress by indirectly regulating metabolic networks, including ensuring energy acquisition, NADH and NADPH production, and phosphate and nitrate transport. This study reveals the mechanism by which sll1019 and slr1259 improve oxidative stress tolerance of strains by glyoxalase pathway. Our findings provide theoretical basis for breeding, seedling, and field production of abiotic stress tolerance-enhanced variety.IMPORTANCEThe glyoxalase system is present in most organisms, and it is the primary pathway for eliminating the toxic metabolite methylglyoxal. Increasing the activity of the glyoxalase system can enhance plant resistance to environmental stress, but the relevant mechanism is poorly understood. This study revealed the physiological functions of glyoxalase II genes sll1019 and slr1259 in Synechocystis sp. PCC 6803 under abiotic stress conditions and their regulatory effects on oxidative stress tolerance of strains. Under stress conditions, sll1019 and slr1259 enhanced the activity of the glyoxalase pathway and the antioxidant system, maintained photosynthesis, ensured energy acquisition, NADH and NADPH production, and phosphate and nitrate transport, thereby protecting the strain against oxidative stress. This study lays a foundation for further deciphering the mechanism by which the glyoxalase system enhances the tolerance of cells to abiotic stress, providing important information for breeding, seedling, and selection of plants with strong stress resistance.
Collapse
Affiliation(s)
- Kai Ji
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yihang Zhang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Tianyuan Zhang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Daixi Li
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yuan Yuan
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Li Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qiaoyun Huang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China
- Hubei Key Laboratory of Soil Environment and Pollution Remediation, College of Resources and Environment, Huazhong Agricultural University, Wuhan, China
| | - Wenli Chen
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
10
|
Park S, Jebamani P, Seo YG, Wu S. Computational Study of Network and Type-I Functional Divergence in Alcohol Dehydrogenase Enzymes Across Species Using Molecular Dynamics Simulation. Biomolecules 2024; 14:1473. [PMID: 39595650 PMCID: PMC11591965 DOI: 10.3390/biom14111473] [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: 10/01/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Alcohol dehydrogenases (ADHs) are critical enzymes involved in the oxidation of alcohols, contributing to various metabolic pathways across organisms. This study investigates type I functional divergence within three ADH1 families: Saccharomyces cerevisiae (PDB ID: 4W6Z), Gadus morhua (PDB ID: 1CDO), and Homo sapiens (PDB ID: 1HDX). Understanding the molecular evolution and mechanisms underlying functional divergence of ADHs is essential for comprehending their adaptive significance. For this purpose, we performed a computational analysis that included structural characterization of ADHs through three-dimensional modeling, site-specific analysis to evaluate selective pressures and evolutionary constraints, and network analysis to elucidate relationships between structural features and functional divergence. Our findings indicate substantial variations in evolutionary and structural adaptations among the ADH families.
Collapse
Affiliation(s)
- Suhyun Park
- Department of Physics, Pukyong National University, Busan 48513, Republic of Korea; (S.P.); (Y.G.S.)
| | - Petrina Jebamani
- Department of Chemical Engineering, Pusan National University, Busan 46241, Republic of Korea;
| | - Yeon Gyo Seo
- Department of Physics, Pukyong National University, Busan 48513, Republic of Korea; (S.P.); (Y.G.S.)
| | - Sangwook Wu
- Department of Physics, Pukyong National University, Busan 48513, Republic of Korea; (S.P.); (Y.G.S.)
- PharmCADD, 1102-ho, 60, Centum Jungang-ro, Haeundae-gu, Busan 48059, Republic of Korea
| |
Collapse
|
11
|
Thomas LA, Hopkinson RJ. The biochemistry of the carcinogenic alcohol metabolite acetaldehyde. DNA Repair (Amst) 2024; 144:103782. [PMID: 39566398 DOI: 10.1016/j.dnarep.2024.103782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/14/2024] [Accepted: 10/30/2024] [Indexed: 11/22/2024]
Abstract
Acetaldehyde (AcH) is the first metabolite of ethanol and is proposed to be responsible for the genotoxic effects of alcohol consumption. As an electrophilic aldehyde, AcH can form multiple adducts with DNA and other biomolecules, leading to function-altering and potentially toxic and carcinogenic effects. In this review, we describe sources of AcH in humans, including AcH biosynthesis mechanisms, and outline the structures, properties and functions of AcH-derived adducts with biomolecules. We also describe human AcH detoxification mechanisms and discuss ongoing challenges in the field.
Collapse
Affiliation(s)
- Liam A Thomas
- Institute of Structural and Chemical Biology, School of Chemistry, University of Leicester, Leicester LE1 7RH, UK
| | - Richard J Hopkinson
- Institute of Structural and Chemical Biology, School of Chemistry, University of Leicester, Leicester LE1 7RH, UK.
| |
Collapse
|
12
|
Luan L, Zhang Y, Ji X, Guo B, Song S, Huang Y, Zhang S. Electro-Driven Multi-Enzymatic Cascade Conversion of CO 2 to Ethylene Glycol in Nano-Reactor. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407204. [PMID: 39231322 PMCID: PMC11538636 DOI: 10.1002/advs.202407204] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/13/2024] [Indexed: 09/06/2024]
Abstract
Multi-enzymatic cascade reaction provides a new avenue for C─C coupling directly from CO2 under mild conditions. In this study, a new pathway with four enzymes including formate dehydrogenase (PaFDH), formaldehyde dehydrogenase (BmFADH), glycolaldehyde synthase (PpGALS), and alcohol dehydrogenase (GoADH) is developed for directly converting CO2 gas molecules to ethylene glycol (EG) in vitro. A rhodium-based NADH regeneration electrode is constructed to continuously provide the proton and electron of this multi-enzymatic cascade reaction. The prepared electrode can reach the Faradaic Efficiency (FE) of 82.9% at -0.6 V (vs. Ag/AgCl) and the NADH productivity of 0.737 mM h-1. Shortening the reaction path is crucial for multi-enzymatic cascade reactions. Here, a hydrogen-bonded organic framework (HOF) nano-reactor is successfully developed to immobilize four enzymes in one pot with a striking enzyme loading capacity (990 mg enzyme g-1 material). Through integrating and optimization of NADH electro-regeneration and enzymatic catalysis in one pot, 0.15 mM EG is achieved with an average conversion rate of 7.15 × 10-7 mmol CO2 min-1 mg-1 enzymes in 6 h. These results shed light on electro-driven multi-enzymatic cascade conversion of C─C coupling from CO2 in the nano-reactor.
Collapse
Affiliation(s)
- Likun Luan
- Beijing Key Laboratory of Ionic Liquids Clean ProcessCAS Key Laboratory of Green Process and EngineeringState Key Laboratory of Mesoscience and EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
- Sino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijing101408P. R. China
| | - Yingfang Zhang
- Beijing Key Laboratory of Ionic Liquids Clean ProcessCAS Key Laboratory of Green Process and EngineeringState Key Laboratory of Mesoscience and EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
| | - Xiuling Ji
- Beijing Key Laboratory of Ionic Liquids Clean ProcessCAS Key Laboratory of Green Process and EngineeringState Key Laboratory of Mesoscience and EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
| | - Boxia Guo
- Beijing Key Laboratory of Ionic Liquids Clean ProcessCAS Key Laboratory of Green Process and EngineeringState Key Laboratory of Mesoscience and EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
- Sino‐Danish CollegeUniversity of Chinese Academy of SciencesBeijing101408P. R. China
| | - Shaoyu Song
- Beijing Key Laboratory of Ionic Liquids Clean ProcessCAS Key Laboratory of Green Process and EngineeringState Key Laboratory of Mesoscience and EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
| | - Yuhong Huang
- Beijing Key Laboratory of Ionic Liquids Clean ProcessCAS Key Laboratory of Green Process and EngineeringState Key Laboratory of Mesoscience and EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
| | - Suojiang Zhang
- Beijing Key Laboratory of Ionic Liquids Clean ProcessCAS Key Laboratory of Green Process and EngineeringState Key Laboratory of Mesoscience and EngineeringInstitute of Process EngineeringChinese Academy of SciencesBeijing100190P. R. China
- Longzihu New Energy LaboratoryZhengzhou Institute of Emerging Industrial TechnologyHenan UniversityZhengzhou450000P. R. China
| |
Collapse
|
13
|
Pei W, Zhang X, Zeng Y, Li J, Li Z, Yang J, Du X, Zhu Y, Sun Y. Rational design of a zinc-dependence secondary alcohol dehydrogenase to boost its oxidation activity for α-hydroxyketones biosynthesis. Int J Biol Macromol 2024; 282:137183. [PMID: 39488308 DOI: 10.1016/j.ijbiomac.2024.137183] [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: 09/25/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/04/2024]
Abstract
α-Hydroxyketones, which have significant industrial applications, can be sustainably synthesized through the oxidation of secondary alcohols using secondary alcohol dehydrogenase (SADH). However, the activity of the SADH oxidation reaction is generally low, making it unsuitable for large-scale production. In this study, a rational design approach was employed to computationally engineer SADH derived from Ogataea parapolymorpha (OpSADH), significantly enhancing its oxidation activity towards (R)-1,2-propanediol (PDO). The mutant M2 (S222T/S316A) exhibited a 14.2-fold increase in specific enzyme activity compared to the wild type (WT) and was employed as a catalyst for high-concentration hydroxyacetone production, facilitated by an NAD+ regeneration system. By applying an appropriate Zn2+ ion force field in molecular dynamics (MD) simulations, it was found that two mutation sites could stabilize the conformations of NAD+ and PDO, thereby revealing the molecular mechanism behind the enhanced activity of this metalloenzyme mutant. Notably, we first uncovered the dynamic mechanism by which four key residues (Cys39, His75, Glu76, and Glu175) and PDO within the active pocket contribute to the formation of coordination bonds with Zn2+. The findings of this study provide robust support for researching the catalytic mechanisms and dynamics processes of metalloenzymes.
Collapse
Affiliation(s)
- Wenwen Pei
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China; National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
| | - Xuewen Zhang
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
| | - Yan Zeng
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
| | - Jiao Li
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
| | - Ziyi Li
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
| | - Jiangang Yang
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
| | - Xinjun Du
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China.
| | - Yueming Zhu
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China.
| | - Yuanxia Sun
- National Engineering Laboratory for Industrial Enzymes, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
| |
Collapse
|
14
|
Schröder S, Schulze Westhoff M, Bleich S, Bode H, Jendretzky KF, Krichevsky B, Glahn A, Heck J. Influence of inpatient withdrawal treatment on drug safety in alcohol use disorder - a quasi-experimental pre-post study. BMC Psychiatry 2024; 24:733. [PMID: 39455970 PMCID: PMC11515795 DOI: 10.1186/s12888-024-06188-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
OBJECTIVE Most patients with alcohol use disorder (AUD) regularly take medication. Alcohol interacts negatively with many commonly prescribed medications. Little is known about whether the risk of potential alcohol-medication and drug-drug interactions increases or decreases in patients with AUD during inpatient withdrawal treatment. The aim of our study was to determine the prevalence and characteristics of potential alcohol-medication and drug-drug interactions in patients with AUD before and after withdrawal treatment in an addiction unit. DESIGN Prospective monocentric quasi-experimental pre-post study. METHODS Medication records before and after withdrawal treatment were analyzed and screened for potential alcohol-medication (pAMI) and drug-drug interactions (pDDI) using the drugs.com classification and the AiDKlinik® electronic interaction program, respectively. RESULTS We enrolled 153 patients with AUD who were treated in an addiction unit of a university hospital in Germany. Of these, 67.3% experienced at least one pAMI before and 91.5% after withdrawal treatment. In total, there were 278 pAMIs classified as "mild," "moderate," or "severe" before and 370 pAMIs after withdrawal treatment. Additionally, there were 76 pDDIs classified as "moderate," "severe," or "contraindicated combinations" both before and after withdrawal treatment. CONCLUSION The risk of exposure to pAMIs and pDDIs increases during inpatient withdrawal treatment in patients with AUD. Improvements in the quality of prescribing should particularly focus on the use of antihypertensives and opioids.
Collapse
Affiliation(s)
- Sebastian Schröder
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Martin Schulze Westhoff
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Stefan Bleich
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Henry Bode
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | | | - Benjamin Krichevsky
- Institute for General Practice and Palliative Care, Hannover Medical School, Hannover, Germany
| | - Alexander Glahn
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Johannes Heck
- Institute for Clinical Pharmacology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
15
|
Liu Y, Yu J, An X, Rao H, Qiu Z, Ke J, Wu L, Zhu Z, Deng H, Wu F, Zhang Z, Li S. TSA attenuates the progression of c-Myc-driven hepatocarcinogenesis by pAKT-ADH4 pathway. BMC Cancer 2024; 24:1049. [PMID: 39187747 PMCID: PMC11346213 DOI: 10.1186/s12885-024-12781-x] [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: 09/23/2023] [Accepted: 08/08/2024] [Indexed: 08/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the primary malignant tumor of the liver. c-Myc is one of the most common oncogenes in clinical settings, and amplified levels of c-Myc are frequently found in HCC. Histone deacetylase inhibitors (HDACi), such as Trichostatin A (TSA), hold enormous promise for the treatment of HCC. However, the potential and mechanism of TSA in the treatment of c-Myc-induced HCC are unclear. In this study, we investigated the effects of TSA treatment on a c-Myc-induced HCC model in mice. TSA treatment delayed the development of HCC, and liver function indicators such as ALT, AST, liver weight ratio, and spleen weight ratio demonstrated the effectiveness of TSA treatment. Oil red staining further demonstrated that TSA attenuated lipid accumulation in the HCC tissues of mice. Through mRNA sequencing, we identified that TSA mainly affected cell cycle and fatty acid degradation genes, with alcohol dehydrogenase 4 (ADH4) potentially being the core molecular downstream target. QPCR, immunohistochemistry, and western blot analysis revealed that ADH4 expression was repressed by c-Myc and restored after TSA treatment both in vitro and in vivo. Furthermore, we observed that the levels of total NAD+ and NADH, NAD+, NAD+/NADH, and ATP concentration increased after c-Myc transfection in liver cells but decreased after TSA intervention. The levels of phosphorylated protein kinase B (p-AKT) and p-mTOR were identified as targets regulated by TSA, and they governed the ADH4 expression and the downstream regulation of total NAD+ and NADH, NAD+, NAD+/NADH, and ATP concentration. Overall, our study suggests that TSA has a therapeutic effect on c-Myc-induced HCC through the AKT-mTOR-ADH4 pathway. These findings provide valuable insights into the potential treatment of HCC using TSA and shed light on the underlying molecular mechanisms involved.
Collapse
Grants
- JC2020003, JC202109, YC2023033, YC2024007 Innovative Research Program for Graduates of Institute of Hubei University of Medicine
- JC2020003, JC202109, YC2023033, YC2024007 Innovative Research Program for Graduates of Institute of Hubei University of Medicine
- JC2020003, JC202109, YC2023033, YC2024007 Innovative Research Program for Graduates of Institute of Hubei University of Medicine
- JC2020003, JC202109, YC2023033, YC2024007 Innovative Research Program for Graduates of Institute of Hubei University of Medicine
- 2023AFB882, 2022CFB004 the Hubei Provincial Natural Science Foundation
- 2023AFB882, 2022CFB004 the Hubei Provincial Natural Science Foundation
- S202110929011 the Provincial Training Program of Innovation and Entrepreneurship for undergraduates
- Q20212103 the Natural Science Foundation of Hubei Provincial Department of Education
- Q20212103 the Natural Science Foundation of Hubei Provincial Department of Education
- 2020QDJZR018 Cultivating Project for Young Scholar at Hubei University of Medicine
- FDFR201901 Free Exploration Project of Hubei University of Medicine
Collapse
Affiliation(s)
- Yang Liu
- Sinopharm Dongfeng General Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, People's Republic of China
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, People's Republic of China
| | - Juan Yu
- Sinopharm Dongfeng General Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, People's Republic of China
| | - Xiaotong An
- Sinopharm Dongfeng General Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, People's Republic of China
| | - Huiling Rao
- Sinopharm Dongfeng General Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, People's Republic of China
| | - Zhenpeng Qiu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, People's Republic of China
| | - Jing Ke
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, People's Republic of China
| | - Lun Wu
- Sinopharm Dongfeng General Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, People's Republic of China
| | - Zhengpeng Zhu
- Sinopharm Dongfeng General Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, People's Republic of China
| | - Haojun Deng
- Sinopharm Dongfeng General Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, People's Republic of China
| | - Fuyun Wu
- Sinopharm Dongfeng General Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, People's Republic of China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, People's Republic of China.
- Institute of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, People's Republic of China.
| | - Zhaoyang Zhang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, People's Republic of China.
- Institute of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, People's Republic of China.
| | - Shan Li
- Sinopharm Dongfeng General Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, People's Republic of China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, People's Republic of China.
- Institute of Basic Medical Science, Hubei University of Medicine, Shiyan, 442000, People's Republic of China.
| |
Collapse
|
16
|
Jennings MV, Martínez-Magaña JJ, Courchesne-Krak NS, Cupertino RB, Vilar-Ribó L, Bianchi SB, Hatoum AS, Atkinson EG, Giusti-Rodriguez P, Montalvo-Ortiz JL, Gelernter J, Artigas MS, Elson SL, Edenberg HJ, Fontanillas P, Palmer AA, Sanchez-Roige S. A phenome-wide association and Mendelian randomisation study of alcohol use variants in a diverse cohort comprising over 3 million individuals. EBioMedicine 2024; 103:105086. [PMID: 38580523 PMCID: PMC11121167 DOI: 10.1016/j.ebiom.2024.105086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Alcohol consumption is associated with numerous negative social and health outcomes. These associations may be direct consequences of drinking, or they may reflect common genetic factors that influence both alcohol consumption and other outcomes. METHODS We performed exploratory phenome-wide association studies (PheWAS) of three of the best studied protective single nucleotide polymorphisms (SNPs) in genes encoding ethanol metabolising enzymes (ADH1B: rs1229984-T, rs2066702-A; ADH1C: rs698-T) using up to 1109 health outcomes across 28 phenotypic categories (e.g., substance-use, mental health, sleep, immune, cardiovascular, metabolic) from a diverse 23andMe cohort, including European (N ≤ 2,619,939), Latin American (N ≤ 446,646) and African American (N ≤ 146,776) populations to uncover new and perhaps unexpected associations. These SNPs have been consistently implicated by both candidate gene studies and genome-wide association studies of alcohol-related behaviours but have not been investigated in detail for other relevant phenotypes in a hypothesis-free approach in such a large cohort of multiple ancestries. To provide insight into potential causal effects of alcohol consumption on the outcomes significant in the PheWAS, we performed univariable two-sample and one-sample Mendelian randomisation (MR) analyses. FINDINGS The minor allele rs1229984-T, which is protective against alcohol behaviours, showed the highest number of PheWAS associations across the three cohorts (N = 232, European; N = 29, Latin American; N = 7, African American). rs1229984-T influenced multiple domains of health. We replicated associations with alcohol-related behaviours, mental and sleep conditions, and cardio-metabolic health. We also found associations with understudied traits related to neurological (migraines, epilepsy), immune (allergies), musculoskeletal (fibromyalgia), and reproductive health (preeclampsia). MR analyses identified evidence of causal effects of alcohol consumption on liability for 35 of these outcomes in the European cohort. INTERPRETATION Our work demonstrates that polymorphisms in genes encoding alcohol metabolising enzymes affect multiple domains of health beyond alcohol-related behaviours. Understanding the underlying mechanisms of these effects could have implications for treatments and preventative medicine. FUNDING MVJ, NCK, SBB, SSR and AAP were supported by T32IR5226 and 28IR-0070. SSR was also supported by NIDA DP1DA054394. NCK and RBC were also supported by R25MH081482. ASH was supported by funds from NIAAA K01AA030083. JLMO was supported by VA 1IK2CX002095. JLMO and JJMM were also supported by NIDA R21DA050160. JJMM was also supported by the Kavli Postdoctoral Award for Academic Diversity. EGA was supported by K01MH121659 from the NIMH/NIH, the Caroline Wiess Law Fund for Research in Molecular Medicine and the ARCO Foundation Young Teacher-Investigator Fund at Baylor College of Medicine. MSA was supported by the Instituto de Salud Carlos III and co-funded by the European Union Found: Fondo Social Europeo Plus (FSE+) (P19/01224, PI22/00464 and CP22/00128).
Collapse
Affiliation(s)
- Mariela V Jennings
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - José Jaime Martínez-Magaña
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, Orange, West Haven, CT, USA
| | | | - Renata B Cupertino
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Laura Vilar-Ribó
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
| | - Sevim B Bianchi
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Alexander S Hatoum
- Department of Psychology & Brain Sciences, Washington University in St. Louis, St Louis, MO, USA
| | - Elizabeth G Atkinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Paola Giusti-Rodriguez
- Department of Psychiatry, University of Florida College of Medicine, Gainesville, FL, USA
| | - Janitza L Montalvo-Ortiz
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, Orange, West Haven, CT, USA; National Center of Posttraumatic Stress Disorder, VA CT Healthcare Center, West Haven, CT, USA
| | - Joel Gelernter
- VA CT Healthcare Center, Department Psychiatry, West Haven, CT, USA; Departments Psychiatry, Genetics, and Neuroscience, Yale Univ. School of Medicine, New Haven, CT, USA
| | - María Soler Artigas
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain; Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | | | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
17
|
Sun M, Manson ML, Guo T, de Lange ECM. CNS Viral Infections-What to Consider for Improving Drug Treatment: A Plea for Using Mathematical Modeling Approaches. CNS Drugs 2024; 38:349-373. [PMID: 38580795 PMCID: PMC11026214 DOI: 10.1007/s40263-024-01082-3] [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] [Accepted: 03/10/2024] [Indexed: 04/07/2024]
Abstract
Neurotropic viruses may cause meningitis, myelitis, encephalitis, or meningoencephalitis. These inflammatory conditions of the central nervous system (CNS) may have serious and devastating consequences if not treated adequately. In this review, we first summarize how neurotropic viruses can enter the CNS by (1) crossing the blood-brain barrier or blood-cerebrospinal fluid barrier; (2) invading the nose via the olfactory route; or (3) invading the peripheral nervous system. Neurotropic viruses may then enter the intracellular space of brain cells via endocytosis and/or membrane fusion. Antiviral drugs are currently used for different viral CNS infections, even though their use and dosing regimens within the CNS, with the exception of acyclovir, are minimally supported by clinical evidence. We therefore provide considerations to optimize drug treatment(s) for these neurotropic viruses. Antiviral drugs should cross the blood-brain barrier/blood cerebrospinal fluid barrier and pass the brain cellular membrane to inhibit these viruses inside the brain cells. Some antiviral drugs may also require intracellular conversion into their active metabolite(s). This illustrates the need to better understand these mechanisms because these processes dictate drug exposure within the CNS that ultimately determine the success of antiviral drugs for CNS infections. Finally, we discuss mathematical model-based approaches for optimizing antiviral treatments. Thereby emphasizing the potential of CNS physiologically based pharmacokinetic models because direct measurement of brain intracellular exposure in living humans faces ethical restrictions. Existing physiologically based pharmacokinetic models combined with in vitro pharmacokinetic/pharmacodynamic information can be used to predict drug exposure and evaluate efficacy of antiviral drugs within the CNS, to ultimately optimize the treatments of CNS viral infections.
Collapse
Affiliation(s)
- Ming Sun
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Martijn L Manson
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Tingjie Guo
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Elizabeth C M de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
18
|
Imani MM, Moradi MM, Rezaei F, Mozaffari HR, Sharifi R, Safaei M, Azizi F, Basamtabar M, Sohrabi Z, Shalchi M, Sadeghi M. Association between alcohol dehydrogenase polymorphisms (rs1229984, rs1573496, rs1154460, and rs284787) and susceptibility to head and neck cancers: A systematic review and meta-analysis. Arch Oral Biol 2024; 160:105898. [PMID: 38278126 DOI: 10.1016/j.archoralbio.2024.105898] [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: 10/26/2023] [Revised: 12/25/2023] [Accepted: 01/19/2024] [Indexed: 01/28/2024]
Abstract
OBJECTIVE Head and neck cancer (HNC) is a prevalent and complex group of malignancies with increasing incidence globally. Alcohol dehydrogenases (ADHs) play a crucial role in alcohol metabolism, and their polymorphisms have been linked to HNC risk. This systematic review and meta-analysis aims to evaluate the association between ADH polymorphisms and susceptibility to HNCs, incorporating additional analyses and adding more studies to increase power and accuracy of the results. DESIGN Subgroup analysis, meta-regression analysis, and sensitivity analyses were conducted to explore potential differences within the data and assess the stability of pooled odds ratios (ORs). To mitigate the risk of false conclusions from meta-analyses, a trial sequential analysis was performed. RESULTS For ADH1B rs1229984, the pooled OR (95 % confidence interval (CI)) was 0.73 (0.65, 0.82), 0.42 (0.35, 0.50), 0.57 (0.44, 0.73), 0.56 (0.50, 0.62), and 0.80 (0.73, 0.88), as well as for ADH7 rs1573496, the pooled OR was 0.72 (0.62, 0.85), 0.36 (0.17, 0.74), 0.76 (0.64, 0.91), 0.80 (0.71, 0.91), and 0.38 (0.18, 0.78) with a p < 0.05 in all allelic, homozygous, heterozygous, recessive, and dominant models, respectively. However, no significant association was found between the ADH7 rs1154460 and rs284787 polymorphisms and the risk of HNC with pooled ORs of 1.11 (p = 0.19) and 1.09 (p = 0.24) for the recessive model, respectively. The ethnicities, tumor subsites, control sources, sample sizes, quality scores, and Hardy-Weinberg equilibrium statuses were confounding factors. CONCLUSION The ADH1B rs1229984 and ADH7 rs1573496 polymorphisms are significantly associated with a reduced risk of HNC.
Collapse
Affiliation(s)
- Mohammad Moslem Imani
- Department of Orthodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohamad Mehdi Moradi
- Students Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farzad Rezaei
- Department of Oral and Maxillofacial Surgery, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hamid Reza Mozaffari
- Department of Oral and Maxillofacial Medicine, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Roohollah Sharifi
- Department of Endodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Safaei
- Advanced Dental Sciences Research Center, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Azizi
- Department of Orthodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoumeh Basamtabar
- Department of Orthodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Sohrabi
- Department of periodontology, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Majid Shalchi
- Orthodontic Department, Guilan University of Medical Sciences, School of Dentistry, Rasht, Iran
| | - Masoud Sadeghi
- Medical Biology Research Centre, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
19
|
John T, Saffoon N, Walsby-Tickle J, Hester SS, Dingler FA, Millington CL, McCullagh JSO, Patel KJ, Hopkinson RJ, Schofield CJ. Aldehyde-mediated inhibition of asparagine biosynthesis has implications for diabetes and alcoholism. Chem Sci 2024; 15:2509-2517. [PMID: 38362406 PMCID: PMC10866355 DOI: 10.1039/d3sc06551k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/01/2024] [Indexed: 02/17/2024] Open
Abstract
Patients with alcoholism and type 2 diabetes manifest altered metabolism, including elevated aldehyde levels and unusually low asparagine levels. We show that asparagine synthetase B (ASNS), the only human asparagine-forming enzyme, is inhibited by disease-relevant reactive aldehydes, including formaldehyde and acetaldehyde. Cellular studies show non-cytotoxic amounts of reactive aldehydes induce a decrease in asparagine levels. Biochemical analyses reveal inhibition results from reaction of the aldehydes with the catalytically important N-terminal cysteine of ASNS. The combined cellular and biochemical results suggest a possible mechanism underlying the low asparagine levels in alcoholism and diabetes. The results will stimulate research on the biological consequences of the reactions of aldehydes with nucleophilic residues.
Collapse
Affiliation(s)
- Tobias John
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Nadia Saffoon
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - John Walsby-Tickle
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Svenja S Hester
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford Oxford UK
| | - Felix A Dingler
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital/Headley Way Oxford OX3 9DS UK
| | - Christopher L Millington
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital/Headley Way Oxford OX3 9DS UK
| | - James S O McCullagh
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Ketan J Patel
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital/Headley Way Oxford OX3 9DS UK
| | - Richard J Hopkinson
- Leicester Institute for Structural and Chemical Biology and School of Chemistry, University of Leicester, Henry Wellcome Building Lancaster Road Leicester LE1 7RH UK
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| |
Collapse
|
20
|
Zeng J, Xu S, Lin K, Yao S, Yang B, Peng Z, Hao T, Yu X, Zhu T, Jiang F, Sun J. Long-term stable and efficient degradation of ornidazole with minimized by-product formation by a biological sulfidogenic process based on elemental sulfur. WATER RESEARCH 2024; 249:120940. [PMID: 38071904 DOI: 10.1016/j.watres.2023.120940] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024]
Abstract
Conventional biological treatment processes cannot efficiently and completely degrade nitroimidazole antibiotics, due to the formation of highly antibacterial and carcinogenic nitroreduction by-products. This study investigated the removal of a typical nitroimidazole antibiotic (ornidazole) during wastewater treatment by a biological sulfidogenic process based on elemental sulfur (S0-BSP). Efficient and stable ornidazole degradation and organic carbon mineralization were simultaneously achieved by the S0-BSP in a 798-day bench-scale trial. Over 99.8 % of ornidazole (200‒500 μg/L) was removed with the removal rates of up to 0.59 g/(m3·d). Meanwhile, the efficiencies of organic carbon mineralization and sulfide production were hardly impacted by the dosed ornidazole, and their rates were maintained at 0.15 kg C/(m3·d) and 0.49 kg S/(m3·d), respectively. The genera associated with ornidazole degradation were identified (e.g., Sedimentibacter, Trichococcus, and Longilinea), and their abundances increased significantly. Microbial degradation of ornidazole proceeded by several functional genes, such as dehalogenases, cysteine synthase, and dioxygenases, mainly through dechlorination, denitration, N-heterocyclic ring cleavage, and oxidation. More importantly, the nucleophilic substitution of nitro group mediated by in-situ formed reducing sulfur species (e.g., sulfide, polysulfides, and cysteine hydropolysulfides), instead of nitroreduction, enhanced the complete ornidazole degradation and minimized the formation of carcinogenic and antibacterial nitroreduction by-products. The findings suggest that S0-BSP can be a promising approach to treat wastewater containing multiple contaminants, such as emerging organic pollutants, organic carbon, nitrate, and heavy metals.
Collapse
Affiliation(s)
- Jiajia Zeng
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, School of Environment, South China Normal University, Guangzhou 510006, China; State Environmental Protection Key Laboratory of Drinking Water Source Management and Technology, Shenzhen Key Laboratory of Emerging Contaminants Detection and Control in Water Environment, Shenzhen Academy of Environmental Sciences, Shenzhen 518001, China
| | - Shuqun Xu
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, School of Environment, South China Normal University, Guangzhou 510006, China
| | - Keyue Lin
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, School of Environment, South China Normal University, Guangzhou 510006, China
| | - Si Yao
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, School of Environment, South China Normal University, Guangzhou 510006, China
| | - Bin Yang
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, School of Environment, South China Normal University, Guangzhou 510006, China
| | - Zhanhui Peng
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, School of Environment, South China Normal University, Guangzhou 510006, China
| | - Tianwei Hao
- Department of Civil and Environmental Engineering, Faculty of Science and Technology, University of Macau, Macau 999078, China
| | - Xiaoyu Yu
- Guangdong Polytechnic of Environmental Protection Engineering, Foshan 528216, China
| | - Tingting Zhu
- State Environmental Protection Key Laboratory of Drinking Water Source Management and Technology, Shenzhen Key Laboratory of Emerging Contaminants Detection and Control in Water Environment, Shenzhen Academy of Environmental Sciences, Shenzhen 518001, China
| | - Feng Jiang
- School of Environmental Science & Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Jianliang Sun
- Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, School of Environment, South China Normal University, Guangzhou 510006, China.
| |
Collapse
|
21
|
Sun L, Yang M, Su W, Xu H, Xue F, Lu C, Wu R. Transcriptomic analysis of maize uncovers putative genes involved in metabolic detoxification under four safeners treatment. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 194:105465. [PMID: 37532342 DOI: 10.1016/j.pestbp.2023.105465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/10/2023] [Accepted: 05/14/2023] [Indexed: 08/04/2023]
Abstract
Isoxadifen-ethyl (IDF) and cyprosulfamide (CSA) can effectively protect maize from nicosulfuron (NIC) injury, while mefenpyr-diethyl (MPR) and fenchlorazole-ethyl (FCO) did not. Their chemical diversity and requirement to use them in combination with the corresponding herbicides suggest that their elicitation of gene expression are complex and whether it is associated with the safening activity remains elusive. In this study, our first objective was to determine whether or not the ability of four safeners to enhance the metabolic rate of nicosulfuron. It was found that nicosulfuron degradation in maize was accelerated by IDF and CSA, but not by FCO and MPR. Transcriptomic analysis showed that the number of genes induced by IDF and CSA were larger than that induced by FCO and MPR. Overall, 34 genes associated with detoxification were identified, including glutathione S-transferase (GST), cytochrome P450 (CYP450), UDP-glucosyltransferase (UGT), transporter and serine. Moreover, 14 detoxification genes were screened for further verification by real-time PCR in two maize inbred lines. Two maize inbred lines exhibited high expression levels of four genes (GST31, GST39, AGXT2 and ADH) after IDF treatment. GST6, GST19, MATE, SCPL18 and UF3GT were specifically up-regulated in telerant maize inbred line under IDF and IDF + NIC treatments. Seven genes, namely GST31, GST6, GST19, UF3GT, MATE, ADH and SCPL18, are induced by IDF and CSA to play a vital role in regulating the detoxification process of NIC. Accordingly, the GST activity in maize was accelerated by IDF and CSA, but not by FCO and MPR. This result is consistent with transcriptome and metabolic data.These results indicate that the mitigation of NIC damage is associated with enhanced herbicide metabolism. IDF and CSA were more effective in protecting maize from NIC injury due to their ability to enhance the detoxification of specific types of herbicides, compared to FCO and MPR. The chemical specificity of four safeners is attributed to the up-regulated genes related to the detoxification pathway.
Collapse
Affiliation(s)
- Lanlan Sun
- Henan Key Laboratory of Crop Pest Control, Institute of Plant Protection, Henan Academy of Agricultural Sciences, Zhengzhou, Henan 450002, China
| | - Muhan Yang
- Henan Key Laboratory of Crop Pest Control, Institute of Plant Protection, Henan Academy of Agricultural Sciences, Zhengzhou, Henan 450002, China
| | - Wangcang Su
- Henan Key Laboratory of Crop Pest Control, Institute of Plant Protection, Henan Academy of Agricultural Sciences, Zhengzhou, Henan 450002, China
| | - Hongle Xu
- Henan Key Laboratory of Crop Pest Control, Institute of Plant Protection, Henan Academy of Agricultural Sciences, Zhengzhou, Henan 450002, China
| | - Fei Xue
- Henan Key Laboratory of Crop Pest Control, Institute of Plant Protection, Henan Academy of Agricultural Sciences, Zhengzhou, Henan 450002, China
| | - Chuantao Lu
- Henan Key Laboratory of Crop Pest Control, Institute of Plant Protection, Henan Academy of Agricultural Sciences, Zhengzhou, Henan 450002, China
| | - Renhai Wu
- Henan Key Laboratory of Crop Pest Control, Institute of Plant Protection, Henan Academy of Agricultural Sciences, Zhengzhou, Henan 450002, China.
| |
Collapse
|
22
|
Chan TS, Byer-Alcorace AJ, Latli B, Liu P, Maw HH, Raymond KG, Scaringella YS, Teitelbaum AM, Wang T, Whitcher-Johnstone A, Taub ME. Characterization of Divergent Metabolic Pathways in Elucidating an Unexpected, Slow-Forming, and Long Half-Life Major Metabolite of Iclepertin. Pharm Res 2023; 40:1901-1913. [PMID: 37280472 DOI: 10.1007/s11095-023-03530-z] [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: 03/09/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023]
Abstract
PURPOSE After single oral dosing of the glycine reuptake transporter (GlyT1) inhibitor, iclepertin (BI 425809), a single major circulating metabolite, M530a, was identified. However, upon multiple dosing, a second major metabolite, M232, was observed with exposure levels ~ twofold higher than M530a. Studies were conducted to characterize the metabolic pathways and enzymes responsible for formation of both major human metabolites. METHODS In vitro studies were conducted with human and recombinant enzyme sources and enzyme-selective inhibitors. The production of iclepertin metabolites was monitored by LC-MS/MS. RESULTS Iclepertin undergoes rapid oxidation to a putative carbinolamide that spontaneously opens to an aldehyde, M528, which then undergoes reduction by carbonyl reductase to the primary alcohol, M530a. However, the carbinolamide can also undergo a much slower oxidation by CYP3A to form an unstable imide metabolite, M526, that is subsequently hydrolyzed by a plasma amidase to form M232. This difference in rate of metabolism of the carbinolamine explains why high levels of the M232 metabolite were not observed in vitro and in single dose studies in humans, but were observed in longer-term multiple dose studies. CONCLUSIONS The long half-life iclepertin metabolite M232 is formed from a common carbinolamine intermediate, that is also a precursor of M530a. However, the formation of M232 occurs much more slowly, likely contributing to its extensive exposure in vivo. These results highlight the need to employ adequate clinical study sampling periods and rigorous characterization of unexpected metabolites, especially when such metabolites are categorized as major, thus requiring safety assessment.
Collapse
Affiliation(s)
- Tom S Chan
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT, 06877, USA.
| | - Alexander J Byer-Alcorace
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT, 06877, USA
| | - Bachir Latli
- Department of Chemical Development, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT, 06877, USA
| | - Pingrong Liu
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT, 06877, USA
| | - Hlaing H Maw
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT, 06877, USA
| | - Klairynne G Raymond
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT, 06877, USA
| | - Young-Sun Scaringella
- Department of Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT, 06877, USA
| | - Aaron M Teitelbaum
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Ting Wang
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT, 06877, USA
| | - Andrea Whitcher-Johnstone
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT, 06877, USA
- DMPK Oncology, AstraZeneca Inc., 35 Gatehouse Dr., Waltham, MA, 02451, USA
| | - Mitchell E Taub
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., 900 Ridgebury Rd., Ridgefield, CT, 06877, USA
| |
Collapse
|
23
|
Luo ML, Chen H, Chen GY, Wang S, Wang Y, Yang FQ. Preparation of Alcohol Dehydrogenase-Zinc Phosphate Hybrid Nanoflowers through Biomimetic Mineralization and Its Application in the Inhibitor Screening. Molecules 2023; 28:5429. [PMID: 37513303 PMCID: PMC10386709 DOI: 10.3390/molecules28145429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
A biomimetic mineralization method was used in the facile and rapid preparation of nanoflowers for immobilizing alcohol dehydrogenase (ADH). The method mainly uses ADH as an organic component and zinc phosphate as an inorganic component to prepare flower-like ADH/Zn3(PO4)2 organic-inorganic hybrid nanoflowers (HNFs) with the high specific surface area through a self-assembly process. The synthesis conditions of the ADH HNFs were optimized and its morphology was characterized. Under the optimum enzymatic reaction conditions, the Michaelis-Menten constant (Km) of ADH HNFs (β-NAD+ as substrate) was measured to be 3.54 mM, and the half-maximal inhibitory concentration (IC50) of the positive control ranitidine (0.2-0.8 mM) was determined to be 0.49 mM. Subsequently, the inhibitory activity of natural medicine Penthorum chinense Pursh and nine small-molecule compounds on ADH was evaluated using ADH HNFs. The inhibition percentage of the aqueous extract of P. chinense is 57.9%. The vanillic acid, protocatechuic acid, gallic acid, and naringenin have obvious inhibitory effects on ADH, and their percentages of inhibition are 55.1%, 68.3%, 61.9%, and 75.5%, respectively. Moreover, molecular docking analysis was applied to explore the binding modes and sites of the four most active small-molecule compounds to ADH. The results of this study can broaden the application of immobilized enzymes through biomimetic mineralization, and provide a reference for the discovery of ADH inhibitors from natural products.
Collapse
Affiliation(s)
- Mao-Ling Luo
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 401331, China
| | - Hua Chen
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 401331, China
| | - Guo-Ying Chen
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 401331, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Feng-Qing Yang
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 401331, China
| |
Collapse
|
24
|
Beers J, Authement AK, Isoherranen N, Jackson KD. Cytosolic Enzymes Generate Cannabinoid Metabolites 7-Carboxycannabidiol and 11-Nor-9-carboxytetrahydrocannabinol. ACS Med Chem Lett 2023; 14:614-620. [PMID: 37197460 PMCID: PMC10184666 DOI: 10.1021/acsmedchemlett.3c00017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/17/2023] [Indexed: 05/19/2023] Open
Abstract
The cannabinoids cannabidiol (CBD) and delta-9-tetrahydrocannabinol (THC) undergo extensive oxidative metabolism in the liver. Although cytochromes P450 form the primary, pharmacologically active, hydroxylated metabolites of CBD and THC, less is known about the enzymes that generate the major in vivo circulating metabolites of CBD and THC, 7-carboxy-CBD and 11-carboxy-THC, respectively. The purpose of this study was to elucidate the enzymes involved in forming these metabolites. Cofactor dependence experiments with human liver subcellular fractions revealed that 7-carboxy-CBD and 11-carboxy-THC formation is largely dependent on cytosolic NAD+-dependent enzymes, with lesser contributions from NADPH-dependent microsomal enzymes. Experiments with chemical inhibitors provided evidence that 7-carboxy-CBD formation is mainly dependent on aldehyde dehydrogenases and 11-carboxy-THC formation is mediated also in part by aldehyde oxidase. This study is the first to demonstrate the involvement of cytosolic drug-metabolizing enzymes in generating major in vivo metabolites of CBD and THC and addresses a knowledge gap in cannabinoid metabolism.
Collapse
Affiliation(s)
- Jessica
L. Beers
- Division
of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| | - Aurora K. Authement
- Department
of Pharmaceutics, University of Washington
School of Pharmacy, Seattle, Washington 98195, United States
| | - Nina Isoherranen
- Department
of Pharmaceutics, University of Washington
School of Pharmacy, Seattle, Washington 98195, United States
| | - Klarissa D. Jackson
- Division
of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill Eshelman School of Pharmacy, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
25
|
Peng M, Wang Z, Sun X, Guo X, Wang H, Li R, Liu Q, Chen M, Chen X. Deep Learning-Based Label-Free Surface-Enhanced Raman Scattering Screening and Recognition of Small-Molecule Binding Sites in Proteins. Anal Chem 2022; 94:11483-11491. [PMID: 35968807 DOI: 10.1021/acs.analchem.2c01158] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Identification of small-molecule binding sites in proteins is of great significance in analysis of protein function and drug design. Modified sites can be recognized via proteolytic cleavage followed by liquid chromatography-mass spectrometry (LC-MS); however, this has always been impeded by the complexity of peptide mixtures and the elaborate synthetic design for tags. Here, we demonstrate a novel technique for identifying protein binding sites using a deep learning-based label-free surface-enhanced Raman scattering (SERS) screening (DLSS) strategy. In DLSS, the deep learning model that was trained with large SERS signals could detect signal features of small molecules with high accuracy (>99%). Without any secondary tag, the small molecules are directly complexed with proteins. After proteolysis and LC, SERS signals of all LC fractions are collected and input into the model, whereby the fractions containing the small-molecule-modified peptides can be recognized by the model and sent to MS/MS to identify the binding site(s). By using an automated DLSS system, we successfully identified the modification sites of fomepizole in alcohol dehydrogenase, which is coordinated with zinc along with three peptides. We also showed that the DLSS strategy works for identification of amino-acid residues that covalently bond with ibrutinib in Bruton tyrosine kinase. These results suggest that the DLSS strategy, which provides high molecular recognition capability to LC-MS analysis, has potential in drug discovery, proteomics, and metabolomics.
Collapse
Affiliation(s)
- Mei Peng
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Zi Wang
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Xiaotong Sun
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Xiangwei Guo
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Haoyang Wang
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Ruili Li
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Qi Liu
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Miao Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China.,School of Life Sciences, Central South University, Changsha 410013, China
| | - Xiaoqing Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
26
|
Inhibition mechanism of baicalein against alcohol dehydrogenase in vitro via biological techniques, spectroscopy and computer simulation. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
27
|
Bolleddula J, Chen H, Cohen L, Zhou X, Pusalkar S, Berger A, Sedarati F, Venkatakrishnan K, Chowdhury SK. Metabolism and Disposition of [ 14C]Pevonedistat, a First-in-Class NEDD8-Activating Enzyme Inhibitor, after Intravenous Infusion to Patients with Advanced Solid Tumors. Drug Metab Dispos 2022; 50:989-997. [PMID: 35504658 DOI: 10.1124/dmd.122.000842] [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: 01/27/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022] Open
Abstract
Metabolism and disposition of pevonedistat, an investigational, first-in-class inhibitor of the NEDD8-activating enzyme (NAE), were characterized in patients with advanced solid tumors after intravenous infusion of [14C]pevonedistat at 25 mg/m2 (∼60-85 μCi radioactive dose). More than 94% of the administered dose was recovered, with ∼41% and ∼53% of drug-related material eliminated in urine and feces, respectively. The metabolite profiles of [14C]pevonedistat were established in plasma using an accelerator mass spectrometer and excreta with traditional radiometric analysis. In plasma, unchanged parent drug accounted for approximately 49% of the total drug-related material. Metabolites M1 and M2 were major (>10% of the total drug-related material) circulating metabolites and accounted for approximately 15% and 22% of the drug-related material, respectively. Unchanged [14C]pevonedistat accounted for approximately 4% and 17% of the dose in urine and feces, respectively. Oxidative metabolites M1, M2, and M3 appeared as the most abundant drug-related components in the excreta and represented approximately 27%, 26%, and 15% of the administered dose, respectively. Based on the unbound plasma exposure in cancer patients and in vitro NAE inhibition, the contribution of metabolites M1 and M2 to overall in vivo pharmacological activity is anticipated to be minimal. The exposure to these metabolites was higher at safe and well tolerated doses in rat and dog (the two preclinical species used in toxicology evaluation) plasma than that observed in human plasma. Reaction phenotyping studies revealed that CYP3A4/5 are primary enzymes responsible for the metabolic clearance of pevonedistat. SIGNIFICANCE STATEMENT: This study details the metabolism and clearance mechanisms of pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, after intravenous administration to patients with cancer. Pevonedistat is biotransformed to two major circulating metabolites with higher exposure in nonclinical toxicological species than in humans. The pharmacological activity contribution of these metabolites is minimal compared to the overall target pharmacological effect of pevonedistat. Renal clearance was not an important route of excretion of unchanged pevonedistat (∼4% of the dose).
Collapse
Affiliation(s)
| | - Hao Chen
- Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Lawrence Cohen
- Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Xiaofei Zhou
- Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | | | - Allison Berger
- Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | - Farhad Sedarati
- Takeda Development Center Americas, Inc., Lexington, Massachusetts
| | | | | |
Collapse
|
28
|
Sato R, Ohmori K, Umetsu M, Takao M, Tano M, Grant G, Porter B, Bet A, Terasaki T, Uchida Y. An Atlas of the Quantitative Protein Expression of Anti-Epileptic-Drug Transporters, Metabolizing Enzymes and Tight Junctions at the Blood-Brain Barrier in Epileptic Patients. Pharmaceutics 2021; 13:pharmaceutics13122122. [PMID: 34959403 PMCID: PMC8708024 DOI: 10.3390/pharmaceutics13122122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/02/2021] [Accepted: 12/08/2021] [Indexed: 01/06/2023] Open
Abstract
The purpose of the present study was to quantitatively elucidate the levels of protein expression of anti-epileptic-drug (AED) transporters, metabolizing enzymes and tight junction molecules at the blood–brain barrier (BBB) in the focal site of epilepsy patients using accurate SWATH (sequential window acquisition of all theoretical fragment ion spectra) proteomics. Brain capillaries were isolated from focal sites in six epilepsy patients and five normal brains; tryptic digests were produced and subjected to SWATH analysis. MDR1 and BCRP were significantly downregulated in the epilepsy group compared to the normal group. Out of 16 AED-metabolizing enzymes detected, the protein expression levels of GSTP1, GSTO1, CYP2E1, ALDH1A1, ALDH6A1, ALDH7A1, ALDH9A1 and ADH5 were significantly 2.13-, 6.23-, 2.16-, 2.80-, 1.73-, 1.67-, 2.47- and 2.23-fold greater in the brain capillaries of epileptic patients than those of normal brains, respectively. The protein expression levels of Claudin-5, ZO-1, Catenin alpha-1, beta-1 and delta-1 were significantly lower, 1.97-, 2.51-, 2.44-, 1.90- and 1.63-fold, in the brain capillaries of epileptic patients compared to those of normal brains, respectively. Consistent with these observations, leakage of blood proteins was also observed. These results provide for a better understanding of the therapeutic effect of AEDs and molecular mechanisms of AED resistance in epileptic patients.
Collapse
Affiliation(s)
- Risa Sato
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (R.S.); (K.O.); (M.U.); (T.T.)
| | - Kotaro Ohmori
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (R.S.); (K.O.); (M.U.); (T.T.)
| | - Mina Umetsu
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (R.S.); (K.O.); (M.U.); (T.T.)
| | - Masaki Takao
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki 372-0006, Japan; (M.T.); (M.T.)
- Department of Clinical Laboratory, National Center of Neurology and Psychiatry, National Center Hospital, Kodaira 187-8551, Japan
| | - Mitsutoshi Tano
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki 372-0006, Japan; (M.T.); (M.T.)
| | - Gerald Grant
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; (G.G.); (A.B.)
| | - Brenda Porter
- Department of Neurology, Stanford University, Stanford, CA 94305, USA;
| | - Anthony Bet
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; (G.G.); (A.B.)
| | - Tetsuya Terasaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (R.S.); (K.O.); (M.U.); (T.T.)
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan; (R.S.); (K.O.); (M.U.); (T.T.)
- Correspondence: ; Tel.: +81-22-795-6832
| |
Collapse
|
29
|
Nardotto GHB, Bollela VR, Rocha A, Della Pasqua O, Lanchote VL. No implication of HIV coinfection on the plasma exposure to rifampicin, pyrazinamide, and ethambutol in tuberculosis patients. Clin Transl Sci 2021; 15:514-523. [PMID: 34670022 PMCID: PMC8841449 DOI: 10.1111/cts.13169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/31/2021] [Accepted: 09/05/2021] [Indexed: 11/28/2022] Open
Abstract
There are contrasting findings regarding the effect of HIV on the pharmacokinetics of first‐line anti‐tubercular drugs (FLATDs) due to a lack of prospective controlled clinical studies, including patients with tuberculosis (TB) and patients with TB living with HIV. This study aims to assess the effect of HIV coinfection and antiviral therapy on the plasma exposure to FLATDs in patients with TB. HIV negative (TB‐HIV− group; n = 15) and HIV positive (TB‐HIV+ group; n = 18) adult patients with TB were enrolled during the second month of FLATDs treatment. All TB‐HIV+ patients were on treatment with lamivudine, tenofovir (or zidovudine), and raltegravir (or efavirenz). Serial blood sampling was collected over 24 h and FLATDs pharmacokinetic parameters were evaluated using noncompartmental methods. In the TB‐HIV+ patients, dose‐normalized plasma exposure area under the curve from zero to 24 h (nAUC0–24; geometric mean and 95% confidence interval [CI]) values at steady‐state to rifampicin, pyrazinamide, and ethambutol were 18.38 (95% CI 13.74–24.59), 238.21 (95% CI 191.09–296.95), and 18.33 (95% CI 14.56–23.09) µg∙h/ml, respectively. Similar plasma exposure was found in the TB‐HIV− patients. The geometric mean and 90% CI of the ratios between TB‐HIV− and TB‐HIV+ groups suggest no significant pharmacokinetic interaction between the selected antivirals and FLATDs. Likewise, HIV coinfection itself does not appear to have any effect on the plasma exposure to FLATDs.
Collapse
Affiliation(s)
| | - Valdes Roberto Bollela
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Adriana Rocha
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Oscar Della Pasqua
- Clinical Pharmacology & Therapeutics Group, School of Pharmacy - University College London, London, UK
| | - Vera Lucia Lanchote
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
30
|
Non-cytochrome P450 enzymes involved in the oxidative metabolism of xenobiotics: Focus on the regulation of gene expression and enzyme activity. Pharmacol Ther 2021; 233:108020. [PMID: 34637840 DOI: 10.1016/j.pharmthera.2021.108020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/25/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Oxidative metabolism is one of the major biotransformation reactions that regulates the exposure of xenobiotics and their metabolites in the circulatory system and local tissues and organs, and influences their efficacy and toxicity. Although cytochrome (CY)P450s play critical roles in the oxidative reaction, extensive CYP450-independent oxidative metabolism also occurs in some xenobiotics, such as aldehyde oxidase, xanthine oxidoreductase, flavin-containing monooxygenase, monoamine oxidase, alcohol dehydrogenase, or aldehyde dehydrogenase-dependent oxidative metabolism. Drugs form a large portion of xenobiotics and are the primary target of this review. The common reaction mechanisms and roles of non-CYP450 enzymes in metabolism, factors affecting the expression and activity of non-CYP450 enzymes in terms of inhibition, induction, regulation, and species differences in pharmaceutical research and development have been summarized. These non-CYP450 enzymes are detoxifying enzymes, although sometimes they mediate severe toxicity. Synthetic or natural chemicals serve as inhibitors for these non-CYP450 enzymes. However, pharmacokinetic-based drug interactions through these inhibitors have rarely been reported in vivo. Although multiple mechanisms participate in the basal expression and regulation of non-CYP450 enzymes, only a limited number of inducers upregulate their expression. Therefore, these enzymes are considered non-inducible or less inducible. Overall, this review focuses on the potential xenobiotic factors that contribute to variations in gene expression levels and the activities of non-CYP450 enzymes.
Collapse
|
31
|
de Matos IL, Birolli WG, Santos DDA, Nitschke M, Porto ALM. Stereoselective reduction of flavanones by marine-derived fungi. MOLECULAR CATALYSIS 2021. [DOI: 10.1016/j.mcat.2021.111734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
32
|
Dhuria NV, Haro B, Kapadia A, Lobo KA, Matusow B, Schleiff MA, Tantoy C, Sodhi JK. Recent developments in predicting CYP-independent metabolism. Drug Metab Rev 2021; 53:188-206. [PMID: 33941024 DOI: 10.1080/03602532.2021.1923728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
As lead optimization efforts have successfully reduced metabolic liabilities due to cytochrome P450 (CYP)-mediated metabolism, there has been an increase in the frequency of involvement of non-CYP enzymes in the metabolism of investigational compounds. Although there have been numerous notable advancements in the characterization of non-CYP enzymes with respect to their localization, reaction mechanisms, species differences and identification of typical substrates, accurate prediction of non-CYP-mediated clearance, with a particular emphasis with the difficulties in accounting for any extrahepatic contributions, remains a challenge. The current manuscript comprehensively summarizes the recent advancements in the prediction of drug metabolism and the in vitro to in vitro extrapolation of clearance for substrates of non-CYP drug metabolizing enzymes.
Collapse
Affiliation(s)
- Nikhilesh V Dhuria
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bianka Haro
- School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Amit Kapadia
- California Poison Control Center, University of California San Francisco, San Diego, CA, USA
| | | | - Bernice Matusow
- Department of Drug Metabolism and Pharmacokinetics, Plexxikon Inc, Berkeley, CA, USA
| | - Mary A Schleiff
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Christina Tantoy
- Department of Drug Metabolism and Pharmacokinetics, Plexxikon Inc, Berkeley, CA, USA
| | - Jasleen K Sodhi
- Department of Drug Metabolism and Pharmacokinetics, Plexxikon Inc, Berkeley, CA, USA.,Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|