1
|
Li H, Zhou Y, Cai C, Liang H, Li X, Huang M, Fan S, Bi H. Fenofibrate induces liver enlargement in aging mice via activating the PPARα-YAP signaling pathway. Chem Biol Interact 2025; 405:111286. [PMID: 39442682 DOI: 10.1016/j.cbi.2024.111286] [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: 09/06/2024] [Revised: 10/12/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024]
Abstract
Fenofibrate is a clinically prescribed drug for treating hypertriglyceridemia, which is also a classic peroxisome proliferator-activated receptor α (PPARα) agonist. We previously reported that fenofibrate induced liver enlargement in adult mice partially through activation of the yes-associated protein (YAP) signaling pathway. However, the effects of fenofibrate on liver enlargement and the YAP signaling pathway in aging mice remain unclear. In this study, D-galactose-induced aging mice, naturally aging mice, and senescence-accelerated mice P8 (SAMP8) were used to investigate the effects of aging on fenofibrate-induced liver enlargement and YAP signaling activation. The results showed that fenofibrate-induced liver enlargement in aging mice was consistent with that of adult mice. The effects of fenofibrate on hepatocyte enlargement around the central vein (CV) area and hepatocyte proliferation around the portal vein (PV) area were comparable between adult and aging mice. There was no significant difference in the upregulation of PPARα downstream proteins between the two groups following fenofibrate treatment. Fenofibrate treatment also increased the expression of proliferation-related proteins and activated the YAP signaling pathway to a similar degree in both groups. In summary, these results demonstrate that the fenofibrate-induced liver enlargement and activation of the YAP pathway are consistent between adult and aging mice, indicating that the effects of fenofibrate on promoting liver enlargement and its activation of the PPARα and YAP pathway were independent of aging. These findings offer a new perspective for the clinical use of fenofibrate in elderly patients and provide a new insight for the role of PPARα in liver enlargement.
Collapse
Affiliation(s)
- Huilin Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Yanying Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Chenghui Cai
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Hangfei Liang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Xuan Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Shicheng Fan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.
| | - Huichang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China; The State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School of Peking University, Shenzhen, 518055, China.
| |
Collapse
|
2
|
Kaya B, Akduman H, Dilli D, Kaya Ö, Çitli R, Zenciroğlu A. The Utilization of Acetaminophen for Managing PGE1-Induced Fever in Neonates with Critical Congenital Heart Disease. CHILDREN 2024; 11:1547. [DOI: 10.3390/children11121547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Introduction: We aimed to retrospectively evaluate the use of acetaminophen, which may be a risk factor for the ductal canal, in the treatment of fever due to prostaglandin E1 (PGE1) infusion in newborns with critical congenital heart disease (CCHD). Methods: The study included newborns who were followed-up in our neonatal intensive care unit with the diagnosis of critical congenital heart disease, developed fever due to PGE1 infusion and had acetaminophen administered for antipyretic treatment. The patent ductus arteriosus diameters of the patients were evaluated by echocardiographic imaging before intravenous acetaminophen treatment and at the end of the day of acetaminophen treatment. Results: PGE1 fever was observed in 10 (6%) of 156 infants with ductus-dependent congenital heart disease. Intravenous acetaminophen treatment administered as an antipyretic in these infants did not cause the narrowing or closure of the ductal canal diameter, which would lead to clinical decompensation if it was closed, and the patients remained hemodynamically stable until surgery. Conclusions: It can be considered that controlled and rapidly administered intravenous acetaminophen therapy in the management of fever in neonates with congenital heart disease who develop fever as a side effect of high-dose intravenous PGE1 infusion therapy (≥0.3 mcg/kg/min) may prevent hemodynamic decompensation in these critically ill infants, and as a secondary outcome, it can be speculated that avoiding rapid daily increases in PGE1 maintenance infusion doses may be necessary to limit the number of paracetamol administrations in these infants.
Collapse
Affiliation(s)
- Başak Kaya
- Department of Neonatology, SBU. Dr. Sami Ulus Maternity and Child Research and Training Hospital, Babur St., Number: 36, Altındag 06080, Turkey
| | - Hasan Akduman
- Department of Neonatology, SBU. Dr. Sami Ulus Maternity and Child Research and Training Hospital, Babur St., Number: 36, Altındag 06080, Turkey
| | - Dilek Dilli
- Department of Neonatology, SBU. Dr. Sami Ulus Maternity and Child Research and Training Hospital, Babur St., Number: 36, Altındag 06080, Turkey
| | - Özkan Kaya
- Department of Pediatric Cardiology, SBU. Dr. Sami Ulus Maternity and Child Research and Training Hospital, Babur St., Number: 36, Altındag 06080, Turkey
| | - Rumeysa Çitli
- Department of Neonatology, SBU. Dr. Sami Ulus Maternity and Child Research and Training Hospital, Babur St., Number: 36, Altındag 06080, Turkey
| | - Ayşegül Zenciroğlu
- Department of Neonatology, SBU. Dr. Sami Ulus Maternity and Child Research and Training Hospital, Babur St., Number: 36, Altındag 06080, Turkey
| |
Collapse
|
3
|
Mokhosoev IM, Astakhov DV, Terentiev AA, Moldogazieva NT. Cytochrome P450 monooxygenase systems: Diversity and plasticity for adaptive stress response. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 193:19-34. [PMID: 39245215 DOI: 10.1016/j.pbiomolbio.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/21/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Superfamily of cytochromes P450 (CYPs) is composed of heme-thiolate-containing monooxygenase enzymes, which play crucial roles in the biosynthesis, bioactivation, and detoxification of a variety of organic compounds, both endogenic and exogenic. Majority of CYP monooxygenase systems are multi-component and contain various redox partners, cofactors and auxiliary proteins, which contribute to their diversity in both prokaryotes and eukaryotes. Recent progress in bioinformatics and computational biology approaches make it possible to undertake whole-genome and phylogenetic analyses of CYPomes of a variety of organisms. Considerable variations in sequences within and between CYP families and high similarity in secondary and tertiary structures between all CYPs along with dramatic conformational changes in secondary structure elements of a substrate binding site during catalysis have been reported. This provides structural plasticity and substrate promiscuity, which underlie functional diversity of CYPs. Gene duplication and mutation events underlie CYP evolutionary diversity and emergence of novel selectable functions, which provide the involvement of CYPs in high adaptability to changing environmental conditions and dietary restrictions. In our review, we discuss the recent advancements and challenges in the elucidating the evolutionary origin and mechanisms underlying the CYP monooxygenase system diversity and plasticity. Our review is in the view of hypothesis that diversity of CYP monooxygenase systems is translated into the broad metabolic profiles, and this has been acquired during the long evolutionary time to provide structural plasticity leading to high adaptative capabilities to environmental stress conditions.
Collapse
Affiliation(s)
| | - Dmitry V Astakhov
- Department of Biochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Alexander A Terentiev
- Department of Biochemistry and Molecular Biology, N.I. Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | | |
Collapse
|
4
|
Zhu D, Arnold M, Samuelson BA, Wu JZ, Mueller A, Sinclair DA, Kane AE. Sex dimorphism and tissue specificity of gene expression changes in aging mice. Biol Sex Differ 2024; 15:89. [PMID: 39482778 PMCID: PMC11529319 DOI: 10.1186/s13293-024-00666-4] [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: 07/31/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Aging is a complex process that involves all tissues in an organism and shows sex dimorphism. While transcriptional changes in aging have been well characterized, the majority of studies have focused on a single sex and sex differences in gene expression in aging are poorly understood. In this study, we explore sex dimorphism in gene expression in aging mice across three tissues. METHODS We collected gastrocnemius muscle, liver and white adipose tissue from young (6 months, n = 14) and old (24 months, n = 14) female and male C57BL/6NIA mice and performed RNA-seq. To investigate sex dimorphism in aging, we considered two levels of comparisons: (a) differentially expressed genes between females and males in the old age group and (b) comparisons between females and males across the aging process. We utilized differential expression analysis and gene feature selection to investigate candidate genes. Gene set enrichment analysis was performed to identify candidate molecular pathways. Furthermore, we performed a co-expression network analysis and chose the gene module(s) associated with aging independent of sex or tissue-type. RESULTS We identified both tissue-specific and tissue-independent genes associated with sex dimorphism in aged mice. Unique differentially expressed genes between old males and females across tissues were mainly enriched for pathways related to specific tissue function. We found similar results when exploring sex differences in the aging process, with the exception that in the liver genes enriched for lipid metabolism and digestive system were identified in both females and males. Combining enriched pathways across analyses, we identified amino acid metabolism, digestive system, and lipid metabolism as the core mechanisms of sex dimorphism in aging. Although the vast majority of age-related genes were sex and tissue specific, we identified 127 hub genes contributing to aging independent of sex and tissue that were enriched for the immune system and signal transduction. CONCLUSIONS There are clear sex differences in gene expression in aging across liver, muscle and white adipose. Core pathways, including amino acid metabolism, digestive system and lipid metabolism, contribute to sex differences in aging.
Collapse
Affiliation(s)
- Dantong Zhu
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Matt Arnold
- Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Blavatnik Institute, Boston, MA, 02115, USA
| | | | - Judy Z Wu
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Amber Mueller
- Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Blavatnik Institute, Boston, MA, 02115, USA
- Cell Press, Cambridge, MA, 02139, USA
| | - David A Sinclair
- Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Blavatnik Institute, Boston, MA, 02115, USA
| | - Alice E Kane
- Institute for Systems Biology, Seattle, WA, 98109, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98115, USA.
| |
Collapse
|
5
|
Guerrero L, Carmona-Rodríguez L, Santos FM, Ciordia S, Stark L, Hierro L, Pérez-Montero P, Vicent D, Corrales FJ. Molecular basis of progressive familial intrahepatic cholestasis 3. A proteomics study. Biofactors 2024; 50:794-809. [PMID: 38284625 DOI: 10.1002/biof.2041] [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: 07/07/2023] [Accepted: 12/18/2023] [Indexed: 01/30/2024]
Abstract
Progressive familial intrahepatic cholestasis type 3 (PFIC3) is a severe rare liver disease that affects between 1/50,000 and 1/100,000 children. In physiological conditions, bile is produced by the liver and stored in the gallbladder, and then it flows to the small intestine to play its role in fat digestion. To prevent tissue damage, bile acids (BAs) are kept in phospholipid micelles. Mutations in phosphatidyl choline transporter ABCB4 (MDR3) lead to intrahepatic accumulation of free BAs that result in liver damage. PFIC3 onset usually occurs at early ages, progresses rapidly, and the prognosis is poor. Currently, besides the palliative use of ursodeoxycholate, the only available treatment for this disease is liver transplantation, which is really challenging for short-aged patients. To gain insight into the pathogenesis of PFIC3 we have performed an integrated proteomics and phosphoproteomics study in human liver samples to then validate the emerging functional hypotheses in a PFIC3 murine model. We identified 6246 protein groups, 324 proteins among them showing differential expression between control and PFIC3. The phosphoproteomic analysis allowed the identification of 5090 phosphopeptides, from which 215 corresponding to 157 protein groups, were differentially phosphorylated in PFIC3, including MDR3. Regulation of essential cellular processes and structures, such as inflammation, metabolic reprogramming, cytoskeleton and extracellular matrix remodeling, and cell proliferation, were identified as the main drivers of the disease. Our results provide a strong molecular background that significantly contributes to a better understanding of PFIC3 and provides new concepts that might prove useful in the clinical management of patients.
Collapse
Affiliation(s)
- Laura Guerrero
- Functional Proteomics Labortory, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | | | - Fátima Milhano Santos
- Functional Proteomics Labortory, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Sergio Ciordia
- Functional Proteomics Labortory, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Luiz Stark
- IdiPAZ, Instituto de Investigación Sanitaria [Health Research Institute] of Hospital Universitario La Paz, Madrid, Spain
| | - Loreto Hierro
- IdiPAZ, Instituto de Investigación Sanitaria [Health Research Institute] of Hospital Universitario La Paz, Madrid, Spain
| | - Pablo Pérez-Montero
- Servicio de Anatomía Patológica, Hospital Universitario La Paz, Madrid, Spain
| | - David Vicent
- IdiPAZ, Instituto de Investigación Sanitaria [Health Research Institute] of Hospital Universitario La Paz, Madrid, Spain
| | - Fernando J Corrales
- Functional Proteomics Labortory, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| |
Collapse
|
6
|
Cho S, Jo H, Hwang YJ, Kim C, Jo YH, Yun JW. Potential impact of underlying diseases influencing ADME in nonclinical safety assessment. Food Chem Toxicol 2024; 188:114636. [PMID: 38582343 DOI: 10.1016/j.fct.2024.114636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/19/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
Nonclinical studies involve in vitro, in silico, and in vivo experiments to assess the toxicokinetics, toxicology, and safety pharmacology of drugs according to regulatory requirements by a national or international authority. In this review, we summarize the potential effects of various underlying diseases governing the absorption, distribution, metabolism, and excretion (ADME) of drugs to consider the use of animal models of diseases in nonclinical trials. Obesity models showed alterations in hepatic metabolizing enzymes, transporters, and renal pathophysiology, which increase the risk of drug-induced toxicity. Diabetes models displayed changes in hepatic metabolizing enzymes, transporters, and glomerular filtration rates (GFR), leading to variability in drug responses and susceptibility to toxicity. Animal models of advanced age exhibited impairment of drug metabolism and kidney function, thereby reducing the drug-metabolizing capacity and clearance. Along with changes in hepatic metabolic enzymes, animal models of metabolic syndrome-related hypertension showed renal dysfunction, resulting in a reduced GFR and urinary excretion of drugs. Taken together, underlying diseases can induce dysfunction of organs involved in the ADME of drugs, ultimately affecting toxicity. Therefore, the use of animal models of representative underlying diseases in nonclinical toxicity studies can be considered to improve the predictability of drug side effects before clinical trials.
Collapse
Affiliation(s)
- Sumin Cho
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Harin Jo
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yeon Jeong Hwang
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Changuk Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Yong Hyeon Jo
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
7
|
Abudahab S, Slattum PW, Price ET, McClay JL. Epigenetic regulation of drug metabolism in aging: utilizing epigenetics to optimize geriatric pharmacotherapy. Pharmacogenomics 2024; 25:41-54. [PMID: 38126340 PMCID: PMC10794944 DOI: 10.2217/pgs-2023-0199] [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: 10/19/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
We explore the relationship between epigenetic aging and drug metabolism. We review current evidence for changes in drug metabolism in normal aging, followed by a description of how epigenetic modifications associated with age can regulate the expression and functionality of genes. In particular, we focus on the role of epigenome-wide studies of human and mouse liver in understanding these age-related processes with respect to xenobiotic processing. We highlight genes encoding drug metabolizing enzymes and transporters revealed to be affected by epigenetic aging in these studies. We conclude that substantial evidence exists for epigenetic aging impacting drug metabolism and transport genes, but more work is needed. We further highlight the promise of pharmacoepigenetics applied to enhancing drug safety in older adults.
Collapse
Affiliation(s)
- Sara Abudahab
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Patricia W Slattum
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
- Virginia Center on Aging, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Elvin T Price
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Joseph L McClay
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
8
|
Umar TP, Tanasov A, Stevanny B, Agustini D, Dave T, Nabhan A, Madany M, Ibrahim M, Nguyen D, Jain S, Jain N. A Digital Health Perspective on Medication Use and Polypharmacy Management for Improving Healthcare Outcomes in Geriatric Patients. ADVANCES IN MEDICAL DIAGNOSIS, TREATMENT, AND CARE 2023:1-39. [DOI: 10.4018/979-8-3693-0260-6.ch001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The high prevalence of multiple comorbidities poses unique medication-related challenges for geriatric patients. Polypharmacy is a particular concern since taking several medications simultaneously increases the likelihood of adverse drug events and the risk of drug interactions while decreasing patient adherence. These factors are associated with suboptimal health outcomes and a heightened burden on the healthcare system (insurance claims) and the patient (out-of-pocket expenses). These challenges can significantly affect the quality of life of geriatric patients. This chapter critically examines the impact of medication use and polypharmacy on the quality of life of older patients. In addition, the authors discuss how artificial intelligence-based digital tools and precision medicine can address these issues by streamlining medical decision-making, improving the patient experience, and allowing remote monitoring. Finally, they interpret the findings from the lens of ethical considerations associated with the adoption and implementation of digital applications and gadgets.
Collapse
Affiliation(s)
| | - Andrei Tanasov
- Carol Davila University of Medicine and Pharmacy, Romania
| | | | | | - Tirth Dave
- Bukovinian State Medical University, Ukraine
| | - Ayman Nabhan
- Al Andalus University for Medical Sciences, Syria
| | | | - Muiz Ibrahim
- International Higher School of Medicine, International University of Kyrgyzstan, Kyrgyzstan
| | | | - Shivani Jain
- Genesis Institute of Dental Sciences and Research, India
| | | |
Collapse
|
9
|
Haduch A, Bromek E, Kuban W, Basińska-Ziobroń A, Danek PJ, Alenina N, Bader M, Daniel WA. The effect of brain serotonin deficit (TPH2-KO) on the expression and activity of liver cytochrome P450 enzymes in aging male Dark Agouti rats. Pharmacol Rep 2023; 75:1522-1532. [PMID: 37848703 PMCID: PMC10661807 DOI: 10.1007/s43440-023-00540-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: 08/14/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Liver cytochrome P450 (CYP) greatly contributes to the metabolism of endogenous substances and drugs. Recent studies have demonstrated that CYP expression in the liver is controlled by the central nervous system via hormonal pathways. In particular, the expression of hepatic CYPs is negatively regulated by the brain serotoninergic system. The present study aimed to investigate changes in the function of the main liver drug-metabolizing CYP enzymes as a result of serotonin depletion in the brain of aging rats, caused by knockout of brain tryptophan hydroxylase gene (TPH2-KO). METHODS The hepatic CYP mRNA (qRT-PCR), protein level (Western blotting) and activity (HPLC), and serum hormone levels (ELISA) were measured in Dark Agouti wild-type (WT) male rats (mature 3.5-month-old and senescent 21-month-old) and in TPH2-KO senescent animals. RESULTS The expression/activity of the studied CYPs decreased with age in the liver of wild-type rats. The deprivation of serotonin in the brain of aging males decreased the mRNA level of most of the studied CYPs (CYP1A/2A/2B/3A), and lowered the protein level of CYP2C11 and CYP3A. In contrast, the activities of CYP2C11, CYP3A and CYP2C6 were increased. The expression of cytochrome b5 decreased in aging rats, but increased in TPH2-deficient senescent animals. The serum concentration of growth hormone declined in the aged and further dropped down in TPH2-deficient senescent rats. CONCLUSIONS Rat liver cytochrome P450 functions deteriorate with age, which may impair drug metabolism. The TPH2 knockout, which deprives brain serotonin, affects cytochrome P450 expression and activity differently in mature and senescent male rats.
Collapse
Affiliation(s)
- Anna Haduch
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Ewa Bromek
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Wojciech Kuban
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Agnieszka Basińska-Ziobroń
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Przemysław J Danek
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland
| | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
- Charité University Medicine, Berlin, Germany
| | - Władysława A Daniel
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| |
Collapse
|
10
|
Wright CJ, McCulley DJ, Mitra S, Jensen EA. Acetaminophen for the patent ductus arteriosus: has safety been adequately demonstrated? J Perinatol 2023; 43:1230-1237. [PMID: 37169914 PMCID: PMC10626600 DOI: 10.1038/s41372-023-01697-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
Patent ductus arteriosus (PDA) is the most common cardiovascular condition diagnosed in premature infants. Acetaminophen was first proposed as a potential treatment for PDA in 2011. Since that time acetaminophen use among extremely preterm neonates has increased substantially. The limited available data demonstrate that acetaminophen reduces PDA without evident hepatotoxicity. These findings have led some to suggest that acetaminophen is a safe and effective therapy for PDA closure. However, the lack of apparent hepatoxicity is predictable. Acetaminophen induced cellular injury is due to CYP2E1 derived metabolites; and hepatocyte CYP2E1 expression is low in the fetal and neonatal period. Here, we review preclinical and clinical data that support the hypothesis that the lung, which expresses high levels of CYP2E1 during fetal and early postnatal development, may be particularly susceptible to acetaminophen induced toxicity. Despite these emerging data, the true potential pulmonary risks and benefits of acetaminophen for PDA closure are largely unknown. The available clinical studies in are marked by significant weakness including low sample sizes and minimal evaluation of extremely preterm infants who are typically at highest risk of pulmonary morbidity. We propose that studies interrogating mechanisms linking developmentally regulated, cell-specific CYP2E1 expression and acetaminophen-induced toxicity as well as robust assessment of pulmonary outcomes in large trials that evaluate the safety and efficacy of acetaminophen in extremely preterm infants are needed.
Collapse
Affiliation(s)
- Clyde J Wright
- Section of Neonatology, Department of Pediatrics, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO, USA.
| | - David J McCulley
- Division of Neonatology, Department of Pediatrics, University of California, San Diego, CA, USA
| | - Souvik Mitra
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Dalhousie University and IWK Health Centre, Halifax, NS, Canada
| | - Erik A Jensen
- Division of Neonatology, Department of Pediatrics, The Children's Hospital of Philadelphia and The University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
11
|
Zhang J, Qiu Z, Zhang Y, Wang G, Hao H. Intracellular spatiotemporal metabolism in connection to target engagement. Adv Drug Deliv Rev 2023; 200:115024. [PMID: 37516411 DOI: 10.1016/j.addr.2023.115024] [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: 04/25/2023] [Revised: 07/05/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
The metabolism in eukaryotic cells is a highly ordered system involving various cellular compartments, which fluctuates based on physiological rhythms. Organelles, as the smallest independent sub-cell unit, are important contributors to cell metabolism and drug metabolism, collectively designated intracellular metabolism. However, disruption of intracellular spatiotemporal metabolism can lead to disease development and progression, as well as drug treatment interference. In this review, we systematically discuss spatiotemporal metabolism in cells and cell subpopulations. In particular, we focused on metabolism compartmentalization and physiological rhythms, including the variation and regulation of metabolic enzymes, metabolic pathways, and metabolites. Additionally, the intricate relationship among intracellular spatiotemporal metabolism, metabolism-related diseases, and drug therapy/toxicity has been discussed. Finally, approaches and strategies for intracellular spatiotemporal metabolism analysis and potential target identification are introduced, along with examples of potential new drug design based on this.
Collapse
Affiliation(s)
- Jingwei Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Zhixia Qiu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yongjie Zhang
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing, China; Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK-PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing, China.
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism & Pharmacokinetics, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
12
|
Song R, Hu M, Qin X, Qiu L, Wang P, Zhang X, Liu R, Wang X. The Roles of Lipid Metabolism in the Pathogenesis of Chronic Diseases in the Elderly. Nutrients 2023; 15:3433. [PMID: 37571370 PMCID: PMC10420821 DOI: 10.3390/nu15153433] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Lipid metabolism plays crucial roles in cellular processes such as hormone synthesis, energy production, and fat storage. Older adults are at risk of the dysregulation of lipid metabolism, which is associated with progressive declines in the physiological function of various organs. With advancing age, digestion and absorption commonly change, thereby resulting in decreased nutrient uptake. However, in the elderly population, the accumulation of excess fat becomes more pronounced due to a decline in the body's capacity to utilize lipids effectively. This is characterized by enhanced adipocyte synthesis and reduced breakdown, along with diminished peripheral tissue utilization capacity. Excessive lipid accumulation in the body, which manifests as hyperlipidemia and accumulated visceral fat, is linked to several chronic lipid-related diseases, including cardiovascular disease, type 2 diabetes, obesity, and nonalcoholic fatty liver disease. This review provides a summary of the altered lipid metabolism during aging, including lipid digestion, absorption, anabolism, and catabolism, as well as their associations with age-related chronic diseases, which aids in developing nutritional interventions for older adults to prevent or alleviate age-related chronic diseases.
Collapse
Affiliation(s)
- Rui Song
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (R.S.); (M.H.); (X.Q.); (L.Q.)
| | - Mengxiao Hu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (R.S.); (M.H.); (X.Q.); (L.Q.)
| | - Xiyu Qin
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (R.S.); (M.H.); (X.Q.); (L.Q.)
| | - Lili Qiu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (R.S.); (M.H.); (X.Q.); (L.Q.)
| | - Pengjie Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (P.W.); (X.Z.); (R.L.)
| | - Xiaoxu Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (P.W.); (X.Z.); (R.L.)
| | - Rong Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (P.W.); (X.Z.); (R.L.)
| | - Xiaoyu Wang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (R.S.); (M.H.); (X.Q.); (L.Q.)
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (P.W.); (X.Z.); (R.L.)
| |
Collapse
|
13
|
Abdrabouh AES. Toxicological and histopathological alterations in the heart of young and adult albino rats exposed to mosquito coil smoke. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:93070-93087. [PMID: 37501034 PMCID: PMC10447284 DOI: 10.1007/s11356-023-28812-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023]
Abstract
Mosquito coil repellents are well-known indoor air pollutant with significant health concerns. The present study investigated the toxic effects of mosquito coil smoke on the heart of young and adult male rats. The animals were subjected to the smoke for 6 h/day, 6 days/week, for 4 weeks. Within the first hour after lighting the coil, significant amounts of formaldehyde, total volatile organic compounds, and particulate matter (PM2.5 and PM10) were detected. Both exposed ages, particularly the young group, showed a significant increase in the activities of serum aspartate aminotransferase, lactate dehydrogenase, creatine kinase-MB, and the levels of troponin I, myoglobin, Na+ levels, lipid profile, and inflammatory markers (interleukin-6 and C-reactive protein) as well as a significant decrease in K+ levels and cardiac Na-K ATPase activity, indicating development of cardiac inflammation and dysfunction. Furthermore, the toxic stress response was validated by significant downregulation at expression of the detoxifying enzyme cytochrome p450. Histopathological studies in both age groups, especially the young group, revealed cardiomyocyte degeneration and necrotic areas. Moreover, upregulation at the pro-apoptotic markers, caspase3, P53, and cytochrome C expressions, was detected by immunohistochemical approach in heart sections of the exposed groups. Finally, the myocardial dysfunctional effects of the coil active ingredient, meperfluthrin, were confirmed by the docking results which indicated a high binding affinity of meperfluthrin, with Na-K ATPase and caspase 3. In conclusion, both the young and adult exposed groups experienced significant cardiac toxicity changes evidenced by cell apoptosis and histopathological alterations as well as disruption of biochemical indicators.
Collapse
|
14
|
Du X, Cai H, Jin N, Wu Z, Wang L, Wang Z, Xie B. Differences in the pharmacokinetics and steady-state blood concentrations of orally administered lenvatinib in adult and juvenile rats. Front Pharmacol 2023; 14:1140849. [PMID: 37576809 PMCID: PMC10420079 DOI: 10.3389/fphar.2023.1140849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023] Open
Abstract
Objective: The aim of this study was to compare the pharmacokinetics and steady-state serum concentrations of lenvatinib in adult and juvenile rats. Experimental study: An ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) method was developed to quantify lenvatinib in the serum and liver of rats. Six juvenile and six adult rats in each group were orally administered with a single dose of 7.0 mg/kg lenvatinib suspension for pharmacokinetics. Another 12 juvenile and adult rats were subjected to oral gavage with 7.0 mg/kg lenvatinib once daily for 5 days. Biofluild samples were pre-treated by protein precipitation and sorafenib was used as the internal standard for UPLC-MS analysis. The pharmacokinetic parameters were estimated by compartment and statistical model. The mRNA expression of CYP3A2 and SLC22A1 in liver of adult and juvenile rats was measured by real-time fluorescence quantitative PCR (RT-qPCR). Results: The UPLC-MS method met the requirements for quantitative analysis of lenvatinib in serum and liver. The pharmacokinetic results showed that the mean retention time (MRT(0-∞)) was 19.64 ± 7.64 h and 126.38 ± 130.18 h, with AUC(0-∞) values of 3.97 ± 0.73 μg‧mL-1 h and 5.95 ± 2.27 μg mL-1 h in adult and juvenile rats, respectively. When comparing adult rats (0.35 ± 0.15 μg/mL) to juvenile rats, no significant differences were observed in steady-state serum lenvatinib (0.32 ± 0.11 μg/mL), but a noteworthy decrease to one-third of steady-state liver lenvatinib was observed after multiple oral doses of lenvatinib in juvenile rats. Additional findings revealed that the mRNA expression of CYP3A2 and SLC22A1 was notably increased by 6.86 and 14.67 times, respectively, in juvenile rats compared to adult rats. Conclusion: Juvenile rats exhibit lower levels of lenvatinib in the liver's steady-state, potentially due to the disparity in CYP3A2 mRNA expression. These results imply that the dosage of lenvatinib for pediatric patients may need to be augmented in order to attain the desired clinical outcome.
Collapse
Affiliation(s)
- Xiaoyue Du
- Jiaxing University Master Degree Cultivation Base, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Medical College of Jiaxing University, Key Laboratory of Medical Electronics and Digital Health of Zhejiang Province, Jiaxing University, Jiaxing, China
| | - Hongxin Cai
- Jiaxing University Master Degree Cultivation Base, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Medical College of Jiaxing University, Key Laboratory of Medical Electronics and Digital Health of Zhejiang Province, Jiaxing University, Jiaxing, China
| | - Nan Jin
- Medical College of Jiaxing University, Key Laboratory of Medical Electronics and Digital Health of Zhejiang Province, Jiaxing University, Jiaxing, China
| | - Zhiguo Wu
- Medical College of Jiaxing University, Key Laboratory of Medical Electronics and Digital Health of Zhejiang Province, Jiaxing University, Jiaxing, China
| | - Lele Wang
- Medical College of Jiaxing University, Key Laboratory of Medical Electronics and Digital Health of Zhejiang Province, Jiaxing University, Jiaxing, China
| | - Zeyu Wang
- Medical College of Jiaxing University, Key Laboratory of Medical Electronics and Digital Health of Zhejiang Province, Jiaxing University, Jiaxing, China
| | - Baogang Xie
- Jiaxing University Master Degree Cultivation Base, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Medical College of Jiaxing University, Key Laboratory of Medical Electronics and Digital Health of Zhejiang Province, Jiaxing University, Jiaxing, China
| |
Collapse
|
15
|
Chen JH, Lin IH, Hsueh TY, Dalley JW, Tsai TH. Pharmacokinetics and transplacental transfer of codeine and codeine metabolites from Papaver somniferum L. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115623. [PMID: 36007718 DOI: 10.1016/j.jep.2022.115623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/04/2022] [Accepted: 08/06/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Papaveris Pericarpium, which is the dried husk of Papaver somniferum L., has been used as a phytomedicine to relieve cough, diarrhea and pain. The alkaloid codeine contained therein via biotransformation converts to morphine and potentially produces addictive and toxic effects. Due to the healthy concern for a pregnant woman, our hypothesis is that codeine and its metabolites can penetrate the placental barrier to reach the foetus and amniotic fluid, and these processes may be modulated by the transporter. AIM OF THE STUDY Because codeine is also considered a prodrug of morphine, it has a good analgesic effect. It is often used by pregnant women but may expose the foetus to the risk of morphine harm. The aim of this study is to investigate the metabolic rate, distribution and transplacental transfer mechanism of codeine and its metabolites morphine and morphine-3-glucuronide (M3G) in pregnant rats and to assess the risk of medication for pregnant women. MATERIALS AND METHODS Ultrahigh-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) combined with a microdialysis system was developed to monitor codeine, morphine and M3G in multiple sites of maternal blood, placenta, foetus and amniotic fluid after codeine administration. A compartmental model was used to calculate the pharmacokinetic parameters of codeine in blood after codeine administration (10 mg/kg, i.v.). The area under the concentration (AUC) ratio of AUCmetabolite/AUCcodeine and AUCtissue/AUCblood was used to represent the metabolic biotransformation ratio and the drug from blood-to-tissue transfer ratio, respectively. RESULTS The pharmacokinetic results demonstrated that codeine fit well with a two-compartment model and went through rapid metabolism to morphine and M3G in pregnant rats after codeine administration (10 mg/kg, i.v.). The biotransformation ratios of AUCmorphine/AUCcodeine, AUCM3G/AUCmorphine and AUCM3G/AUCcodeine were 0.12 ± 0.03, 54.45 ± 20.61 and 6.53 ± 2.47, respectively, after codeine administration (10 mg/kg, i.v.), which suggested that codeine was easily metabolized into M3G through morphine. The tissue distribution results demonstrated that all of the analytes penetrated into the foetus through the placenta; however, the blood-to-tissue transfer ratio (AUCtissue/AUCblood) of morphine and M3G was relatively lower than that of codeine after codeine administration (10 mg/kg, i.v.), which suggested that the blood-placenta barrier blocks the penetration of morphine and M3G into the foetus. Thus, the tissue transfer of morphine in the placenta and foetus was significantly enhanced by treatment with corticosterone, an inhibitor of organic cation transporter (OCT). CONCLUSION Based on microdialysis coupled to a validated UHPLC-MS/MS system, the pharmacokinetics and metabolic biotransformation of codeine and its metabolites were analyzed and clarified. The potential mechanism of morphine placental transfer was modulated by OCT transporters.
Collapse
Affiliation(s)
- Jung-Hung Chen
- Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - I-Hsin Lin
- Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Thomas Y Hsueh
- Division of Urology, Department of Surgery, Taipei City Hospital Renai Branch, Taipei, 106, Taiwan; Department of Urology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Jeffrey W Dalley
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK; Department of Psychiatry, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan; Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK.
| |
Collapse
|
16
|
ROS: Basic Concepts, Sources, Cellular Signaling, and its Implications in Aging Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1225578. [PMID: 36312897 PMCID: PMC9605829 DOI: 10.1155/2022/1225578] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022]
Abstract
Reactive oxygen species (ROS) are bioproducts of cellular metabolism. There is a range of molecules with oxidizing properties known as ROS. Despite those molecules being implied negatively in aging and numerous diseases, their key role in cellular signaling is evident. ROS control several biological processes such as inflammation, proliferation, and cell death. The redox signaling underlying these cellular events is one characteristic of the new generation of scientists aimed at defining the role of ROS in the cellular environment. The control of redox potential, which includes the balance of the sources of ROS and the antioxidant system, implies an important target for understanding the cells' fate derived from redox signaling. In this review, we summarized the chemical, the redox balance, the signaling, and the implications of ROS in biological aging.
Collapse
|
17
|
Carberry CK, Ferguson SS, Beltran AS, Fry RC, Rager JE. Using liver models generated from human-induced pluripotent stem cells (iPSCs) for evaluating chemical-induced modifications and disease across liver developmental stages. Toxicol In Vitro 2022; 83:105412. [PMID: 35688329 PMCID: PMC9296547 DOI: 10.1016/j.tiv.2022.105412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/20/2022] [Accepted: 06/03/2022] [Indexed: 01/09/2023]
Abstract
The liver is a pivotal organ regulating critical developmental stages of fetal metabolism and detoxification. Though numerous studies have evaluated links between prenatal/perinatal exposures and adverse health outcomes in the developing fetus, the central role of liver to health disruptions resulting from these exposures remains understudied, especially concerning early development and later-in-life health outcomes. While numerous in vitro methods for evaluating liver toxicity have been established, the use of iPSC-derived hepatocytes appears to be particularly well suited to contribute to this critical research gap due to their potential to model a diverse range of disease phenotypes and different stages of liver development. The following key aspects are reviewed: (1) an introduction to developmental liver toxicity; (2) an introduction to embryonic and induced pluripotent stem cell models; (3) methods and challenges for deriving liver cells from stem cells; and (4) applications for iPSC-derived hepatocytes to evaluate liver developmental stages and their associated responses to insults. We conclude that iPSC-derived hepatocytes have great potential for informing liver toxicity and underlying disease mechanisms via the generation of patient-specific iPSCs; implementing large-scale drug and chemical screening; evaluating general biological responses as a potential surrogate target cell; and evaluating inter-individual disease susceptibility and response variability.
Collapse
Affiliation(s)
- Celeste K Carberry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephen S Ferguson
- Biomolecular Screening Branch, National Toxicology Program, Research Triangle Park, NC, USA
| | - Adriana S Beltran
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Julia E Rager
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Curriculum in Toxicology and Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
18
|
Satomoto K, Suzuki I, Mita K, Wakita A, Yamagata H, Mitsumoto T, Hamada S. The effect of aging on the repeated-dose liver micronucleus assay using diethylnitrosamine. GENES AND ENVIRONMENT : THE OFFICIAL JOURNAL OF THE JAPANESE ENVIRONMENTAL MUTAGEN SOCIETY 2022; 44:21. [PMID: 35982479 PMCID: PMC9387043 DOI: 10.1186/s41021-022-00250-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 08/02/2022] [Indexed: 11/22/2022]
Abstract
Background The repeated-dose liver micronucleus (RDLMN) assay has been well-developed and applied because of its simplicity and the ease of integration into general toxicity studies which is the preferred method from the 3R’s point of view. In this assay, we observed micronucleated hepatocytes which accumulated during a rather long-term dosing period. When considering integration into general toxicity studies, the effects of age of the animals used in the micronucleus assay becomes a major issue. The effect of age on the micronucleus induction rate has been reported in bone marrow micronucleus assays, and it is considered that the decrease in cell proliferation rate due to aging is the cause of the decrease in sensitivity. A decrease in sensitivity due to aging was also reported in a liver micronucleus assay using clofibrate and the cause is considered to be a decrease in hepatocyte proliferation activity due to aging. However, no actual decrease in hepatocyte proliferation rate due to aging has been reported. In addition, there are no reports, so far, on whether similar effects of aging appear when other substances were administered. To investigate the effects of aging in the RDLMN assay, this study focused on the effects of 14-day repeated administration of DEN, a well-known genotoxic hepatocarcinogen with the hepatocyte toxicity which should cause an elevation of cell proliferation rate as a reflective regeneration. Results The liver micronuclei induced by DEN were equivalent between the two age groups (i.e., six and eight weeks of age at the start of dosing). In the histopathological examination for the liver, single cell necrosis, karyomegaly, and increased mitosis were observed in the hepatocytes, and the frequency and severity were increased dose-dependently. Ki-67 immunohistochemical analysis which can detect all cells in the cell cycle other than those in the G0 phase revealed dose-dependent increase of cell proliferation activity, and the difference between ages was not observed. Conclusion The effect of aging on the RDLMN assay could not be recognized when DEN was administered for 14 days in rats. Meanwhile, it was supported by the histopathological examination and Ki-67 immunohistochemical analysis that such an effect of aging was masked by the compensatory hepatocyte proliferation which was induced by the hepatocyte toxicity of DEN.
Collapse
Affiliation(s)
- Kensuke Satomoto
- Gotemba Laboratory, BoZo Research Center Inc, 1284 Kamado, Gotemba-shi, Shizuoka, 412-0039, Japan.
| | - Isamu Suzuki
- Gotemba Laboratory, BoZo Research Center Inc, 1284 Kamado, Gotemba-shi, Shizuoka, 412-0039, Japan
| | - Koji Mita
- Gotemba Laboratory, BoZo Research Center Inc, 1284 Kamado, Gotemba-shi, Shizuoka, 412-0039, Japan
| | - Atsushi Wakita
- Gotemba Laboratory, BoZo Research Center Inc, 1284 Kamado, Gotemba-shi, Shizuoka, 412-0039, Japan
| | - Hiroshi Yamagata
- Gotemba Laboratory, BoZo Research Center Inc, 1284 Kamado, Gotemba-shi, Shizuoka, 412-0039, Japan
| | - Tatsuya Mitsumoto
- Gotemba Laboratory, BoZo Research Center Inc, 1284 Kamado, Gotemba-shi, Shizuoka, 412-0039, Japan
| | - Shuichi Hamada
- Gotemba Laboratory, BoZo Research Center Inc, 1284 Kamado, Gotemba-shi, Shizuoka, 412-0039, Japan
| |
Collapse
|
19
|
Ko PJ, Milad MA, Radulovic LL, Gibson D. Pharmacokinetics of Levonorgestrel in Rat and Minipig and Pharmacokinetics of Etonogestrel in Rat Following Various Administration Routes. Xenobiotica 2022; 52:575-582. [PMID: 35975955 DOI: 10.1080/00498254.2022.2079023] [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: 10/15/2022]
Abstract
1. The objective of these studies was to determine the pharmacokinetics of levonorgestrel and etonogestrel in Sprague-Dawley rat or Göttingen minipig following various administration routes.2. Four sequential crossover studies were conducted: Study 1 administered levonorgestrel 30 µg intravenously and intradermally in four minipigs; Study 2 administered levonorgestrel 30 µg intravenously in 12 rats; Study 3 administered levonorgestrel 60 µg intravenously and subcutaneously in 12 rats; and Study 4 administered etonogestrel 30 µg intravenously in 12 rats. Samples were quantified using liquid chromatography-tandem mass spectrometry and pharmacokinetic parameters were estimated via noncompartmental analysis.3. Cmax and AUCinf for etonogestrel and levonorgestrel were similar following 30 µg intravenous bolus in rat, suggesting comparable pharmacokinetics. Levonorgestrel exposure was dose-proportional in rats, based on two-fold higher AUCinf following levonorgestrel 60 versus 30 µg. Bioavailability of intradermal and subcutaneous levonorgestrel was 97.7% (Study 1) and 90.3% (Study 3), respectively. The minipig levonorgestrel clearance was 21.5 L/hr, which was about 10-fold higher than both the rat levonorgestrel (range: 0.985 to 1.45 L/hr) and etonogestrel clearance (range: 0.803 to 0.968 L/hr).4. These studies contribute to the gap in knowledge of nonclinical levonorgestrel and etonogestrel pharmacokinetics, which is necessary for ongoing development of long-acting reversible contraceptives.
Collapse
Affiliation(s)
- Paul J Ko
- Milad Pharmaceutical Consulting LLC, Plymouth, Michigan, United States
| | - Mark A Milad
- Milad Pharmaceutical Consulting LLC, Plymouth, Michigan, United States
| | - Louis L Radulovic
- Innovative Pharma Consulting LLC, Superior Township, Michigan, United States
| | - Don Gibson
- DMG III Pharma Project Management Consulting LLC, East Lyme, Connecticut, United States
| |
Collapse
|
20
|
Jiang Y, Qin L, Lu A, Chen J, Wang J, Zhang Q, Lu Y, Gong Q, Gao J, Ma H, Tan D, He Y. CYP3A1 metabolism-based neurotoxicity of strychnine in rat. Toxicology 2022; 471:153156. [PMID: 35358606 DOI: 10.1016/j.tox.2022.153156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 10/18/2022]
Abstract
Strychnine is one of the main bioactive and toxic constituents of Semen Strychni. In the present study, the neurotoxic effects of strychnine, and the role of individual differences in metabolism on susceptibility to neurotoxicity of strychnine were investigated. The acute toxicity was observed by a single dose of strychnine (2.92 mg/kg, i.g.) in rats, the epileptic stages of rats were scored according to Racine's scale. The neurotoxicity of strychnine was evaluated by the levels of ROS, MDA, SOD and GSH in hippocampus, striatum, and cortex tissues measurements and histopathological analysis. The concentrations of strychnine in the plasma, hippocampus, striatum, and cortex tissues were determined using high performance liquid chromatography tandem mass spectrometry (LC-MS/MS). The expressions of the cytochrome P450, which is the most critical protein family involved in drugs metabolism, were detected by proteomics. The mechanism of susceptibility to neurotoxicity of strychnine was elucidated by correlation analysis among above indicators. The results indicated that striatum and cortex were the main toxic targets of strychnine, and the CYP3A1 might be a susceptible biomarker to neurotoxicity of strychnine. These results provide valuable insights into the neurotoxic susceptibility of strychnine that will aid in the rational clinical use of strychnine (possibly including Semen Strychni).
Collapse
Affiliation(s)
- Yuan Jiang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Lin Qin
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Anjing Lu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Juan Chen
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jianmei Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Qianru Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Yanliu Lu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Qihai Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jianmei Gao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Hong Ma
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, China
| | - Daopeng Tan
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Yuqi He
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
21
|
Wu Y, Li M, Guo Y, Liu T, Zhong L, Huang C, Ye C, Liu Q, Ren Z, Wang Y. The Effects of AT-533 and AT-533 gel on Liver Cytochrome P450 Enzymes in Rats. Eur J Drug Metab Pharmacokinet 2022; 47:345-352. [PMID: 35137361 DOI: 10.1007/s13318-022-00757-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND OBJECTIVES AT-533 is a novel heat shock protein 90 inhibitor, which exhibits various biological activities in vitro and in vivo. Cytochrome P450 (CYP) enzymes in the liver are involved in the biotransformation of drugs and considered to be essential indicators of liver toxicity. The aim of this study was to assess the effect of AT-533, either as active pharmaceutical ingredient or in gel form, on liver CYP enzymes. METHODS The effect of AT-533 or AT-533 gel on rat liver cytochrome P450 enzymes, including CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4, was analyzed using LC-MS/MS. RESULTS AT-533 and AT-533 gel did not significantly increase or reduce the enzymatic activity of CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4 at any treatment dose. CONCLUSIONS AT-533 and AT-533 gel did not have any effect on CYP activity and may be considered safe for external use in gel form, as an alternative to conventional treatment.
Collapse
Affiliation(s)
- Yanting Wu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, No. 601, Whampoa Road West, Guangzhou, 510632, People's Republic of China.,Department of Cell Biology, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, People's Republic of China
| | - Menghe Li
- Department of Cell Biology, College of Life Science and Technology, Jinan University, No. 601, Whampoa Road West, Guangzhou, 510632, People's Republic of China.,Department of Cell Biology, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, People's Republic of China
| | - Yuying Guo
- Department of Cell Biology, Guangzhou Jinan Biomedicine Research and Development Center Co. Ltd, Guangzhou, People's Republic of China
| | - Tao Liu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, No. 601, Whampoa Road West, Guangzhou, 510632, People's Republic of China.,Department of Cell Biology, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, People's Republic of China
| | - Lishan Zhong
- Department of Cell Biology, College of Life Science and Technology, Jinan University, No. 601, Whampoa Road West, Guangzhou, 510632, People's Republic of China.,Department of Cell Biology, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, People's Republic of China
| | - Chen Huang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, No. 601, Whampoa Road West, Guangzhou, 510632, People's Republic of China.,Department of Cell Biology, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, People's Republic of China
| | - Cuifang Ye
- Department of Cell Biology, College of Life Science and Technology, Jinan University, No. 601, Whampoa Road West, Guangzhou, 510632, People's Republic of China.,Department of Cell Biology, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, People's Republic of China
| | - Qiuying Liu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, No. 601, Whampoa Road West, Guangzhou, 510632, People's Republic of China.,Department of Cell Biology, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, People's Republic of China.,Department of pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhe Ren
- Department of Cell Biology, College of Life Science and Technology, Jinan University, No. 601, Whampoa Road West, Guangzhou, 510632, People's Republic of China.,Department of Cell Biology, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, People's Republic of China
| | - Yifei Wang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, No. 601, Whampoa Road West, Guangzhou, 510632, People's Republic of China. .,Department of Cell Biology, Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, National Engineering Research Center of Genetic Medicine, Guangzhou, People's Republic of China. .,Department of Cell Biology, Guangzhou Jinan Biomedicine Research and Development Center Co. Ltd, Guangzhou, People's Republic of China.
| |
Collapse
|
22
|
A pilot study for the prediction of liver function related scores using breath biomarkers and machine learning. Sci Rep 2022; 12:2032. [PMID: 35132067 PMCID: PMC8821604 DOI: 10.1038/s41598-022-05808-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/13/2022] [Indexed: 02/07/2023] Open
Abstract
Volatile organic compounds (VOCs) present in exhaled breath can help in analysing biochemical processes in the human body. Liver diseases can be traced using VOCs as biomarkers for physiological and pathophysiological conditions. In this work, we propose non-invasive and quick breath monitoring approach for early detection and progress monitoring of liver diseases using Isoprene, Limonene, and Dimethyl sulphide (DMS) as potential biomarkers. A pilot study is performed to design a dataset that includes the biomarkers concentration analysed from the breath sample before and after study subjects performed an exercise. A machine learning approach is applied for the prediction of scores for liver function diagnosis. Four regression methods are performed to predict the clinical scores using breath biomarkers data as features set by the machine learning techniques. A significant difference was observed for isoprene concentration (p < 0.01) and for DMS concentration (p < 0.0001) between liver patients and healthy subject's breath sample. The R-square value between actual clinical score and predicted clinical score is found to be 0.78, 0.82, and 0.85 for CTP score, APRI score, and MELD score, respectively. Our results have shown a promising result with significant different breath profiles between liver patients and healthy volunteers. The use of machine learning for the prediction of scores is found very promising for use of breath biomarkers for liver function diagnosis.
Collapse
|
23
|
Haduch A, Danek PJ, Kuban W, Pukło R, Alenina N, Gołębiowska J, Popik P, Bader M, Daniel WA. Cytochrome P450 2D (CYP2D) enzyme dysfunction associated with aging and serotonin deficiency in the brain and liver of female Dark Agouti rats. Neurochem Int 2022; 152:105223. [PMID: 34780807 DOI: 10.1016/j.neuint.2021.105223] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022]
Abstract
Among the enzymes that support brain metabolism, cytochrome P450 (CYP) enzymes occupy an important place. These enzymes catalyze the biotransformation pathways of neuroactive endogenous substrates (neurosteroids, neurotransmitters) and are necessary for the detoxification processes. The aim of the present study was to assess changes in the CYP2D activity and protein level during the aging process and as a result of serotonin deficiency in the female brain. The CYP2D activity was measured in brain and liver microsomes of Dark Agouti wild type (WT) female rats (mature 15-week-old and senescent 18-month-old rats) and in tryptophan hydroxylase 2 (TPH2)-deficient senescent female rats. The CYP2D activity in mature WT Dark Agouti females was independent of the changing phases of the estrous cycle. In senescent WT females rats, the CYP2D activity and protein level were decreased in the cerebral cortex, hippocampus, cerebellum and liver, but increased in the brain stem. In the other examined structures (frontal cortex, hypothalamus, thalamus, striatum), the enzyme activity did not change. In aging TPH2-deficient females, the CYP2D activity and protein levels were decreased in the frontal cortex, hypothalamus and brain stem (activity only), remaining unchanged in other brain structures and liver, relative to senescent WT females. In summary, the aging process and TPH2 deficit affect the CYP2D activity and protein level in female rats, which may have a negative impact on the compensatory capacity of CYP2D in the synthesis of serotonin and dopamine in cerebral structures involved in cognitive and emotional functions. In the liver, the CYP2D-catalyzed drug metabolism may be diminished in elderly females. The results in female rats are compared with those obtained previously in males. It is concluded that aging and serotonin deficiency exert sex-dependent effects on brain CYP2D, which seem to be less favorable in females concerning CYP2D-mediated neurotransmitter synthesis, but beneficial regarding slower neurosteroid metabolism.
Collapse
Affiliation(s)
- Anna Haduch
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Przemysław J Danek
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Wojciech Kuban
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Renata Pukło
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Joanna Gołębiowska
- Department of Behavioral Neuroscience and Drug Development, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Piotr Popik
- Department of Behavioral Neuroscience and Drug Development, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany; Institute for Biology, University of Lübeck, Germany; Charité University Medicine, Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Władysława A Daniel
- Department of Pharmacokinetics and Drug Metabolism, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland.
| |
Collapse
|
24
|
Fernández-Eslava B, Alonso D, Alonso-Alvarez C. An age-related decline in the expression of a red carotenoid-based ornament in wild birds. Evolution 2021; 75:3142-3153. [PMID: 34643274 DOI: 10.1111/evo.14378] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 09/21/2021] [Accepted: 09/27/2021] [Indexed: 01/02/2023]
Abstract
The past decades have provided valuable information on how animals age in the wild. However, examples of male reproductive senescence are scarce. In particular, few studies have described an age-related decline in the expression of conspicuous traits influencing mating success. Red ornaments could be good candidates to detect this decline because their expression may depend on the availability of pigments (carotenoids) related to oxidative stress, the latter frequently linked to senescence. Furthermore, it has been argued that efficient mitochondrial metabolism is key to express red carotenoid-based ornaments, and mitochondrial dysfunction is usually associated with senescence. We studied the age-linked expression of a red carotenoid-based trait: the yellow-to-red plumage coloration of male common crossbills (Loxia curvirostra). This coloration has recently been experimentally related to mitochondrial function. Here, we analyzed longitudinal plumage coloration data obtained throughout 28 years in free-living birds. We detected an initial increase in redness during the first 2 years of life and a subsequent decline. The relationship between color and age was unrelated to within-individual body mass variability. As far as we know, this is the first demonstration of an age-related ketocarotenoid-based color decrease detected by simultaneously testing within- and between-individual variability in wild animals.
Collapse
Affiliation(s)
- B Fernández-Eslava
- Department of Environmental Biology, Universidad de Navarra, Pamplona, Spain
| | - D Alonso
- Department of Ornithology, Aranzadi Sciences Society, Donostia-S. Sebastián, Spain
| | - C Alonso-Alvarez
- Department of Evolutionary Ecology, Spanish National Research Council (CSIC), Madrid, Spain
| |
Collapse
|
25
|
Abstract
Significance: During aging, excessive production of reactive species in the liver leads to redox imbalance with consequent oxidative damage and impaired organ homeostasis. Nevertheless, slight amounts of reactive species may modulate several transcription factors, acting as second messengers and regulating specific signaling pathways. These redox-dependent alterations may impact the age-associated decline in liver regeneration. Recent Advances: In the last few decades, relevant findings related to redox alterations in the aging liver were investigated. Consistently, recent research broadened understanding of redox modifications and signaling related to liver regeneration. Other than reporting the effect of oxidative stress, epigenetic and post-translational modifications, as well as modulation of specific redox-sensitive cellular signaling, were described. Among them, the present review focuses on Wnt/β-catenin, the nuclear factor (erythroid-derived 2)-like 2 (NRF2), members of the Forkhead box O (FoxO) family, and the p53 tumor suppressor. Critical Issues: Even though alteration in redox homeostasis occurs both in aging and in impaired liver regeneration, the associative mechanisms are not clearly defined. Of note, antioxidants are not effective in slowing hepatic senescence, and do not clearly improve liver repopulation after hepatectomy or transplant in humans. Future Directions: Further investigations are needed to define mutual redox-dependent molecular pathways involved both in aging and in the decline of liver regeneration. Preclinical studies aimed at the characterization of these pathways would define possible therapeutic targets for human trials. Antioxid. Redox Signal. 35, 832-847.
Collapse
Affiliation(s)
- Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gianluigi Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
26
|
Pereska Z, Simonovska N, Babulovska A, Berat-Huseini A, Naumoski K, Kostadinoski K. Acute severe poisoning with disinfectant in senior aged patient-case report and overview of literature considering age influence on treatment decision in alcohol-based intoxications. SAGE Open Med Case Rep 2021; 9:2050313X211047717. [PMID: 34567557 PMCID: PMC8461117 DOI: 10.1177/2050313x211047717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 09/01/2021] [Indexed: 01/12/2023] Open
Abstract
We present our experiences in the first case of severe suicidal poisoning with 70% ethanol-disinfectant in North Macedonia, in an elderly patient with immunocompromising comorbidities. A 66-year-old unconscious woman was admitted at our clinic, with a history of seropositive rheumatoid arthritis treated with methotrexate. She was in a coma, without signs of serotonin syndrome, recurrent episodes of cardio-respiratory insufficiency under supportive treatment without invasive ventilation, metabolic acidosis, increased D-dimer 3254 ng/mL. The toxicology screening confirmed low benzodiazepines levels and alcoholaemia of 526 mg/dL (5.26 g/L), due to ingestion of 70% ethanol. Considering the decreased biotransformation in the elderly, immunocompromising comorbidities, reports of fatal outcome in poisoned elderly patients with disinfectants under standard fluids supportive protocol, haemodialysis was initiated, with registered associated hypercoagulability which resulted in complete stabilization after 48 h of admission. Treatment protocols of poisoning with ethanol-based disinfectant in the elderly should consider timely performing haemodialysis at lower alcoholaemia levels than recommended.
Collapse
Affiliation(s)
- Zanina Pereska
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| | - Natasha Simonovska
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| | - Aleksandra Babulovska
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| | - Afrodita Berat-Huseini
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| | - Kiril Naumoski
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| | - Kristin Kostadinoski
- Faculty of Medicine, Ss Cyril and Methodius University, University Clinic of Toxicology, Skopje, North Macedonia
| |
Collapse
|
27
|
Gilbert ME, O'Shaughnessy KL, Thomas SE, Riutta C, Wood CR, Smith A, Oshiro WO, Ford RL, Hotchkiss MG, Hassan I, Ford JL. Thyroid Disruptors: Extrathyroidal Sites of Chemical Action and Neurodevelopmental Outcome-An Examination Using Triclosan and Perfluorohexane Sulfonate. Toxicol Sci 2021; 183:195-213. [PMID: 34460931 PMCID: PMC9038230 DOI: 10.1093/toxsci/kfab080] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Many xenobiotics are identified as potential thyroid disruptors due to their action to reduce circulating levels of thyroid hormone, most notably thyroxine (T4). Developmental neurotoxicity is a primary concern for thyroid disrupting chemicals yet correlating the impact of chemically induced changes in serum T4 to perturbed brain development remains elusive. A number of thyroid-specific neurodevelopmental assays have been proposed, based largely on the model thyroid hormone synthesis inhibitor propylthiouracil (PTU). This study examined whether thyroid disrupting chemicals acting distinct from synthesis inhibition would result in the same alterations in brain as expected with PTU. The perfluoroalkyl substance perfluorohexane sulfonate (50 mg/kg/day) and the antimicrobial Triclosan (300 mg/kg/day) were administered to pregnant rats from gestational day 6 to postnatal day (PN) 21, and a number of PTU-defined assays for neurotoxicity evaluated. Both chemicals reduced serum T4 but did not increase thyroid stimulating hormone. Both chemicals increased expression of hepatic metabolism genes, while thyroid hormone-responsive genes in the liver, thyroid gland, and brain were largely unchanged. Brain tissue T4 was reduced in newborns, but despite persistent T4 reductions in serum, had recovered in the PN6 pup brain. Neither treatment resulted in a low dose PTU-like phenotype in either brain morphology or neurobehavior, raising questions for the interpretation of serum biomarkers in regulatory toxicology. They further suggest that reliance on serum hormones as prescriptive of specific neurodevelopmental outcomes may be too simplistic and to understand thyroid-mediated neurotoxicity we must expand our thinking beyond that which follows thyroid hormone synthesis inhibition.
Collapse
Affiliation(s)
- Mary E Gilbert
- Center for Public Health and Environmental Assessment, Public Health Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27709, USA
| | - Katherine L O'Shaughnessy
- Center for Public Health and Environmental Assessment, Public Health Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27709, USA
| | - Susan E Thomas
- Oak Ridge Institute for Science Education, Oak Ridge, Tennesse 37830, USA
| | - Cal Riutta
- Oak Ridge Institute for Science Education, Oak Ridge, Tennesse 37830, USA
| | - Carmen R Wood
- Center for Public Health and Environmental Assessment, Public Health Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27709, USA
| | - Alicia Smith
- Oak Ridge Institute for Science Education, Oak Ridge, Tennesse 37830, USA
| | - Wendy O Oshiro
- Center for Public Health and Environmental Assessment, Public Health Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27709, USA
| | - Richard L Ford
- Oak Ridge Institute for Science Education, Oak Ridge, Tennesse 37830, USA
| | - Michelle Gatien Hotchkiss
- Center for Public Health and Environmental Assessment, Public Health Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27709, USA
| | - Iman Hassan
- Center for Public Health and Environmental Assessment, Public Health Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27709, USA
| | - Jermaine L Ford
- Center for Computational Toxicology and Exposure, Chemical Characterization and Exposure Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
28
|
Abstract
Almost 50% of prescription drugs lack age-appropriate dosing guidelines and therefore are used "off-label." Only ~10% drugs prescribed to neonates and infants have been studied for safety or efficacy. Immaturity of drug metabolism in children is often associated with drug toxicity. This chapter summarizes data on the ontogeny of major human metabolizing enzymes involved in oxidation, reduction, hydrolysis, and conjugation of drugs. The ontogeny data of individual drug-metabolizing enzymes are important for accurate prediction of drug pharmacokinetics and toxicity in children. This information is critical for designing clinical studies to appropriately test pharmacological hypotheses and develop safer pediatric drugs, and to replace the long-standing practice of body weight- or surface area-normalized drug dosing. The application of ontogeny data in physiologically based pharmacokinetic model and regulatory submission are discussed.
Collapse
|
29
|
Du T, Sun R, Etim I, Zheng Z, Liang D, Hu M, Gao S. Age-and Region-Dependent Disposition of Raloxifene in Rats. Pharm Res 2021; 38:1357-1367. [PMID: 34322833 PMCID: PMC8452384 DOI: 10.1007/s11095-021-03084-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] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/15/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Raloxifene undergoes extensive glucuronidation in the gastrointestinal (GI) tract and the liver. However, the impact of age on raloxifene disposition has never been studied. The purpose of this paper is to determine glucuronidation and Pharmacokinetics (PK) profiles of raloxifene in rats at different ages. METHODS Raloxifene glucuronidation was characterized using S9 fractions prepared from different intestinal segments and the liver of F344 rats at 4-, 11-, and 28-week. PK studies were conducted to determine raloxifene oral bioavailability at different ages. Raloxifene and its glucuronides were quantified using LC-MS/MS. RESULTS Raloxifene-6-glucuronide and raloxifene-4'-glucuronide were detected as the major metabolites and the ratio of these two glucuronides were different ranging from 2.1 to 4.9 folds in the ileum, jejunum, liver, and duodenum, and from 14.5 to 50 folds in the colon. The clearances in the duodenum at 4-week for both two glucuronides were significantly lower than those at the other two ages. PK studies showed that the oral bioavailability of raloxifene is age dependent. The absolute oral bioavailability of raloxifene was 3.5-folds higher at 4-week compared to that at 11-weeks. When raloxifene was administered through IV bolus, its half-life was 5.9 ± 1.16 h and 3.7 ± 0.68 h at 11-and 4-week, respectively. CONCLUSION These findings suggested that raloxifene metabolism in the duodenum was significantly slower at young age in rats, which increased the oral bioavailability of raloxifene. At 11-week, enterohepatic recycling efficiency was higher than that of 4-week. Raloxifene's dose at different ages should be carefully considered.
Collapse
Affiliation(s)
- Ting Du
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, Texas, 77004, USA
| | - Rongjin Sun
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 4901 Calhoun Street, Houston, Texas, 77204, USA
| | - Imoh Etim
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, Texas, 77004, USA
| | - Zicong Zheng
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 4901 Calhoun Street, Houston, Texas, 77204, USA
| | - Dong Liang
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, Texas, 77004, USA
| | - Ming Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, The University of Houston, 4901 Calhoun Street, Houston, Texas, 77204, USA.
| | - Song Gao
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne St, Houston, Texas, 77004, USA.
| |
Collapse
|
30
|
Kozaczek M, Bottje W, Albataineh D, Hakkak R. Effects of Short- and Long-Term Soy Protein Feeding on Hepatic Cytochrome P450 Expression in Obese Nonalcoholic Fatty Liver Disease Rat Model. Front Nutr 2021; 8:699620. [PMID: 34262928 PMCID: PMC8273275 DOI: 10.3389/fnut.2021.699620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/03/2021] [Indexed: 12/16/2022] Open
Abstract
Obesity can lead to chronic health complications such as nonalcoholic fatty liver disease (NAFLD). NAFLD is characterized by lipid aggregation in the hepatocytes and inflammation of the liver tissue as a consequence that can contribute to the development of cirrhosis and hepatocellular carcinoma (HCC). Previously, we reported that feeding obese Zucker rats with soy protein isolate (SPI) can reduce liver steatosis when compared with a casein (CAS) diet as a control. However, the effects of SPI on cytochrome P450 (CYP) in an obese rat model are less known. In addition, there is a lack of information concerning the consumption of soy protein in adolescents and its effect in reducing the early onset of NAFLD in this group. Our main goal was to understand if the SPI diet had any impact on the hepatic CYP gene expression when compared with the CAS diet. For this purpose, we used the transcriptomic data obtained in a previous study in which liver samples were collected from obese rats after short-term (eight-week) and long-term (16-week) feeding of SPI (n = 8 per group). To analyze this RNAseq data, we used Ingenuity Pathway Analysis (IPA) software. Comparing short- vs long-term feeding revealed an increase in the number of downregulated CYP genes from three at 8 weeks of SPI diet to five at 16 weeks of the same diet (P ≤ 0.05). On the other hand, upregulated CYP gene numbers showed a small increase in the long-term SPI diet compared to the short-term SPI diet, from 14 genes at 8 weeks to 17 genes at 16 weeks (P ≤ 0.05). The observed changes may have an important role in the attenuation of liver steatosis.
Collapse
Affiliation(s)
- Melisa Kozaczek
- Department of Dietetics and Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Poultry Science and The Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| | - Walter Bottje
- Department of Poultry Science and The Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Diyana Albataineh
- Department of Poultry Science and The Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Reza Hakkak
- Department of Dietetics and Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, AR, United States
| |
Collapse
|
31
|
Serum and Erythrocyte Antioxidant Defense in Colorectal Cancer Patients during Early Postoperative Period: Potential Modifiers and Impact on Clinical Outcomes. Antioxidants (Basel) 2021; 10:antiox10070999. [PMID: 34201536 PMCID: PMC8300787 DOI: 10.3390/antiox10070999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022] Open
Abstract
A better understanding of antioxidant status, its modifiers, and its effect on clinical outcomes in patients undergoing colorectal cancer surgery is needed for effective antioxidant-based interventions. The objectives of this cohort study were: to determine baseline serum (total antioxidant capacity (TAC) and ferric reducing antioxidant power (FRAP); n = 72) and erythrocyte (superoxide dismutase (SOD) and glutathione peroxidase (GPx); n = 47) antioxidant capacity and time-course during the 72 h postoperative period, to identify potential modifiers, and to establish impact on clinical outcomes. Older patients with comorbidities had lower baseline FRAP. TAC was inversely and SOD directly correlated with inflammatory markers. Cancer pathology affected GPx (lower in advanced and more aggressive cancers) and SOD (higher in advanced cancers). Surgical intervention induced a transient increase in FRAP and TAC with greater FRAP elevation in older, obese patients with several comorbidities. SOD activity significantly increased while GPx non-significantly decreased between 8 and 24 h post-incision. Poorer health status was associated with an increase in SOD and a decrease in GPx at 72 h. Clinical manifestation of postoperative ileus was preceded by decreased TAC at 24 h and an increase in SOD between 8 and 24 h and anastomotic leak was manifested by diminished SOD at 72 h compared to activities at 8 and 24 h. The time-frame between 8 and 24 h post-incision might be the most critical regarding oxidant/antioxidant balance and therefore the best suited for antioxidant-based intervention.
Collapse
|
32
|
Alshinnawy AS, El-Sayed WM, Sayed AA, Salem AM, Taha AM. Telomerase activator-65 and pomegranate peel improved the health status of the liver in aged rats; multi-targets involved. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:842-850. [PMID: 34630962 PMCID: PMC8487600 DOI: 10.22038/ijbms.2021.56670.12655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/22/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVES This study was undertaken to investigate the efficacy of telomerase activator-65 (Ta-65) and pomegranate peel against aging-induced deteriorations in the liver. MATERIALS AND METHODS The rats were divided into four groups: control, aged, aged rats treated with Ta-65, and pomegranate orally for two months. RESULTS Aging significantly increased the serum levels of total protein, globulins, and protein carbonyl and reduced the insulin-like growth factor 1 (IGF-1). It also elevated the hepatic malondialdehyde and decreased the hepatic glutathione S-transferase (GST) activity. Aging elevated the expression of thioredoxin reductase1, telomerase reverse transcriptase, and cytochrome 3a1 in the liver; it increased the p53 protein level and elevated the activity of caspase-3 in the liver indicating the occurrence of apoptosis. The architecture of the liver deteriorated in the aged rats, as shown by both light and electron microscopy examinations. The liver of the aged rats had many apoptotic hepatocytes with shrunken nuclei. Many hepatocytes had dilated rough endoplasmic reticulum, many lysosomes, and many fat droplets. Administration of Ta-65 and pomegranate to the aged rats normalized most of the previous biochemical parameters and improved the liver architecture. CONCLUSION Ta-65 and pomegranate have anti-aging activity through targeting multiple cellular pathways. It is also noteworthy that Ta-65 was superior to pomegranate in its alleviative effects.
Collapse
Affiliation(s)
| | - Wael Mohamed El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, Egypt. 11566
| | - Ahmed AbdelAziz Sayed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt. 11566
- Children's Cancer Hospital 57357, Cairo, Egypt
| | - Ahmed Mohamed Salem
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt. 11566
| | - AlShaimaa Mohamed Taha
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt. 11566
| |
Collapse
|
33
|
Abd-Elhakim YM, Moselhy AAA, Aldhahrani A, Beheiry RR, Mohamed WAM, Soliman MM, Saffaf BA, M. El Deib M. Protective Effect of Curcumin against Sodium Salicylate-Induced Oxidative Kidney Damage, Nuclear Factor-Kappa Dysregulation, and Apoptotic Consequences in Rats. Antioxidants (Basel) 2021; 10:826. [PMID: 34064189 PMCID: PMC8224369 DOI: 10.3390/antiox10060826] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
This study examined the effect of sodium salicylates (SS), alone and in combination with curcumin (CUR), on kidney function and architecture in rats. Five rat groups were given 1 mL physiological saline/rat orally, 1 mL olive oil/rat orally, 50 mg CUR/kg bwt orally, 300 mg SS/kg bwt intraperitoneally, or CUR+SS for 15 days. The hematological indices, serum protein profile, serum electrolytes balance, oxidative stress, and lipid peroxidation of kidney tissues were assessed. The histopathological examination and immune expression of Caspase-3 and nuclear factor kappa (NF-κB) were conducted. The findings showed that SS injection induced nephrotoxic activity, including increased serum urea, creatinine, and uric acid levels. It also caused apparent pathological alterations with increased Caspase-3 and NF-κB immuno-expression. In addition, thrombocytopenia, leukocytosis, neutrophilia, hyponatremia, hypochloremia, hypocalcemia, and hypomagnesemia but not hyperkalemia and hyperphosphatemia were evident in SS-injected rats. Moreover, SS exposure increased serum α1 globulin, renal tissue malondialdehyde, and Caspase-3 levels but superoxide dismutase, glutathione peroxidase, and Bcl-2 levels declined. Meanwhile, CUR significantly counteracted the SS harmful impacts on kidneys but SS+CUR co-administration induced an anemic condition. Overall, CUR has an evident protective role against SS-induced renal damage, but the disturbed hematological alterations should be carefully taken into consideration in their combined use.
Collapse
Affiliation(s)
- Yasmina M. Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Attia A. A. Moselhy
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt;
| | - Adil Aldhahrani
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, Turabah 21995, Saudi Arabia; (A.A.); (M.M.S.)
| | - Rasha R. Beheiry
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt;
| | - Wafaa A. M. Mohamed
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt;
| | - Mohamed Mohamed Soliman
- Clinical Laboratory Sciences Department, Turabah University College, Taif University, Turabah 21995, Saudi Arabia; (A.A.); (M.M.S.)
| | - Bayan A. Saffaf
- Pharmacology Department, Faculty of Pharmacy, Future University, City of the Future 41639, Egypt;
| | - Maha M. El Deib
- Department of Biochemistry, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt;
| |
Collapse
|
34
|
Schwab N, Ju Y, Hazrati LN. Early onset senescence and cognitive impairment in a murine model of repeated mTBI. Acta Neuropathol Commun 2021; 9:82. [PMID: 33964983 PMCID: PMC8106230 DOI: 10.1186/s40478-021-01190-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/03/2021] [Indexed: 12/19/2022] Open
Abstract
Mild traumatic brain injury (mTBI) results in broad neurological symptoms and an increased risk of being diagnosed with a neurodegenerative disease later in life. While the immediate oxidative stress response and post-mortem pathology of the injured brain has been well studied, it remains unclear how early pathogenic changes may drive persistent symptoms and confer susceptibility to neurodegeneration. In this study we have used a mouse model of repeated mTBI (rmTBI) to identify early gene expression changes at 24 h or 7 days post-injury (7 dpi). At 24 h post-injury, gene expression of rmTBI mice shows activation of the DNA damage response (DDR) towards double strand DNA breaks, altered calcium and cell–cell signalling, and inhibition of cell death pathways. By 7 dpi, rmTBI mice had a gene expression signature consistent with induction of cellular senescence, activation of neurodegenerative processes, and inhibition of the DDR. At both timepoints gliosis, microgliosis, and axonal damage were evident in the absence of any gross lesion, and by 7 dpi rmTBI also mice had elevated levels of IL1β, p21, 53BP1, DNA2, and p53, supportive of DNA damage-induced cellular senescence. These gene expression changes reflect establishment of processes usually linked to brain aging and suggests that cellular senescence occurs early and most likely prior to the accumulation of toxic proteins. These molecular changes were accompanied by spatial learning and memory deficits in the Morris water maze. To conclude, we have identified DNA damage-induced cellular senescence as a repercussion of repeated mild traumatic brain injury which correlates with cognitive impairment. Pathways involved in senescence may represent viable treatment targets of post-concussive syndrome. Senescence has been proposed to promote neurodegeneration and appears as an effective target to prevent long-term complications of mTBI, such as chronic traumatic encephalopathy and other related neurodegenerative pathologies.
Collapse
|
35
|
van Groen BD, Nicolaï J, Kuik AC, Van Cruchten S, van Peer E, Smits A, Schmidt S, de Wildt SN, Allegaert K, De Schaepdrijver L, Annaert P, Badée J. Ontogeny of Hepatic Transporters and Drug-Metabolizing Enzymes in Humans and in Nonclinical Species. Pharmacol Rev 2021; 73:597-678. [PMID: 33608409 DOI: 10.1124/pharmrev.120.000071] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The liver represents a major eliminating and detoxifying organ, determining exposure to endogenous compounds, drugs, and other xenobiotics. Drug transporters (DTs) and drug-metabolizing enzymes (DMEs) are key determinants of disposition, efficacy, and toxicity of drugs. Changes in their mRNA and protein expression levels and associated functional activity between the perinatal period until adulthood impact drug disposition. However, high-resolution ontogeny profiles for hepatic DTs and DMEs in nonclinical species and humans are lacking. Meanwhile, increasing use of physiologically based pharmacokinetic (PBPK) models necessitates availability of underlying ontogeny profiles to reliably predict drug exposure in children. In addition, understanding of species similarities and differences in DT/DME ontogeny is crucial for selecting the most appropriate animal species when studying the impact of development on pharmacokinetics. Cross-species ontogeny mapping is also required for adequate translation of drug disposition data in developing nonclinical species to humans. This review presents a quantitative cross-species compilation of the ontogeny of DTs and DMEs relevant to hepatic drug disposition. A comprehensive literature search was conducted on PubMed Central: Tables and graphs (often after digitization) in original manuscripts were used to extract ontogeny data. Data from independent studies were standardized and normalized before being compiled in graphs and tables for further interpretation. New insights gained from these high-resolution ontogeny profiles will be indispensable to understand cross-species differences in maturation of hepatic DTs and DMEs. Integration of these ontogeny data into PBPK models will support improved predictions of pediatric hepatic drug disposition processes. SIGNIFICANCE STATEMENT: Hepatic drug transporters (DTs) and drug-metabolizing enzymes (DMEs) play pivotal roles in hepatic drug disposition. Developmental changes in expression levels and activities of these proteins drive age-dependent pharmacokinetics. This review compiles the currently available ontogeny profiles of DTs and DMEs expressed in livers of humans and nonclinical species, enabling robust interpretation of age-related changes in drug disposition and ultimately optimization of pediatric drug therapy.
Collapse
Affiliation(s)
- B D van Groen
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - J Nicolaï
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - A C Kuik
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - S Van Cruchten
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - E van Peer
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - A Smits
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - S Schmidt
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - S N de Wildt
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - K Allegaert
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - L De Schaepdrijver
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - P Annaert
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| | - J Badée
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands (B.D.v.G., K.A.); Development Science, UCB BioPharma SRL, Braine-l'Alleud, Belgium (J.N.); Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.C.K.); Department of Veterinary Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium (S.V.C.); Fendigo sa/nvbv, An Alivira Group Company, Brussels, Belgium (E.v.P.); Department of Development and Regeneration KU Leuven, Leuven, Belgium (A.S.); Neonatal intensive care unit, University Hospitals Leuven, Leuven, Belgium (A.S.); Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida (S.S.); Department of Pharmacology and Toxicology, Radboud Institute of Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (S.N.d.W.); Departments of Development and Regeneration and of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (K.A.); Department of Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, The Netherlands (K.A.); Nonclinical Safety, Janssen R&D, Beerse, Belgium (L.D.S.); Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium (P.A.); and Department of PK Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland (J.B.)
| |
Collapse
|
36
|
First assessment of POPs and cytochrome P450 expression in Cuvier's beaked whales (Ziphius cavirostris) skin biopsies from the Mediterranean Sea. Sci Rep 2020; 10:21891. [PMID: 33318545 PMCID: PMC7736872 DOI: 10.1038/s41598-020-78962-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022] Open
Abstract
The Cuvier's beaked whale (Ziphius cavirostris) is one of the least known cetacean species worldwide. The decreasing population trend and associated threats has led to the IUCN categorising the Mediterranean subpopulation as Vulnerable on the Red List of Threatened Species. This study aimed to investigate for the first time the ecotoxicological status of Cuvier's beaked whale in the NW Mediterranean Sea. The study sampled around the 20% of the individuals belonging to the Ligurian subpopulation, collecting skin biopsies from free-ranging specimens. The levels of polychlorinated biphenyl (PCBs), polybrominated diphenyl ethers (PBDEs) and induction of cytochrome's P450 (CYP1A1 and CYP2B isoforms) were evaluated. Results highlighted that the pattern of concentration for the target contaminants was PCBs > PBDEs and the accumulation values were linked to age and sex, with adult males showing significantly higher levels than juvenile. Concerns raised by the fact that 80% of the individuals had PCB levels above the toxicity threshold for negative physiological effects in marine mammals. Therefore, these findings shed light on this silent and serious threat never assessed in the Mediterranean Cuvier’s beaked whale population, indicating that anthropogenic pressures, including chemical pollution, may represent menaces for the conservation of this species in the Mediterranean Sea.
Collapse
|
37
|
Li X, Lu Y, Ou X, Zeng S, Wang Y, Qi X, Zhu L, Liu Z. Changes and sex- and age-related differences in the expression of drug metabolizing enzymes in a KRAS-mutant mouse model of lung cancer. PeerJ 2020; 8:e10182. [PMID: 33240601 PMCID: PMC7680056 DOI: 10.7717/peerj.10182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/23/2020] [Indexed: 01/16/2023] Open
Abstract
Background This study aimed to systematically profile the alterations and sex- and age-related differences in the drug metabolizing enzymes (DMEs) in a KRAS-mutant mouse model of lung cancer (KRAS mice). Methodology In this study, the LC-MS/MS approach and a probe substrate method were used to detect the alterations in 21 isoforms of DMEs, as well as the enzymatic activities of five isoforms, respectively. Western blotting was applied to study the protein expression of four related receptors. Results The proteins contents of CYP2C29 and CYP3A11, were significantly downregulated in the livers of male KRAS mice at 26 weeks (3.7- and 4.4-fold, respectively, p < 0.05). SULT1A1 and SULT1D1 were upregulated by 1.8- to 7.0- fold at 20 (p = 0.015 and 0.017, respectively) and 26 weeks (p = 0.055 and 0.031, respectively). There were positive correlations between protein expression and enzyme activity for CYP2E1, UGT1A9, SULT1A1 and SULT1D1 (r2 ≥ 0.5, p < 0.001). Western blotting analysis revealed the downregulation of AHR, FXR and PPARα protein expression in male KRAS mice at 26 weeks. For sex-related differences, CYP2E1 was male-predominant and UGT1A2 was female-predominant in the kidney. UGT1A1 and UGT1A5 expression was female-predominant, whereas UGT2B1 exhibited male-predominant expression in liver tissue. For the tissue distribution of DMEs, 21 subtypes of DMEs were all expressed in liver tissue. In the intestine, the expression levels of CYP2C29, CYP27A1, UGT1A2, 1A5, 1A6a, 1A9, 2B1, 2B5 and 2B36 were under the limitation of quantification. The subtypes of CYP7A1, 1B1, 2E1 and UGT1A1, 2A3, 2B34 were detected in kidney tissue. Conclusions This study, for the first time, unveils the variations and sex- and age-related differences in DMEs in C57 BL/6 (WT) mice and KRAS mice.
Collapse
Affiliation(s)
- Xiaoyan Li
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yiyan Lu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojun Ou
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sijing Zeng
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Wang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoxiao Qi
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lijun Zhu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| |
Collapse
|
38
|
Daujat-Chavanieu M, Gerbal-Chaloin S. Regulation of CAR and PXR Expression in Health and Disease. Cells 2020; 9:E2395. [PMID: 33142929 PMCID: PMC7692647 DOI: 10.3390/cells9112395] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/22/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Pregnane X receptor (PXR, NR1I2) and constitutive androstane receptor (CAR, NR1I3) are members of the nuclear receptor superfamily that mainly act as ligand-activated transcription factors. Their functions have long been associated with the regulation of drug metabolism and disposition, and it is now well established that they are implicated in physiological and pathological conditions. Considerable efforts have been made to understand the regulation of their activity by their cognate ligand; however, additional regulatory mechanisms, among which the regulation of their expression, modulate their pleiotropic effects. This review summarizes the current knowledge on CAR and PXR expression during development and adult life; tissue distribution; spatial, temporal, and metabolic regulations; as well as in pathological situations, including chronic diseases and cancers. The expression of CAR and PXR is modulated by complex regulatory mechanisms that involve the interplay of transcription factors and also post-transcriptional and epigenetic modifications. Moreover, many environmental stimuli affect CAR and PXR expression through mechanisms that have not been elucidated.
Collapse
Affiliation(s)
| | - Sabine Gerbal-Chaloin
- IRMB, University of Montpellier, INSERM, CHU Montpellier, 34295 Montpellier, France;
| |
Collapse
|
39
|
Walsh J, Gheorghe CE, Lyte JM, van de Wouw M, Boehme M, Dinan TG, Cryan JF, Griffin BT, Clarke G, Hyland NP. Gut microbiome-mediated modulation of hepatic cytochrome P450 and P-glycoprotein: impact of butyrate and fructo-oligosaccharide-inulin. J Pharm Pharmacol 2020; 72:1072-1081. [PMID: 32337713 DOI: 10.1111/jphp.13276] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/21/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Our objective was to demonstrate microbial regulation of hepatic genes implicated in drug metabolism and transport using germ-free (GF) mice and to explore the impact of a microbial metabolite, butyrate, and a prebiotic dietary intervention on hepatic gene expression in mice. METHODS Using reverse-transcriptase PCR, we investigated cytochrome P450 (CYP) and multidrug-resistance protein 1 (MDR1) expression in conventional, GF and colonised GF mice. To investigate the effects of butyrate, sodium butyrate (3 g/l) was administered for 21 days to conventional or GF mice. In the prebiotic study, young adult and middle-aged mice received diet enriched with 10% fructo-oligosaccharide (FOS)-inulin for 14 weeks. KEY FINDINGS Colonisation of GF animals normalised expression of Cyp3a11 and Mdr1b to conventional levels. Butyrate upregulated Cyp2b10 in conventional mice (P < 0.05) but overall did not induce widespread changes in hepatic genes. FOS-inulin increased Cyp3a13 expression and had the opposite effect on Mdr1a expression in young adult mice (P < 0.05). Age, on the other hand, influenced the prebiotic effect on Cyp2a4 expression (P < 0.01). CONCLUSION The expression of hepatic genes implicated in drug metabolism and transport displays sensitivity to the microbiome, microbiome-derived metabolites and a microbial-targeted intervention. Our study may provide the impetus to explore microbiota-targeted interventions in normalising host metabolic activity and reducing inter-individual variability in drug pharmacokinetics.
Collapse
Affiliation(s)
- Jacinta Walsh
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Cassandra E Gheorghe
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Joshua M Lyte
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Brendan T Griffin
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Niall P Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
40
|
Fu L, Zhou L, Geng S, Li M, Lu W, Lu Y, Feng Z, Zhou X. Catalpol coordinately regulates phase I and II detoxification enzymes of Triptolide through CAR and NRF2 pathways to reduce Triptolide-induced hepatotoxicity. Biomed Pharmacother 2020; 129:110379. [PMID: 32563148 DOI: 10.1016/j.biopha.2020.110379] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 06/03/2020] [Accepted: 06/07/2020] [Indexed: 01/17/2023] Open
Abstract
Triptolide (TP), as the main component of Tripterygium Wilfordii (TW), can induce obvious liver injury when exerting the therapeutic effect. However, in our previous study, Catalpol (CAT), the main active ingredient of Rehmannia Glutinosa (RG), was shown to increase the drug clearance rate of TP and to attenuate TP-induced hepatotoxicity. Thus the present study aims to address the roles of phase I and II metabolic enzymes and the nuclear receptors in the detoxification process of TP, to analyze the mechanism of CAT reducing hepatotoxicity. For this purpose, SD rats and human liver cell line L-02 and HepG2 cells were selected, and treated with TP or the combination of TP and CAT in our study. Then the effect of CAT on detoxification of TP was analyzed, and the roles of phase I metabolic enzymes cytochrome P450 3A2/4 (CYP3A2/4) and phase II metabolic enzyme UDP-glucuronosyltransferase 1A6 (UGT1A6) and their related nuclear receptor regulations were evaluated. It was found that TP inhibited the transcription of CYP3A2/4. And through the constitutive androstane receptor (CAR) pathway, CAT not only significantly changed this inhibition and increased the expression of CYP3A2/4 but also increased the expression of CYP2C9, both of which are phase I detoxification enzymes of TP. And with the gene-silenced experiment, it was confirmed that this regulation was CAR-dependent. We also found that CAT could continue to exert a certain protective effect after CAR was silenced, with UGT1A6, the phase II detoxification enzyme of TP, significantly induced. And this was closely related to the enhanced transcriptional regulation of the nuclear factor erythroid 2-related factor 2 (NRF2) pathway. In conclusion, our results reveal that CAT can induce TP's phase I detoxification enzymes CYP3A2/4 and CYP2C9 through the CAR pathway, and induce TP's phase II detoxification enzyme UGT1A6 via the NRF2 pathway when CAR is strongly inhibited. And this coordinate regulation of CAT may be an important source of the effect for CAT to increase TP metabolic conversion and reduce TP hepatotoxicity.
Collapse
Affiliation(s)
- Ling Fu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lingling Zhou
- Jiangsu Provincial Key Laboratory of Pharmacology and Safety Evaluation of Material Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shan Geng
- Jiangsu Provincial Key Laboratory of Pharmacology and Safety Evaluation of Material Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Sichuan Kelun Pharmaceutical CO., LTD., Chengdu 610071, China
| | - Ming Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Lu
- Jiangsu Provincial Key Laboratory of Pharmacology and Safety Evaluation of Material Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yan Lu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China; Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210001, China
| | - Zhe Feng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xueping Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
41
|
Walsh J, Olavarria-Ramirez L, Lach G, Boehme M, Dinan TG, Cryan JF, Griffin BT, Hyland NP, Clarke G. Impact of host and environmental factors on β-glucuronidase enzymatic activity: implications for gastrointestinal serotonin. Am J Physiol Gastrointest Liver Physiol 2020; 318:G816-G826. [PMID: 32146834 DOI: 10.1152/ajpgi.00026.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The gastrointestinal tract houses a reservoir of bacterial-derived enzymes that can directly catalyze the metabolism of drugs, dietary elements and endogenous molecules. Both host and environmental factors may influence this enzymatic activity, with the potential to dictate the availability of the biologically-active form of endogenous molecules in the gut and influence inter-individual variation in drug metabolism. We aimed to investigate the influence of the microbiota, and the modulation of its composition, on fecal enzymatic activity. Intrinsic factors related to the host, including age, sex and genetic background, were also explored. Fecalase, a cell-free extract of feces, was prepared and used in a colorimetric-based assay to quantify enzymatic activity. To demonstrate the functional effects of fecal enzymatic activity, we examined β-glucuronidase-mediated cleavage of serotonin β-d-glucuronide (5-HT-GLU) and the resultant production of free 5-HT by HPLC. As expected, β-glucuronidase and β-glucosidase activity were absent in germ-free mice. Enzymatic activity was significantly influenced by mouse strain and animal species. Sex and age significantly altered metabolic activity with implications for free 5-HT. β-Glucuronidase and β-glucosidase activity remained at reduced levels for nearly two weeks after cessation of antibiotic administration. This effect on fecalase corresponded to significantly lower 5-HT levels as compared with incubation with pre-antibiotic fecalase from the same mice. Dietary targeting of the microbiota using prebiotics did not alter β-glucuronidase or β-glucosidase activity. Our data demonstrate that multiple factors influence the activity of bacterial-derived enzymes which may have potential clinical implications for drug metabolism and the deconjugation of host-produced glucuronides in the gut.NEW & NOTEWORTHY This article explores a comprehensive range of host and environmental factors that introduce variability in the expression of bacterial-derived metabolic enzymes. Our results demonstrate that altered β-glucuronidase activity has implications for the bioavailability of luminal serotonin. The experimental approach employed, fecalase, provides a mechanistic basis and translational platform to further delineate the functional outputs of altered metabolic activity, and the associated physiological effects of microbiota-targeted interventions on host response to drugs and host-produced glucuronides.
Collapse
Affiliation(s)
- Jacinta Walsh
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Loreto Olavarria-Ramirez
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - Gilliard Lach
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Brendan T Griffin
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Niall P Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland.,Irish Centre for Maternal and Child Health Research (INFANT), University College Cork, Cork, Ireland
| |
Collapse
|
42
|
Morales-Prieto N, Huertas-Abril PV, López de Lerma N, Pacheco IL, Pérez J, Peinado R, Abril N. Pedro Ximenez sun-dried grape must: a dietary supplement for a healthy longevity. Food Funct 2020; 11:4387-4402. [DOI: 10.1039/d0fo00204f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Sun-dried Pedro Ximénez white grapes must (PXM) is a potent antioxidant that regularizes apoptosis, proliferation, and regeneration of the structure and the function of aged mice liver. PXM consumption contributes to a healthy aging process.
Collapse
Affiliation(s)
- Noelia Morales-Prieto
- Departamento de Bioquímica y Biología Molecular
- Campus de Excelencia Internacional Agroalimentario CeiA3
- Universidad de Córdoba
- 14071 Córdoba
- Spain
| | - Paula V. Huertas-Abril
- Departamento de Bioquímica y Biología Molecular
- Campus de Excelencia Internacional Agroalimentario CeiA3
- Universidad de Córdoba
- 14071 Córdoba
- Spain
| | | | - Isabel. L. Pacheco
- Departamento de Anatomía y Anatomía Patológica Comparadas. Facultad de Veterinaria. Universidad de Córdoba
- 14071 Córdoba
- Spain
| | - José Pérez
- Departamento de Anatomía y Anatomía Patológica Comparadas. Facultad de Veterinaria. Universidad de Córdoba
- 14071 Córdoba
- Spain
| | - Rafael Peinado
- Departamento de Química Agrícola
- Universidad de Córdoba
- 14071 Córdoba
- Spain
| | - Nieves Abril
- Departamento de Bioquímica y Biología Molecular
- Campus de Excelencia Internacional Agroalimentario CeiA3
- Universidad de Córdoba
- 14071 Córdoba
- Spain
| |
Collapse
|