1
|
Okyar A, Ozturk Civelek D, Akyel YK, Surme S, Pala Kara Z, Kavakli IH. The role of the circadian timing system on drug metabolism and detoxification: an update. Expert Opin Drug Metab Toxicol 2024; 20:503-517. [PMID: 38753451 DOI: 10.1080/17425255.2024.2356167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION The 24-hour variations in drug absorption, distribution, metabolism, and elimination, collectively known as pharmacokinetics, are fundamentally influenced by rhythmic physiological processes regulated by the molecular clock. Recent advances have elucidated the intricacies of the circadian timing system and the molecular interplay between biological clocks, enzymes and transporters in preclinical level. AREA COVERED Circadian rhythm of the drug metabolizing enzymes and carrier efflux functions possess a major role for drug metabolism and detoxification. The efflux and metabolism function of intestines and liver seems important. The investigations revealed that the ABC and SLC transporter families, along with cytochrome p-450 systems in the intestine, liver, and kidney, play a dominant role in the circadian detoxification of drugs. Additionally, the circadian control of efflux by the blood-brain barrier is also discussed. EXPERT OPINION The influence of the circadian timing system on drug pharmacokinetics significantly impacts the efficacy, adverse effects, and toxicity profiles of various drugs. Moreover, the emergence of sex-related circadian changes in the metabolism and detoxification processes has underscored the importance of considering gender-specific differences in drug tolerability and pharmacology. A better understanding of coupling between central clock and circadian metabolism/transport contributes to the development of more rational drug utilization and the implementation of chronotherapy applications.
Collapse
Affiliation(s)
- Alper Okyar
- Department of Pharmacology, Istanbul University Faculty of Pharmacy, Istanbul, Turkiye
| | - Dilek Ozturk Civelek
- Department of Pharmacology, Faculty of Pharmacy, Bezmialem Vakif University, Istanbul, Turkiye
| | - Yasemin Kubra Akyel
- Department of Medical Pharmacology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
- Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Saliha Surme
- Molecular Biology and Genetics, Koc University, Istanbul, Türkiye
- Chemical and Biological Engineering, Koc University, Istanbul, Türkiye
| | - Zeliha Pala Kara
- Department of Pharmacology, Istanbul University Faculty of Pharmacy, Istanbul, Turkiye
| | - I Halil Kavakli
- Molecular Biology and Genetics, Koc University, Istanbul, Türkiye
- Chemical and Biological Engineering, Koc University, Istanbul, Türkiye
| |
Collapse
|
2
|
Das JK, Banskota N, Candia J, Griswold ME, Orenduff M, de Cabo R, Corcoran DL, Das SK, De S, Huffman KM, Kraus VB, Kraus WE, Martin C, Racette SB, Redman LM, Schilling B, Belsky D, Ferrucci L. Calorie restriction modulates the transcription of genes related to stress response and longevity in human muscle: The CALERIE study. Aging Cell 2023; 22:e13963. [PMID: 37823711 PMCID: PMC10726900 DOI: 10.1111/acel.13963] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 10/13/2023] Open
Abstract
The lifespan extension induced by 40% caloric restriction (CR) in rodents is accompanied by postponement of disease, preservation of function, and increased stress resistance. Whether CR elicits the same physiological and molecular responses in humans remains mostly unexplored. In the CALERIE study, 12% CR for 2 years in healthy humans induced minor losses of muscle mass (leg lean mass) without changes of muscle strength, but mechanisms for muscle quality preservation remained unclear. We performed high-depth RNA-Seq (387-618 million paired reads) on human vastus lateralis muscle biopsies collected from the CALERIE participants at baseline, 12- and 24-month follow-up from the 90 CALERIE participants randomized to CR and "ad libitum" control. Using linear mixed effect model, we identified protein-coding genes and splicing variants whose expression was significantly changed in the CR group compared to controls, including genes related to proteostasis, circadian rhythm regulation, DNA repair, mitochondrial biogenesis, mRNA processing/splicing, FOXO3 metabolism, apoptosis, and inflammation. Changes in some of these biological pathways mediated part of the positive effect of CR on muscle quality. Differentially expressed splicing variants were associated with change in pathways shown to be affected by CR in model organisms. Two years of sustained CR in humans positively affected skeletal muscle quality, and impacted gene expression and splicing profiles of biological pathways affected by CR in model organisms, suggesting that attainable levels of CR in a lifestyle intervention can benefit muscle health in humans.
Collapse
Affiliation(s)
- Jayanta Kumar Das
- Longitudinal Studies Section, Translation Gerontology BranchNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Nirad Banskota
- Computational Biology and Genomics CoreNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Julián Candia
- Longitudinal Studies Section, Translation Gerontology BranchNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | | | - Melissa Orenduff
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Rafael de Cabo
- Translation Gerontology Branch, National Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - David L. Corcoran
- Department of GeneticsUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Sai Krupa Das
- Energy Metabolism, Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
| | - Supriyo De
- Computational Biology and Genomics CoreNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Kim Marie Huffman
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Virginia B. Kraus
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - William E. Kraus
- Duke Molecular Physiology Institute and Department of MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | - Corby K. Martin
- Pennington Biomedical Research CenterLouisiana State UniversityBaton RougeLouisianaUSA
| | - Susan B. Racette
- College of Health SolutionsArizona State UniversityPhoenixArizonaUSA
| | - Leanne M. Redman
- Pennington Biomedical Research CenterLouisiana State UniversityBaton RougeLouisianaUSA
| | | | - Daniel W. Belsky
- Department of Epidemiology & Butler Columbia Aging CenterColumbia University Mailman School of Public HealthNew York CityNew YorkUSA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translation Gerontology BranchNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| |
Collapse
|
3
|
Shu Y, Hassan F, Ostrowski MC, Mehta KD. Role of hepatic PKCβ in nutritional regulation of hepatic glycogen synthesis. JCI Insight 2021; 6:149023. [PMID: 34622807 PMCID: PMC8525638 DOI: 10.1172/jci.insight.149023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 08/12/2021] [Indexed: 01/12/2023] Open
Abstract
The signaling mechanisms by which dietary fat and cholesterol signals regulate central pathways of glucose homeostasis are not completely understood. By using a hepatocyte-specific PKCβ-deficient (PKCβHep-/-) mouse model, we demonstrated the role of hepatic PKCβ in slowing disposal of glucose overload by suppressing glycogenesis and increasing hepatic glucose output. PKCβHep-/- mice exhibited lower plasma glucose under the fed condition, modestly improved systemic glucose tolerance and mildly suppressed gluconeogenesis, increased hepatic glycogen accumulation and synthesis due to elevated glucokinase expression and activated glycogen synthase (GS), and suppressed glucose-6-phosphatase expression compared with controls. These events were independent of hepatic AKT/GSK-3α/β signaling and were accompanied by increased HNF-4α transactivation, reduced FoxO1 protein abundance, and elevated expression of GS targeting protein phosphatase 1 regulatory subunit 3C in the PKCβHep-/- liver compared with controls. The above data strongly imply that hepatic PKCβ deficiency causes hypoglycemia postprandially by promoting glucose phosphorylation via upregulating glucokinase and subsequently redirecting more glucose-6-phosphate to glycogen via activating GS. In summary, hepatic PKCβ has a unique and essential ability to induce a coordinated response that negatively affects glycogenesis at multiple levels under physiological postprandial conditions, thereby integrating nutritional fat intake with dysregulation of glucose homeostasis.
Collapse
Affiliation(s)
- Yaoling Shu
- Department of Biological Chemistry & Pharmacology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Faizule Hassan
- Department of Biological Chemistry & Pharmacology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Michael C Ostrowski
- Department of Biochemistry & Molecular Biology, Holling Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kamal D Mehta
- Department of Biological Chemistry & Pharmacology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.,Instacare Therapeutics, Dublin, Ohio, USA
| |
Collapse
|
4
|
Furlan-Magaril M, Ando-Kuri M, Arzate-Mejía RG, Morf J, Cairns J, Román-Figueroa A, Tenorio-Hernández L, Poot-Hernández AC, Andrews S, Várnai C, Virk B, Wingett SW, Fraser P. The global and promoter-centric 3D genome organization temporally resolved during a circadian cycle. Genome Biol 2021; 22:162. [PMID: 34099014 PMCID: PMC8185950 DOI: 10.1186/s13059-021-02374-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 05/05/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Circadian gene expression is essential for organisms to adjust their physiology and anticipate daily changes in the environment. The molecular mechanisms controlling circadian gene transcription are still under investigation. In particular, how chromatin conformation at different genomic scales and regulatory elements impact rhythmic gene expression has been poorly characterized. RESULTS Here we measure changes in the spatial chromatin conformation in mouse liver using genome-wide and promoter-capture Hi-C alongside daily oscillations in gene transcription. We find topologically associating domains harboring circadian genes that switch assignments between the transcriptionally active and inactive compartment at different hours of the day, while their boundaries stably maintain their structure over time. To study chromatin contacts of promoters at high resolution over time, we apply promoter capture Hi-C. We find circadian gene promoters displayed a maximal number of chromatin contacts at the time of their peak transcriptional output. Furthermore, circadian genes, as well as contacted and transcribed regulatory elements, reach maximal expression at the same timepoints. Anchor sites of circadian gene promoter loops are enriched in DNA binding sites for liver nuclear receptors and other transcription factors, some exclusively present in either rhythmic or stable contacts. Finally, by comparing the interaction profiles between core clock and output circadian genes, we show that core clock interactomes are more dynamic compared to output circadian genes. CONCLUSION Our results identify chromatin conformation dynamics at different scales that parallel oscillatory gene expression and characterize the repertoire of regulatory elements that control circadian gene transcription through rhythmic or stable chromatin configurations.
Collapse
Affiliation(s)
- Mayra Furlan-Magaril
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| | - Masami Ando-Kuri
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Rodrigo G Arzate-Mejía
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
- Laboratory of Neuroepigenetics, Medical Faculty of the University of Zurich and Department of Health Science and Technology of the Swiss Federal Institute of Technology, Neuroscience Center Zurich, Zurich, Switzerland
| | - Jörg Morf
- Nuclear Dynamics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, CB2 0AW, UK
| | - Jonathan Cairns
- Nuclear Dynamics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
| | - Abraham Román-Figueroa
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Luis Tenorio-Hernández
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - A César Poot-Hernández
- Unidad de Bioinformática y Manejo de Información, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Simon Andrews
- Bioinformatics Group, The Babraham Institute, Cambridge, CB22 3AT, UK
| | - Csilla Várnai
- Nuclear Dynamics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
- Centre for Computational Biology, University of Birmingham, Birmingham, B15 2FG, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, B15 2SY, UK
| | - Boo Virk
- Bioinformatics Group, The Babraham Institute, Cambridge, CB22 3AT, UK
| | - Steven W Wingett
- Bioinformatics Group, The Babraham Institute, Cambridge, CB22 3AT, UK
- Cell Biology Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| | - Peter Fraser
- Nuclear Dynamics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
5
|
Zhang T, Yu F, Guo L, Chen M, Yuan X, Wu B. Small Heterodimer Partner Regulates Circadian Cytochromes p450 and Drug-Induced Hepatotoxicity. Theranostics 2018; 8:5246-5258. [PMID: 30555544 PMCID: PMC6276094 DOI: 10.7150/thno.28676] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 09/24/2018] [Indexed: 01/01/2023] Open
Abstract
The role of small heterodimer partner (SHP) in regulation of xenobiotic detoxification remains elusive. Here, we uncover a critical role for SHP in circadian regulation of cytochromes P450 (CYPs) and drug-induced hepatotoxicity. Methods: The mRNA and protein levels of CYPs in the livers of wild-type and SHP-/- mice were measured by quantitative real-time polymerase chain reaction and Western blotting, respectively. Regulation of CYP by SHP was investigated using luciferase reporter, mobility shift, chromatin immunoprecipitation, and/or co-immunoprecipitation assays. Results: The circadian rhythmicities of xenobiotic-detoxifying CYP mRNAs and proteins were disrupted in SHP-deficient mice. Of note, SHP ablation up-regulated Cyp2c38 and Cyp2c39, whereas it down-regulated all other CYP genes. Moreover, SHP regulated the expression of CYP genes through different mechanisms. SHP repressed Lrh-1/Hnf4α to down-regulate Cyp2c38, E4bp4 to up-regulate Cyp2a5, Dec2/HNF1α axis to up-regulate Cyp1a2, Cyp2e1 and Cyp3a11, and Rev-erbα to up-regulate Cyp2b10, Cyp4a10 and Cyp4a14. Furthermore, SHP ablation sensitized mice to theophylline (or mitoxantrone)-induced toxicity. Higher level of toxicity was correlated with down-regulated metabolism and clearance of theophylline (or mitoxantrone). In contrast, SHP ablation blunted the circadian rhythmicity of acetaminophen-induced hepatotoxicity and alleviated the toxicity by down-regulating Cyp2e1-mediated metabolism and reducing formation of the toxic metabolite. Toxicity alleviation by SHP ablation was also observed for aflatoxin B1 due to reduced formation of the toxic epoxide metabolite. Conclusion: SHP participates in circadian regulation of CYP enzymes, thereby impacting xenobiotic metabolism and drug-induced hepatotoxicity.
Collapse
|
6
|
Zhang T, Jiang X, Xu M, Wang H, Sang X, Qin M, Bao P, Wang R, Zhang C, Lu H, Li Y, Ren J, Chang HC, Yan J, Sun Q, Xu J. Sleep and circadian abnormalities precede cognitive deficits in R521C FUS knockin rats. Neurobiol Aging 2018; 72:159-170. [PMID: 30273830 DOI: 10.1016/j.neurobiolaging.2018.08.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 12/13/2022]
Abstract
Mutations in fused in sarcoma (Fus) cause familial amyotrophic lateral sclerosis (ALS) and occasionally frontotemporal dementia. Here we report the establishment and characterization of a novel knockin (KI) rat model expressing a Fus point mutation (R521C) via CRISPR/Cas9. The mutant animals developed adult-onset learning and memory behavioral deficits, with reduced spine density in hippocampal neurons. Remarkably, sleep-wake cycle and circadian abnormalities preceded the onset of cognitive deficit. RNA-seq study further demonstrated altered expression of some key sleep and circadian regulators, such as orexin/hypocretin receptor type 2 and casein kinase 1 epsilon, in the mutant rats. Therefore, we have established a rodent model expressing physiological level of a pathogenic mutant FUS, and we found cognitive impairment as a main behavioral deficit at mid age. Furthermore, we have revealed a new role of FUS in sleep and circadian regulation and demonstrated that functional change in FUS could cause sleep-wake and circadian disturbance as early symptoms.
Collapse
Affiliation(s)
- Tao Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Shanghai, China
| | - Xin Jiang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Shanghai, China
| | - Min Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haifang Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiao Sang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Meiling Qin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Puhua Bao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ruiqi Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chenchen Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Huiping Lu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuzhuo Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of New Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hung-Chun Chang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jun Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Jin Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
7
|
Go MJ, Noh JR, Hwang JH, Kim KS, Choi DH, Lee JS, Kim YH, Lee CH. Small heterodimer partner deficiency exacerbates binge drinking‑induced liver injury via modulation of natural killer T cell and neutrophil infiltration. Mol Med Rep 2018; 17:4989-4998. [PMID: 29393499 PMCID: PMC5865959 DOI: 10.3892/mmr.2018.8505] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 01/03/2018] [Indexed: 12/16/2022] Open
Abstract
Binge drinking among alcohol consumers is a common occurrence, and may result in the development of numerous diseases, including liver disorders. It has previously been reported that natural killer T (NKT) cells induce alcohol‑associated liver injury by promoting neutrophil infiltration. In the present study, the role of the orphan nuclear receptor small heterodimer partner (SHP), which is encoded by the NR0B2 gene, in acute binge drinking‑induced liver injury was investigated. SHP‑knockout (KO) and wild‑type (WT) control mice were intragastrically administered single doses of alcohol. The plasma concentrations of alanine aminotransferase and aspartate aminotransferase in SHP‑KO mice following alcohol treatment were significantly increased compared with WT mice. However, results of oil red O staining and 2',7'‑dichlorodihydrofluorescein diacetate staining indicated that levels of acute binge drinking‑associated hepatic lipid accumulation and oxidative stress were not significantly different between WT and SHP‑KO alcohol‑treated mice. Notably, tumor necrosis factor‑α mRNA expression in the liver of SHP‑KO mice was significantly increased following alcohol administration, compared with WT mice. Furthermore, the mRNA expression levels of C‑C motif chemokine ligand 2, C‑X‑C motif chemokine ligand 2 and interleukin‑4, which are all potent chemoattractants of NKT cells, as well as neutrophil expression levels, were significantly increased in the livers of SHP‑KO mice compared with WT mice following alcohol administration, as determined by reverse transcription‑quantitative polymerase chain reaction and flow cytometry. Enhanced infiltration of NKT cells, determined by flow cytometry, was also demonstrated in the livers of SHP‑KO mice following alcohol administration, compared with WT mice. The results of the present study indicate that SHP may be involved in liver‑associated protective mechanisms, with regards to the attenuation of damage caused by acute binge drinking, via regulation of NKT cell and neutrophil migration to the liver. The modulation of SHP may be a novel therapeutic strategy for the treatment of acute binge drinking‑induced liver injury.
Collapse
Affiliation(s)
- Min-Jeong Go
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jung-Ran Noh
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jung Hwan Hwang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Dong-Hee Choi
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jong-Soo Lee
- Department of Veterinary Microbiology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Yong-Hoon Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| |
Collapse
|
8
|
Eggink HM, Oosterman JE, de Goede P, de Vries EM, Foppen E, Koehorst M, Groen AK, Boelen A, Romijn JA, la Fleur SE, Soeters MR, Kalsbeek A. Complex interaction between circadian rhythm and diet on bile acid homeostasis in male rats. Chronobiol Int 2017; 34:1339-1353. [PMID: 29028359 DOI: 10.1080/07420528.2017.1363226] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Desynchronization between the master clock in the brain, which is entrained by (day) light, and peripheral organ clocks, which are mainly entrained by food intake, may have negative effects on energy metabolism. Bile acid metabolism follows a clear day/night rhythm. We investigated whether in rats on a normal chow diet the daily rhythm of plasma bile acids and hepatic expression of bile acid metabolic genes is controlled by the light/dark cycle or the feeding/fasting rhythm. In addition, we investigated the effects of high caloric diets and time-restricted feeding on daily rhythms of plasma bile acids and hepatic genes involved in bile acid synthesis. In experiment 1 male Wistar rats were fed according to three different feeding paradigms: food was available ad libitum for 24 h (ad lib) or time-restricted for 10 h during the dark period (dark fed) or 10 h during the light period (light fed). To allow further metabolic phenotyping, we manipulated dietary macronutrient intake by providing rats with a chow diet, a free choice high-fat-high-sugar diet or a free choice high-fat (HF) diet. In experiment 2 rats were fed a normal chow diet, but food was either available in a 6-meals-a-day (6M) scheme or ad lib. During both experiments, we measured plasma bile acid levels and hepatic mRNA expression of genes involved in bile acid metabolism at eight different time points during 24 h. Time-restricted feeding enhanced the daily rhythm in plasma bile acid concentrations. Plasma bile acid concentrations are highest during fasting and dropped during the period of food intake with all diets. An HF-containing diet changed bile acid pool composition, but not the daily rhythmicity of plasma bile acid levels. Daily rhythms of hepatic Cyp7a1 and Cyp8b1 mRNA expression followed the hepatic molecular clock, whereas for Shp expression food intake was leading. Combining an HF diet with feeding in the light/inactive period annulled CYp7a1 and Cyp8b1 gene expression rhythms, whilst keeping that of Shp intact. In conclusion, plasma bile acids and key genes in bile acid biosynthesis are entrained by food intake as well as the hepatic molecular clock. Eating during the inactivity period induced changes in the plasma bile acid pool composition similar to those induced by HF feeding.
Collapse
Affiliation(s)
- Hannah M Eggink
- a Department Endocrinology and Metabolism , Academic Medical Centre, University of Amsterdam , Amsterdam , The Netherlands
| | - Johanneke E Oosterman
- b Laboratory of Endocrinology, Deptartment Clinical Chemistry, Academic Medical Centre , University of Amsterdam , Amsterdam , The Netherlands
| | - Paul de Goede
- b Laboratory of Endocrinology, Deptartment Clinical Chemistry, Academic Medical Centre , University of Amsterdam , Amsterdam , The Netherlands
| | - Emmely M de Vries
- c Department of Medicine , Academic Medical Centre, University of Amsterdam , Amsterdam , The Netherlands
| | - Ewout Foppen
- b Laboratory of Endocrinology, Deptartment Clinical Chemistry, Academic Medical Centre , University of Amsterdam , Amsterdam , The Netherlands
| | - Martijn Koehorst
- d Department of Laboratory Medicine , University of Groningen, University Medical Centre Groningen , Groningen , The Netherlands
| | - Albert K Groen
- d Department of Laboratory Medicine , University of Groningen, University Medical Centre Groningen , Groningen , The Netherlands.,e Department of Vascular Medicine, Academic Medical Centre , University of Amsterdam , Amsterdam , The Netherlands
| | - Anita Boelen
- b Laboratory of Endocrinology, Deptartment Clinical Chemistry, Academic Medical Centre , University of Amsterdam , Amsterdam , The Netherlands
| | - Johannes A Romijn
- c Department of Medicine , Academic Medical Centre, University of Amsterdam , Amsterdam , The Netherlands
| | - Susanne E la Fleur
- a Department Endocrinology and Metabolism , Academic Medical Centre, University of Amsterdam , Amsterdam , The Netherlands.,b Laboratory of Endocrinology, Deptartment Clinical Chemistry, Academic Medical Centre , University of Amsterdam , Amsterdam , The Netherlands.,f Metabolism and Reward , Netherlands Institute for Neuroscience , Amsterdam , the Netherlands
| | - Maarten R Soeters
- a Department Endocrinology and Metabolism , Academic Medical Centre, University of Amsterdam , Amsterdam , The Netherlands
| | - Andries Kalsbeek
- a Department Endocrinology and Metabolism , Academic Medical Centre, University of Amsterdam , Amsterdam , The Netherlands.,b Laboratory of Endocrinology, Deptartment Clinical Chemistry, Academic Medical Centre , University of Amsterdam , Amsterdam , The Netherlands.,g Hypothalamic Integration Mechanisms , Netherlands Institute for Neuroscience , Amsterdam , The Netherlands
| |
Collapse
|
9
|
Marczak MM, Yan B. Circadian rhythmicity: A functional connection between differentiated embryonic chondrocyte-1 (DEC1) and small heterodimer partner (SHP). Arch Biochem Biophys 2017; 631:11-18. [PMID: 28797635 DOI: 10.1016/j.abb.2017.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/04/2017] [Accepted: 08/05/2017] [Indexed: 12/26/2022]
Abstract
Circadian rhythm misalignment has been increasingly recognized to pose health risk for a wide range of diseases, particularly metabolic disorders. The liver maintains metabolic homeostasis and expresses many circadian genes, such as differentiated embryo chondrocyte-1 (DEC1) and small heterodimer partner (SHP). DEC1 is established to repress transcription through E-box elements, and SHP belongs to the superfamily of nuclear receptors and has multiple E-box elements in its promoter. Importantly, DEC1 and SHP are inversely oscillated. This study was performed to test the hypothesis that the SHP gene is a target gene of DEC1. Cotransfection demonstrated that DEC1 repressed the SHP promoter and attenuated the transactivation of the classic circadian activator complex of Clock/Bmal1. Site-directed mutagenesis, electrophoretic mobility shift assay and chromatin immunoprecipitation established that the repression was achieved through the E-box in the proximal promoter. Transfection of DEC1 suppressed the expression of SHP. In circadian-inducing cells, the epileptic agent valproate inversely altered the expression of DEC1 and SHP. Both DEC1 and SHP are involved in energy balance and valproate is known to induce hepatic steatosis. Our findings collectively establish that DEC1 participates in the negative loop of SHP oscillating expression with potential implications in metabolic homeostasis.
Collapse
Affiliation(s)
- Marek M Marczak
- Department of Biomedical and Pharmaceutical Sciences, Center for Integrated Drug Development, University of Rhode Island, Kingston, RI 02881, United States
| | - Bingfang Yan
- Department of Biomedical and Pharmaceutical Sciences, Center for Integrated Drug Development, University of Rhode Island, Kingston, RI 02881, United States.
| |
Collapse
|
10
|
Li WK, Li H, Lu YF, Li YY, Fu ZD, Liu J. Atorvastatin alters the expression of genes related to bile acid metabolism and circadian clock in livers of mice. PeerJ 2017; 5:e3348. [PMID: 28533986 PMCID: PMC5438592 DOI: 10.7717/peerj.3348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/23/2017] [Indexed: 02/06/2023] Open
Abstract
Aim Atorvastatin is a HMG-CoA reductase inhibitor used for hyperlipidemia. Atorvastatin is generally safe but may induce cholestasis. The present study aimed to examine the effects of atorvastatin on hepatic gene expression related to bile acid metabolism and homeostasis, as well as the expression of circadian clock genes in livers of mice. Methods Adult male mice were given atorvastatin (10, 30, and 100 mg/kg, po) daily for 30 days, and blood biochemistry, histopathology, and gene expression were examined. Results Repeated administration of atorvastatin did not affect animal body weight gain or liver weights. Serum enzyme activities were in the normal range. Histologically, the high dose of atorvastatin produced scattered swollen hepatocytes, foci of feathery-like degeneration, together with increased expression of Egr-1 and metallothionein-1. Atorvastatin increased the expression of Cyp7a1 in the liver, along with FXR and SHP. In contract, atorvastatin decreased the expression of bile acid transporters Ntcp, Bsep, Ostα, and Ostβ. The most dramatic change was the 30-fold induction of Cyp7a1. Because Cyp7a1 is a circadian clock-controlled gene, we further examined the effect of atorvastatin on clock gene expression. Atorvastatin increased the expression of clock core master genes Bmal1 and Npas2, decreased the expression of clock feedback genes Per2, Per3, and the clock targeted genes Dbp and Tef, whereas it had no effect on Cry1 and Nr1d1 expression. Conclusion Repeated administration of atorvastatin affects bile acid metabolism and markedly increases the expression of the bile acid synthesis rate-limiting enzyme gene Cyp7a1, together with alterations in the expression of circadian clock genes.
Collapse
Affiliation(s)
- Wen-Kai Li
- Key Lab for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical College, Zunyi, China.,Department of Pharmacology, Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Huan Li
- Key Lab for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical College, Zunyi, China
| | - Yuan-Fu Lu
- Key Lab for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical College, Zunyi, China
| | - Ying-Ying Li
- Key Lab for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical College, Zunyi, China
| | - Zidong Donna Fu
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States of America
| | - Jie Liu
- Key Lab for Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical College, Zunyi, China
| |
Collapse
|