1
|
Hao J, Hu R, Zhao J, Li Y, Li Q, Zhang X. Metabolomics combined with network pharmacology reveals the protective effect of astragaloside IV on alcoholic liver disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156032. [PMID: 39270570 DOI: 10.1016/j.phymed.2024.156032] [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: 04/23/2024] [Revised: 08/13/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Alcoholic liver disease (ALD) is a significant contributor to liver damage. However, the clinical options for the treatment of ALD are limited. Astragaloside IV (AST-IV) is a saponin isolated from Astragalus membranaceus (AM). This study aimed to explore the underlying mechanisms of action of AST-IV in ALD by integrating metabolomics and network pharmacology. METHODS Sprague-Dawley (SD) rats were used to establish a rat model of ALD. AST-IV and polyene phosphatidyl choline (PPC; a positive control drug) were administered to rats with ALD for 4 weeks. We measured the body weight, liver index, ALT, AST, TC, TG, inflammatory markers (IL-1β, IL-6, and TNF-α), and oxidative stress markers (SOD, MDA) and used H&E and ORO staining to evaluate the hepatoprotective effect of both AST-IV and PPC on ALD. Subsequently, we performed untargeted metabolomics to predict the influence of AST-IV on lipid metabolism in rats with ALD. We then used a network pharmacology approach to identify the core targets through which AST-IV corrected lipid metabolism disorders and validated these targets through molecular docking, qRT-PCR and western blot analyses. Finally, we calculated the relationships between ALD-related biochemical markers, differential liver metabolites, and core targets using Spearman's correlation analysis. RESULTS AST-IV improved pathological damage and reduced lipid accumulation in the hepatocytes of rats with ALD. Furthermore, AST-IV inhibited oxidative stress and inflammatory responses in rats with ALD. The metabolomic results showed that AST-IV corrected hepatic lipid metabolism disorders by targeting linoleic acid, necrosis, sphingolipid, and glycerophospholipid metabolism. The Network pharmacology analysis revealed that the core targets of AST-IV exerting the above effects were p-RIPK3, p-MLKL, CYP1A2, CYP2C19, PPARα, PCSK9. Spearman's correlation analysis showed a strong correlation between ALD-related serum biochemical indices, core targets, and liver differential metabolites. CONCLUSION AST-IV corrects the metabolic disorders of linoleic acid, sphingolipid, and glycerophospholipid, and alleviates necrosis in rats with ALD through the core targets p-RIPK3, p-MLKL, CYP1A2, CYP2C19, PPARα, and PCSK9. This study is the first to reveal the mechanism of ALD protection through AST-IV from the perspective of metabolomics and network pharmacology. Therefore, a novel target has been identified to exert protection against ALD. This study provides a reference for ALD treatment.
Collapse
Affiliation(s)
- Jinfang Hao
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Ruixian Hu
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China; Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Jianming Zhao
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China
| | - Yuanhong Li
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China
| | - Qingshan Li
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| | - Xiaoyan Zhang
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
2
|
Li YY, Madduri SS, Rezeli ET, Santos C, Freeman III H, Peng J, McRitchie SL, Pathmasiri W, Hursting SD, Sumner SJ, Stewart DA. Macronutrient-differential dietary pattern impacts on body weight, hepatic inflammation, and metabolism. Front Nutr 2024; 11:1356038. [PMID: 38868554 PMCID: PMC11168494 DOI: 10.3389/fnut.2024.1356038] [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: 12/14/2023] [Accepted: 04/24/2024] [Indexed: 06/14/2024] Open
Abstract
Introduction Obesity is a multi-factorial disease frequently associated with poor nutritional habits and linked to many detrimental health outcomes. Individuals with obesity are more likely to have increased levels of persistent inflammatory and metabolic dysregulation. The goal of this study was to compare four dietary patterns differentiated by macronutrient content in a postmenopausal model. Dietary patterns were high carbohydrate (HC), high fat (HF), high carbohydrate plus high fat (HCHF), and high protein (HP) with higher fiber. Methods Changes in body weight and glucose levels were measured in female, ovariectomized C57BL/6 mice after 15 weeks of feeding. One group of five mice fed the HCHF diet was crossed over to the HP diet on day 84, modeling a 21-day intervention. In a follow-up study comparing the HCHF versus HP dietary patterns, systemic changes in inflammation, using an 80-cytokine array and metabolism, by untargeted liquid chromatography-mass spectrometry (LCMS)-based metabolomics were evaluated. Results Only the HF and HCHF diets resulted in obesity, shown by significant differences in body weights compared to the HP diet. Body weight gains during the two-diet follow-up study were consistent with the four-diet study. On Day 105 of the 4-diet study, glucose levels were significantly lower for mice fed the HP diet than for those fed the HC and HF diets. Mice switched from the HCHF to the HP diet lost an average of 3.7 grams by the end of the 21-day intervention, but this corresponded with decreased food consumption. The HCHF pattern resulted in dramatic inflammatory dysregulation, as all 80 cytokines were elevated significantly in the livers of these mice after 15 weeks of HCHF diet exposure. Comparatively, only 32 markers changed significantly on the HP diet (24 up, 8 down). Metabolic perturbations in several endogenous biological pathways were also observed based on macronutrient differences and revealed dysfunction in several nutritionally relevant biosynthetic pathways. Conclusion Overall, the HCHF diet promoted detrimental impacts and changes linked to several diseases, including arthritis or breast neoplasms. Identification of dietary pattern-specific impacts in this model provides a means to monitor the effects of disease risk and test interventions to prevent poor health outcomes through nutritional modification.
Collapse
Affiliation(s)
- Yuan-yuan Li
- Metabolomics and Exposome Laboratory, Nutrition Research Institute, Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Supradeep S. Madduri
- Metabolomics and Exposome Laboratory, Nutrition Research Institute, Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Erika T. Rezeli
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Charlene Santos
- Animal Studies Core Lab, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Herman Freeman III
- Metabolomics and Exposome Laboratory, Nutrition Research Institute, Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| | - Jing Peng
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Susan L. McRitchie
- Metabolomics and Exposome Laboratory, Nutrition Research Institute, Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Wimal Pathmasiri
- Metabolomics and Exposome Laboratory, Nutrition Research Institute, Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Stephen D. Hursting
- Metabolomics and Exposome Laboratory, Nutrition Research Institute, Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Susan J. Sumner
- Metabolomics and Exposome Laboratory, Nutrition Research Institute, Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Delisha A. Stewart
- Metabolomics and Exposome Laboratory, Nutrition Research Institute, Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
3
|
Harrison SP, Baumgarten SF, Chollet ME, Stavik B, Bhattacharya A, Almaas R, Sullivan GJ. Parenteral nutrition emulsion inhibits CYP3A4 in an iPSC derived liver organoids testing platform. J Pediatr Gastroenterol Nutr 2024; 78:1047-1058. [PMID: 38529852 DOI: 10.1002/jpn3.12195] [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: 11/15/2022] [Revised: 02/14/2024] [Accepted: 02/28/2024] [Indexed: 03/27/2024]
Abstract
OBJECTIVES Parenteral nutrition (PN) is used for patients of varying ages with intestinal failure to supplement calories. Premature newborns with low birth weight are at a high risk for developing PN associated liver disease (PNALD) including steatosis, cholestasis, and gallbladder sludge/stones. To optimize nutrition regimens, models are required to predict PNALD. METHODS We have exploited induced pluripotent stem cell derived liver organoids to provide a testing platform for PNALD. Liver organoids mimic the developing liver and contain the different hepatic cell types. The organoids have an early postnatal maturity making them a suitable model for premature newborns. To mimic PN treatment we used medium supplemented with either clinoleic (80% olive oil/20% soybean oil) or intralipid (100% soybean oil) for 7 days. RESULTS Homogenous HNF4a staining was found in all organoids and PN treatments caused accumulation of lipids in hepatocytes. Organoids exhibited a dose dependent decrease in CYP3A4 activity and expression of hepatocyte functional genes. The lipid emulsions did not affect overall organoid viability and glucose levels had no contributory effect to the observed results. CONCLUSIONS Liver organoids could be utilized as a potential screening platform for the development of new, less hepatotoxic PN solutions. Both lipid treatments caused hepatic lipid accumulation, a significant decrease in CYP3A4 activity and a decrease in the RNA levels of both CYP3A4 and CYP1A2 in a dose dependent manner. The presence of high glucose had no additive effect, while Clinoleic at high dose, caused significant upregulation of interleukin 6 and TLR4 expression.
Collapse
Affiliation(s)
- Sean P Harrison
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Saphira F Baumgarten
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub-Center of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Research, Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Maria E Chollet
- Research, Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Benedicte Stavik
- Research, Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Anindita Bhattacharya
- Research, Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Runar Almaas
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gareth J Sullivan
- Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
4
|
Hazra S, Singh PA. Safety Aspects of Herb Interactions: Current Understanding and Future Prospects. Curr Drug Metab 2024; 25:28-53. [PMID: 38482621 DOI: 10.2174/0113892002289753240305062601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/11/2024] [Accepted: 02/09/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND The use of herbal medicines is on the rise throughout the world due to their perceived safety profile. However, incidences of herb-drug, herb-herb and herb-food interactions considering safety aspects have opened new arenas for discussion. OBJECTIVE The current study aims to provide comprehensive insights into the various types of herb interactions, the mechanisms involved, their assessment, and historical developments, keeping herbal safety at the central point of discussion. METHODS The authors undertook a focused/targeted literature review and collected data from various databases, including Science Direct, Wiley Online Library, Springer, PubMed, and Google Scholar. Conventional literature on herbal remedies, such as those by the WHO and other international or national organizations. RESULTS The article considered reviewing the regulations, interaction mechanisms, and detection of herb-herb, herb-drug and herb-food interactions in commonly used yet vital plants, including Glycyrrhiza glabra, Mentha piperita, Aloe barbadensis, Zingiber officinale, Gingko biloba, Withania somnifera, etc. The study found that healthcare professionals worry about patients not informing them about their herbal prescriptions (primarily used with conventional treatment), which can cause herb-drug/herb-food/herb-herb interactions. These interactions were caused by altered pharmacodynamic and pharmacokinetic processes, which might be explained using in-vivo, in-vitro, in-silico, pharmacogenomics, and pharmacogenetics. Nutrivigilance may be the greatest method to monitor herb-food interactions, but its adoption is limited worldwide. CONCLUSION This article can serve as a lead for clinicians, guiding them regarding herb-drug, herb-food, and herb-herb interactions induced by commonly consumed plant species. Patients may also be counseled to avoid conventional drugs, botanicals, and foods with a restricted therapeutic window.
Collapse
Affiliation(s)
- Subhajit Hazra
- University Institute of Pharma Sciences (UIPS), Chandigarh University, Mohali-140413, Punjab, India
| | - Preet Amol Singh
- University Institute of Pharma Sciences (UIPS), Chandigarh University, Mohali-140413, Punjab, India
| |
Collapse
|
5
|
Pedersen S, Kverneland M, Rudi K, Gervin K, Landmark CJ, Iversen PO, Selmer KK. Decreased serum concentrations of antiseizure medications in children with drug resistant epilepsy following treatment with ketogenic diet. Epilepsia Open 2023; 8:858-866. [PMID: 37057954 PMCID: PMC10472394 DOI: 10.1002/epi4.12746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/13/2023] [Indexed: 04/15/2023] Open
Abstract
OBJECTIVE To examine the potential influence of a ketogenic diet on serum concentrations of antiseizure medications (ASMs) in children with drug resistant epilepsy. METHODS We investigated the serum concentrations of ASMs in 25 children with drug resistant epilepsy, 2-13 years of age, treated with a classical ketogenic diet for 12 weeks. The patients were recruited from the National Centre for Epilepsy from August 15th, 2017, to January 24th, 2022. Changes in ASM serum concentrations were analyzed using a mixed effect model analysis. Significance level was set at P < 0.05 for all comparisons. RESULTS The participants used 12 different ASMs during the study. The mean number of ASMs was 2.4 (±SD 0.7). None of the participants changed the type or dose of the ASMs during the intervention period. The serum concentrations of clobazam (n = 9, P = 0.002), desmethylclobazam (n = 9, P = 0.010), and lamotrigine (n = 6, P = 0.016) decreased significantly during the dietary treatment. The analytes with the largest reduction in serum concentration after 12 weeks of dietary treatment were clobazam (mean change -38%) and desmethylclobazam (mean change -37%). We found no significant change in the serum concentrations of levetiracetam, topiramate, and valproic acid. SIGNIFICANCE We identified a significant decrease in the serum concentrations of clobazam, desmethylclobazam, and lamotrigine following a 12-week ketogenic diet intervention in children with drug resistant epilepsy. An unintended decrease in the serum concentrations of ASMs may render the patient prone to seizures. Measurements of ASM serum concentrations might be useful in patients on a ketogenic diet, especially in patients with lack of efficacy of the dietary treatment.
Collapse
Affiliation(s)
- Sigrid Pedersen
- National Centre for Epilepsy, Member of the ERN EpiCareOslo University HospitalOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Magnhild Kverneland
- National Centre for Epilepsy, Member of the ERN EpiCareOslo University HospitalOsloNorway
| | - Knut Rudi
- Department of ChemistryNorwegian University of Life SciencesÅsNorway
| | - Kristina Gervin
- Department of Research and InnovationOslo University HospitalOsloNorway
| | - Cecilie Johannessen Landmark
- National Centre for Epilepsy, Member of the ERN EpiCareOslo University HospitalOsloNorway
- Department of PharmacologyOslo University HospitalOsloNorway
- Department of PharmacyOslo Metropolitan UniversityOsloNorway
| | - Per Ole Iversen
- Department of NutritionUniversity of OsloOsloNorway
- Department of HematologyOslo University HospitalOsloNorway
| | - Kaja Kristine Selmer
- National Centre for Epilepsy, Member of the ERN EpiCareOslo University HospitalOsloNorway
- Department of Research and InnovationOslo University HospitalOsloNorway
| |
Collapse
|
6
|
Weger M, Weger BD, Gachon F. Understanding circadian dynamics: current progress and future directions for chronobiology in drug discovery. Expert Opin Drug Discov 2023. [PMID: 37300813 DOI: 10.1080/17460441.2023.2224554] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/08/2023] [Indexed: 06/12/2023]
Abstract
INTRODUCTION Most mammalian physiology is orchestrated by the circadian clock, including drug transport and metabolism. As a result, efficacy and toxicity of many drugs are influenced by the timing of their administration, which has led to the establishment of the field of chronopharmacology. AREAS COVERED In this review, the authors provide an overview of the current knowledge about the time-of-day dependent aspects of drug metabolism and the importance of chronopharmacological strategies for drug development. They also discuss the factors influencing rhythmic drug pharmacokinetic including sex, metabolic diseases, feeding rhythms, and microbiota, that are often overlooked in the context of chronopharmacology. This article summarizes the involved molecular mechanisms and functions and explains why these parameters should be considered in the process of drug discovery. EXPERT OPINION Although chronomodulated treatments have shown promising results, particularly for cancer, the practice is still underdeveloped due to the associated high cost and time investments. However, implementing this strategy at the preclinical stage could offer a new opportunity to translate preclinical discoveries into successful clinical treatments.
Collapse
Affiliation(s)
- Meltem Weger
- Institute for Molecular Bioscience, The University of Queensland, QLD, Australia
| | - Benjamin D Weger
- Institute for Molecular Bioscience, The University of Queensland, QLD, Australia
| | - Frédéric Gachon
- Institute for Molecular Bioscience, The University of Queensland, QLD, Australia
| |
Collapse
|
7
|
Huang YL, De Gregorio C, Silva V, Elorza ÁA, Léniz P, Aliaga-Tobar V, Maracaja-Coutinho V, Budini M, Ezquer F, Ezquer M. Administration of Secretome Derived from Human Mesenchymal Stem Cells Induces Hepatoprotective Effects in Models of Idiosyncratic Drug-Induced Liver Injury Caused by Amiodarone or Tamoxifen. Cells 2023; 12:cells12040636. [PMID: 36831304 PMCID: PMC9954258 DOI: 10.3390/cells12040636] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/19/2023] [Accepted: 02/07/2023] [Indexed: 02/18/2023] Open
Abstract
Drug-induced liver injury (DILI) is one of the leading causes of acute liver injury. While many factors may contribute to the susceptibility to DILI, obese patients with hepatic steatosis are particularly prone to suffer DILI. The secretome derived from mesenchymal stem cell has been shown to have hepatoprotective effects in diverse in vitro and in vivo models. In this study, we evaluate whether MSC secretome could improve DILI mediated by amiodarone (AMI) or tamoxifen (TMX). Hepatic HepG2 and HepaRG cells were incubated with AMI or TMX, alone or with the secretome of MSCs obtained from human adipose tissue. These studies demonstrate that coincubation of AMI or TMX with MSC secretome increases cell viability, prevents the activation of apoptosis pathways, and stimulates the expression of priming phase genes, leading to higher proliferation rates. As proof of concept, in a C57BL/6 mouse model of hepatic steatosis and chronic exposure to AMI, the MSC secretome was administered endovenously. In this study, liver injury was significantly attenuated, with a decrease in cell infiltration and stimulation of the regenerative response. The present results indicate that MSC secretome administration has the potential to be an adjunctive cell-free therapy to prevent liver failure derived from DILI caused by TMX or AMI.
Collapse
Affiliation(s)
- Ya-Lin Huang
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile
| | - Cristian De Gregorio
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile
| | - Verónica Silva
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile
| | - Álvaro A. Elorza
- Instituto de Ciencias Biomédicas, Facultad de Medicina y Ciencias de la Vida, Universidad Andres Bello, Santiago 7610658, Chile
| | - Patricio Léniz
- Unidad de Cirugía Plástica, Reparadora y Estética, Clínica Alemana, Santiago 7610658, Chile
| | - Víctor Aliaga-Tobar
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 7610658, Chile
- Centro de Modelamiento Molecular, Biofísica y Bioinformática (CM2B2), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 7610658, Chile
- Laboratorio de Bioingeniería, Instituto de Ciencias de la Ingeniería, Universidad de O’Higgins, Rancagua 7610658, Chile
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 7610658, Chile
- Centro de Modelamiento Molecular, Biofísica y Bioinformática (CM2B2), Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago 7610658, Chile
| | - Mauricio Budini
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago 7610658, Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile
- Correspondence: (F.E.); (M.E.); Tel.: +56-990-699-272 (F.E.); +56-976-629-880 (M.E.)
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago 7610658, Chile
- Correspondence: (F.E.); (M.E.); Tel.: +56-990-699-272 (F.E.); +56-976-629-880 (M.E.)
| |
Collapse
|
8
|
Short-Term High-Fat Diet Alters Acetaminophen Metabolism in Healthy Individuals. Ther Drug Monit 2022; 44:797-804. [PMID: 35500453 DOI: 10.1097/ftd.0000000000000993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/28/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND Acetaminophen is metabolized through a nontoxic sulfation and glucuronidation pathway and toxic oxidation pathway (via CYP2E1 and CYP1A2). A short-term high-fat diet induces alterations in the steatotic liver and may alter hepatic drug enzyme activity. In the case of acetaminophen, these alterations may result in an increased risk of hepatotoxicity. Therefore, this study was conducted to assess the effect of a 3-day hypercaloric high-fat diet on the plasma levels of acetaminophen metabolites. METHODS Nine healthy subjects participated in this randomized, crossover intervention study. The subjects consumed a regular diet or a regular diet supplemented with 500 mL of cream (1700 kcal) for 3 days and then fasted overnight. After ingesting 1000-mg acetaminophen, the plasma concentration of acetaminophen (APAP) and its metabolites [acetaminophen glucuronide, acetaminophen sulfate, 3-cysteinyl-acetaminophen, and 3-(N-acetyl-L-cystein-S-yl)-acetaminophen, and 3-methoxy-acetaminophen] were measured. RESULTS The 3-day high-fat diet increased the extrapolated area under the concentration-time curve from 0 to infinity (area under the curve 0-inf ) of APAP-Cys by approximately 20% ( P = 0.02) and that from 0 to 8 hours (area under the curve 0-8 ) of APAP-Cys-NAC by approximately 39% ( P = 0.01). The 3-day high-fat diet did not alter the pharmacokinetic parameters of the parent compound acetaminophen and other metabolites. CONCLUSIONS A short-term, hypercaloric, high-fat diet increases the plasma levels of the APAP metabolites formed by the oxidation pathway, which may increase the risk of hepatotoxicity.
Collapse
|
9
|
Santativongchai P, Srisuksai K, Parunyakul K, Thiendedsakul P, Lertwatcharasarakul P, Fungfuang W, Tulayakul P. Effects of Crocodile Oil ( Crocodylus siamensis) on Liver Enzymes: Cytochrome P450 and Glutathione S-Transferase Activities in High-fat DietFed Rats. Vet Med Int 2022; 2022:9990231. [PMID: 36457890 PMCID: PMC9708360 DOI: 10.1155/2022/9990231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 07/30/2023] Open
Abstract
Crocodile oil is a highly effective treatment for ailments ranging from skin conditions to cancer. However, the effects of the oil on liver detoxification pathways are not well studied. This study aimed to investigate the effects of crocodile oil on the detoxification enzyme activities and the mRNA expressions of cytochrome P450 1A2 (CYP1A2), cytochrome P450 2E1 (CYP2E1), and glutathione S-transferase (GST) in rats. The rats were divided into four groups (n = 7/group): rats received a standard diet (C), a high-fat diet or HFD (H), and HFD with 1 ml (HCO1) and 3 ml (HCO3) of the oil per kg body weight. Interestingly, the oil yields from this study presented alpha-linolenic acid (0.96%) at similar levels compared with fish oil. The results revealed that HFD significantly increased the activity and relative gene expression of CYP1A2 in the H group (P < 0.05), whereas 3% crocodile oil normalized the enzyme activities compared to the C group. This suggested inhibiting the HFD-induced expression of CYP1A2 mediated by the omega-3 fatty acids found in the oil. Also, crocodile oil supplementation did not reduce the activities of GST. However, the relative gene expression of GSTA1 was significantly decreased (P < 0.05) in the HCO1 and HCO3 groups compared to the H group, which might be attributed to the lower lipid peroxidation that occurred in the liver tissues. Therefore, it could be suggested that using crocodile oil could help in liver detoxification through the CYP1A2 even when offered with a HFD.
Collapse
Affiliation(s)
- Pitchaya Santativongchai
- Bio-Veterinary Sciences (International Program), Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand
| | - Krittika Srisuksai
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Kongphop Parunyakul
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Piriyaporn Thiendedsakul
- Animal Health and Biomedical Science, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand
| | - Preeda Lertwatcharasarakul
- Department of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Kamphaeng Saen Campus, Nakhon Pathom 73140, Thailand
| | - Wirasak Fungfuang
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Phitsanu Tulayakul
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Kasetsart University, Kamphaeng Saen Campus, Nakhon Pathom 73140, Thailand
- Kasetsart University Research and Development Institute (KURDI), Kasetsart University, Bangkok 10900, Thailand
| |
Collapse
|
10
|
Qu H, Zong L, Sang J, Wa Y, Chen D, Huang Y, Chen X, Gu R. Effect of Lactobacillus rhamnosus hsryfm 1301 Fermented Milk on Lipid Metabolism Disorders in High-Fat-Diet Rats. Nutrients 2022; 14:4850. [PMID: 36432537 PMCID: PMC9698387 DOI: 10.3390/nu14224850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
To further explore and improve the mechanism of probiotics to alleviate the disorder of lipid metabolism, transcriptomic and metabolomic with bioinformatic analysis were combined. In the present study, we successfully established a rat model of lipid metabolism disorder using a high-fat diet. Intervention with Lactobacillus rhamnosus hsryfm 1301 fermented milk resulted in a significant reduction in body weight, serum free fatty acid and blood lipid levels (p < 0.05), which predicted that the lipid metabolism disorder was alleviated in rats. Metabolomics and transcriptomics identified a total of 33 significantly different metabolites and 183 significantly different genes screened in the intervention group compared to the model group. Comparative analysis of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotations identified a total of 61 pathways in which differential metabolites and genes were jointly involved, with linoleic acid metabolism, glycine, serine and threonine metabolism and glutamatergic synapse in both transcriptome and metabolome being found to be significantly altered (p < 0.05). Lactobacillus rhamnosus hsryfm 1301 fermented milk was able to directly regulate lipid metabolism disorders by regulating the metabolic pathways of linoleic acid metabolism, glycerophospholipid metabolism, fatty acid biosynthesis, alpha-linolenic acid metabolism, fatty acid degradation, glycerolipid metabolism and arachidonic acid metabolism. In addition, we found that Lactobacillus rhamnosus hsryfm 1301 fermented milk indirectly regulates lipid metabolism through regulating amino acid metabolism, the nervous system, the endocrine system and other pathways. Lactobacillus rhamnosus hsryfm 1301 fermented milk could alleviate the disorders of lipid metabolism caused by high-fat diet through multi-target synergy.
Collapse
Affiliation(s)
- Hengxian Qu
- College of Food Science and Technology, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Dairy Biotechnology and Safety Control, Yangzhou University, Yangzhou 225000, China
| | - Lina Zong
- College of Food Science and Technology, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Dairy Biotechnology and Safety Control, Yangzhou University, Yangzhou 225000, China
| | - Jian Sang
- Realab Biotechnology Co., Ltd., Beijing 100000, China
| | - Yunchao Wa
- College of Food Science and Technology, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Dairy Biotechnology and Safety Control, Yangzhou University, Yangzhou 225000, China
| | - Dawei Chen
- College of Food Science and Technology, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Dairy Biotechnology and Safety Control, Yangzhou University, Yangzhou 225000, China
| | - Yujun Huang
- College of Food Science and Technology, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Dairy Biotechnology and Safety Control, Yangzhou University, Yangzhou 225000, China
| | - Xia Chen
- College of Food Science and Technology, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Dairy Biotechnology and Safety Control, Yangzhou University, Yangzhou 225000, China
| | - Ruixia Gu
- College of Food Science and Technology, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Dairy Biotechnology and Safety Control, Yangzhou University, Yangzhou 225000, China
| |
Collapse
|
11
|
Krøyer Rasmussen M, Thøgersen R, Horsbøl Lindholm P, Bertram HC, Pilegaard H. Hepatic PGC-1α has minor regulatory effect on the transcriptome and metabolome during high fat high fructose diet and exercise. Gene 2022; 851:147039. [DOI: 10.1016/j.gene.2022.147039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022]
|
12
|
Obesity-related genomic instability and altered xenobiotic metabolism: possible consequences for cancer risk and chemotherapy. Expert Rev Mol Med 2022; 24:e28. [PMID: 35899852 PMCID: PMC9884759 DOI: 10.1017/erm.2022.22] [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] [Indexed: 11/06/2022]
Abstract
The increase in the prevalence of obesity has led to an elevated risk for several associated diseases including cancer. Several studies have investigated the DNA damage in human blood samples and showed a clear trend towards increased DNA damage in obesity. Reduced genomic stability is thus one of the consequences of obesity, which may contribute to the related cancer risk. Whether this is influenced by compromised DNA repair has not been elucidated sufficiently yet. On the other hand, obesity has also been linked to reduced therapy survival and increased adverse effects during chemotherapy, although the available data are controversial. Despite some indications that obesity might alter hepatic metabolism, current literature in humans is insufficient, and results from animal studies are inconclusive. Here we have summarised published data on hepatic drug metabolism to understand the impact of obesity on cancer therapy better. Furthermore, we highlight knowledge gaps in the interrelationship between obesity and drug metabolism from a toxicological perspective.
Collapse
|
13
|
Soundharrajan I, Karnan M, Jung JS, Lee KD, Lee JC, Ramesh T, Kim D, Choi KC. A Transcriptomic Response to Lactiplantibacillus plantarum-KCC48 against High-Fat Diet-Induced Fatty Liver Diseases in Mice. Int J Mol Sci 2022; 23:6750. [PMID: 35743193 PMCID: PMC9224190 DOI: 10.3390/ijms23126750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 02/05/2023] Open
Abstract
The most prevalent chronic liver disorder in the world is fatty liver disease caused by a high-fat diet. We examined the effects of Lactiplantibacillus plantarum-KCC48 on high-fat diet-induced (HFD) fatty liver disease in mice. We used the transcriptome tool to perform a systematic evaluation of hepatic mRNA transcripts changes in high-fat diet (HFD)-fed animals and high-fat diet with L. plantarum (HFLPD)-fed animals. HFD causes fatty liver diseases in animals, as evidenced by an increase in TG content in liver tissues compared to control animals. Based on transcriptome data, 145 differentially expressed genes (DEGs) were identified in the liver of HFD-fed mice compared to control mice. Moreover, 61 genes were differentially expressed in the liver of mice fed the HFLPD compared to mice fed the HFD. Additionally, 43 common DEGs were identified between HFD and HFLPD. These genes were enriched in metabolic processes, retinol metabolism, the PPAR signaling pathway, fatty acid degradation, arachidonic metabolism, and steroid hormone synthesis. Taking these data into consideration, it can be concluded that L. plantarum-KCC48 treatment significantly regulates the expression of genes involved in hepatosteatosis caused by HFD, which may prevent fatty liver disease.
Collapse
Affiliation(s)
- Ilavenil Soundharrajan
- Grassland and Forage Division, Rural Development Administration, National Institute of Animal Science, Cheonan 31000, Korea; (I.S.); (M.K.); (J.-S.J.)
| | - Muthusamy Karnan
- Grassland and Forage Division, Rural Development Administration, National Institute of Animal Science, Cheonan 31000, Korea; (I.S.); (M.K.); (J.-S.J.)
| | - Jeong-Sung Jung
- Grassland and Forage Division, Rural Development Administration, National Institute of Animal Science, Cheonan 31000, Korea; (I.S.); (M.K.); (J.-S.J.)
| | - Kyung-Dong Lee
- Department of Companion Animals, Dongsin University, Naju 58245, Korea;
| | - Jeong-Chae Lee
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea;
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Dahye Kim
- Animal Genomics and Bioinformatics Division, National Institute of Animal Science, Wanju 55365, Korea
| | - Ki-Choon Choi
- Grassland and Forage Division, Rural Development Administration, National Institute of Animal Science, Cheonan 31000, Korea; (I.S.); (M.K.); (J.-S.J.)
| |
Collapse
|
14
|
Garcia WL, Miller CJ, Lomas GX, Gaither KA, Tyrrell KJ, Smith JN, Brandvold KR, Wright AT. Profiling How the Gut Microbiome Modulates Host Xenobiotic Metabolism in Response to Benzo[ a]pyrene and 1-Nitropyrene Exposure. Chem Res Toxicol 2022; 35:585-596. [PMID: 35347982 PMCID: PMC9878584 DOI: 10.1021/acs.chemrestox.1c00360] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The gut microbiome is a key contributor to xenobiotic metabolism. Polycyclic aromatic hydrocarbons (PAHs) are an abundant class of environmental contaminants that have varying levels of carcinogenicity depending on their individual structures. Little is known about how the gut microbiome affects the rates of PAH metabolism. This study sought to determine the role that the gut microbiome has in determining the various aspects of metabolism in the liver, before and after exposure to two structurally different PAHs, benzo[a]pyrene and 1-nitropyrene. Following exposures, the metabolic rates of PAH metabolism were measured, and activity-based protein profiling was performed. We observed differences in PAH metabolism rates between germ-free and conventional mice under both unexposed and exposed conditions. Our activity-based protein profiling (ABPP) analysis showed that, under unexposed conditions, there were only minor differences in total P450 activity in germ-free mice relative to conventional mice. However, we observed distinct activity profiles in response to corn oil vehicle and PAH treatment, primarily in the case of 1-NP treatment. This study revealed that the repertoire of active P450s in the liver is impacted by the presence of the gut microbiome, which modifies PAH metabolism in a substrate-specific fashion.
Collapse
Affiliation(s)
- Whitney L. Garcia
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA),Biological Systems Engineering Department, CAHNRS, Washington State University, Pullman, WA 99163 (USA)
| | - Carson J. Miller
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA)
| | - Gerard X. Lomas
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA)
| | - Kari A. Gaither
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA)
| | - Kimberly J. Tyrrell
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA)
| | - Jordan N. Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA),Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331 (USA)
| | - Kristoffer R. Brandvold
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA),Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202 (USA),Corresponding Authors: Kristoffer R. Brandvold - Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA); , Aaron T. Wright - Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA);
| | - Aaron T. Wright
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA),The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99163 (USA),Corresponding Authors: Kristoffer R. Brandvold - Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA); , Aaron T. Wright - Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352 (USA);
| |
Collapse
|
15
|
Bustamante-Marin XM, Merlino JL, Devericks E, Carson MS, Hursting SD, Stewart DA. Mechanistic Targets and Nutritionally Relevant Intervention Strategies to Break Obesity-Breast Cancer Links. Front Endocrinol (Lausanne) 2021; 12:632284. [PMID: 33815289 PMCID: PMC8011316 DOI: 10.3389/fendo.2021.632284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 02/17/2021] [Indexed: 12/29/2022] Open
Abstract
The worldwide prevalence of overweight and obesity has tripled since 1975. In the United States, the percentage of adults who are obese exceeds 42.5%. Individuals with obesity often display multiple metabolic perturbations, such as insulin resistance and persistent inflammation, which can suppress the immune system. These alterations in homeostatic mechanisms underlie the clinical parameters of metabolic syndrome, an established risk factor for many cancers, including breast cancer. Within the growth-promoting, proinflammatory milieu of the obese state, crosstalk between adipocytes, immune cells and breast epithelial cells occurs via obesity-associated hormones, angiogenic factors, cytokines, and other mediators that can enhance breast cancer risk and/or progression. This review synthesizes evidence on the biological mechanisms underlying obesity-breast cancer links, with emphasis on emerging mechanism-based interventions in the context of nutrition, using modifiable elements of diet alone or paired with physical activity, to reduce the burden of obesity on breast cancer.
Collapse
Affiliation(s)
| | - Jenna L. Merlino
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
| | - Emily Devericks
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
| | - Meredith S. Carson
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
| | - Stephen D. Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC, United States
| | - Delisha A. Stewart
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina, Kannapolis, NC, United States
| |
Collapse
|
16
|
He Y, Yang T, Du Y, Qin L, Ma F, Wu Z, Ling H, Yang L, Wang Z, Zhou Q, Ge G, Lu Y. High fat diet significantly changed the global gene expression profile involved in hepatic drug metabolism and pharmacokinetic system in mice. Nutr Metab (Lond) 2020; 17:37. [PMID: 32489392 PMCID: PMC7245748 DOI: 10.1186/s12986-020-00456-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 05/07/2020] [Indexed: 12/25/2022] Open
Abstract
Background High fat diet impact transcription of hepatic genes responsible for drug metabolism and pharmacokinetics. Until now, researches just focused on a couple specific genes without a global profile showing. Age-dependent manner was also not noted well. This study aims to investigate the high fat diet effect on transcriptome of drug metabolism and pharmacokinetic system in mouse livers and show the age-dependent evidence. Methods C57BL/6 male mice were used in this experiment. High fat diet was used to treat mice for 16 and 38 weeks. Serum total cholesterol, low density lipoprotein cholesterol, aspartate transaminase, and alanine transaminaselevels were measured. Meanwhile, Histology, RNA-Seq, RT-PCR analysis and fourteen major hepatic bile acids quantification were performed for the liver tissues. Data was mined at levels of genes, drug metabolism and pharmacokinetic sysem, and genome wide. Results Treatment with high fat diet for 38 weeks significantly increased levels of serum lipids as well as aspartate transaminase, and alanine transaminase. Meanwhile, lipid accumulation in livers was observed. At week 38 of the experiment, the profile of 612 genes involved in drug metabolism and pharmacokinetics was significantly changed, indicated by a heatmap visulization and a principal component analysis. In total 210 genes were significantly regulated. Cyp3a11, Cyp4a10, and Cyp4a14 were down-regulated by 10–35 folds, while these three genes also were highly expressed in the liver. High fat diet regulated 11% of genome-wide gene while 30% of genes involved in the hepatic drug metabolism and pharmacokinetic system. Genes, including Adh4, Aldh1b1, Cyp3a11, Cyp4a10, Cyp8b1, Fmo2, Gsta3, Nat8f1, Slc22a7, Slco1a4, Sult5a1, and Ugt1a9, were regulated by high fat diet as an aging-dependent manner. Bile acids homeostasis, in which many genes related to metabolism and transportation were enriched, was also changed by high fat diet with an aging-dependet manner. Expression of genes in drug metabolism and disposition system significantly correlated to serum lipid profiles, and frequently correlated with each other. Conclusions High fat diet changed the global transcription profile of hepatic drug metabolism and pharmacokinetic system with a age-dependent manner.
Collapse
Affiliation(s)
- Yuqi He
- The Key Laboratory of the Minstry of Education of the Basic Pharmacology and the Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xue-Fu Road, Zunyi City, 563009 Guizhou China.,Institute of Chinese Materia Medica, Shanghai Key Laboratory of Complex Prescription and the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines , Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Yang
- The Key Laboratory of the Minstry of Education of the Basic Pharmacology and the Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xue-Fu Road, Zunyi City, 563009 Guizhou China
| | - Yimei Du
- The Key Laboratory of the Minstry of Education of the Basic Pharmacology and the Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xue-Fu Road, Zunyi City, 563009 Guizhou China
| | - Lin Qin
- The Key Laboratory of the Minstry of Education of the Basic Pharmacology and the Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xue-Fu Road, Zunyi City, 563009 Guizhou China
| | - Feifei Ma
- The Key Laboratory of the Minstry of Education of the Basic Pharmacology and the Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xue-Fu Road, Zunyi City, 563009 Guizhou China
| | - Zunping Wu
- The Key Laboratory of the Minstry of Education of the Basic Pharmacology and the Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xue-Fu Road, Zunyi City, 563009 Guizhou China
| | - Hua Ling
- School of Pharmacy, Philadelphia College of Osteopathic Medicine, Suwanee, GA USA
| | - Li Yang
- Institute of Chinese Materia Medica, Shanghai Key Laboratory of Complex Prescription and the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines , Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhengtao Wang
- Institute of Chinese Materia Medica, Shanghai Key Laboratory of Complex Prescription and the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines , Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qingdi Zhou
- School of Chemistry, The University of Sydney, Camperdown, NSW2006 Australia
| | - Guangbo Ge
- The Key Laboratory of the Minstry of Education of the Basic Pharmacology and the Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xue-Fu Road, Zunyi City, 563009 Guizhou China.,Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanliu Lu
- The Key Laboratory of the Minstry of Education of the Basic Pharmacology and the Joint International Research Laboratory of Ethnomedicine of the Ministry of Education, School of Pharmacy, Zunyi Medical University, 6 West Xue-Fu Road, Zunyi City, 563009 Guizhou China.,Institute of Chinese Materia Medica, Shanghai Key Laboratory of Complex Prescription and the Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines , Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Background Diet Influences TMAO Concentrations Associated with Red Meat Intake without Influencing Apparent Hepatic TMAO-Related Activity in a Porcine Model. Metabolites 2020; 10:metabo10020057. [PMID: 32041174 PMCID: PMC7074160 DOI: 10.3390/metabo10020057] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/30/2020] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
Red meat has been associated with an increased cardiovascular disease (CVD) risk, possibly through gut microbial-derived trimethylamine-N-oxide (TMAO). However, previous reports are conflicting, and influences from the background diet may modulate the impact of meat consumption. This study investigated the effect of red and white meat intake combined with two different background diets on urinary TMAO concentration and its association with the colon microbiome in addition to apparent hepatic TMAO-related activity. For 4 weeks, 32 pigs were fed chicken or red and processed meat combined with a prudent or western background diet. 1H NMR-based metabolomics analysis was conducted on urine samples and hepatic Mrna expression of TMAO-related genes determined. Lower urinary TMAO concentrations were observed after intake of red and processed meat when consumed with a prudent compared to a western background diet. In addition, correlation analyses between urinary TMAO concentrations and relative abundance of colon bacterial groups suggested an association between TMAO and specific bacterial taxa. Diet did not affect the hepatic Mrna expression of genes related to TMAO formation. The results suggest that meat-induced TMAO formation is regulated by mechanisms other than alterations at the hepatic gene expression level, possibly involving modulations of the gut microbiota.
Collapse
|
18
|
Wang P, Shao X, Bao Y, Zhu J, Chen L, Zhang L, Ma X, Zhong XB. Impact of obese levels on the hepatic expression of nuclear receptors and drug-metabolizing enzymes in adult and offspring mice. Acta Pharm Sin B 2020; 10:171-185. [PMID: 31993314 PMCID: PMC6976990 DOI: 10.1016/j.apsb.2019.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/30/2019] [Accepted: 09/18/2019] [Indexed: 12/13/2022] Open
Abstract
The prevalence of obesity-associated conditions raises new challenges in clinical medication. Although altered expression of drug-metabolizing enzymes (DMEs) has been shown in obesity, the impacts of obese levels (overweight, obesity, and severe obesity) on the expression of DMEs have not been elucidated. Especially, limited information is available on whether parental obese levels affect ontogenic expression of DMEs in children. Here, a high-fat diet (HFD) and three feeding durations were used to mimic different obese levels in C57BL/6 mice. The hepatic expression of five nuclear receptors (NRs) and nine DMEs was examined. In general, a trend of induced expression of NRs and DMEs (except for Cyp2c29 and 3a11) was observed in HFD groups compared to low-fat diet (LFD) groups. Differential effects of HFD on the hepatic expression of DMEs were found in adult mice at different obese levels. Family-based dietary style of an HFD altered the ontogenic expression of DMEs in the offspring older than 15 days. Furthermore, obese levels of parental mice affected the hepatic expression of DMEs in offspring. Overall, the results indicate that obese levels affected expression of the DMEs in adult individuals and that of their children. Drug dosage might need to be optimized based on the obese levels.
Collapse
Key Words
- 18-HA, adult mice fed with 18 weeks HFD
- 18-LA, adult mice fed with 18 weeks LFD
- 4-HA, adult mice fed with 4 weeks HFD
- 4-LA, adult mice fed with 4 weeks LFD
- 7-ER, 7-ethoxyresorufin
- 8-HA, adult mice fed with 8 weeks HFD
- 8-LA, adult mice fed with 8 weeks LFD
- AhR, aryl hydrocarbon receptor
- BMI, body mass index
- CAR, constitutive androstane receptor
- CHZ, chlorzoxazone
- CYP2E1, cytochrome P450 2E1
- DIO, diet-induced obesity
- DMEs, drug-metabolizing enzymes
- Diet-induced obesity
- Drug-metabolizing enzymes
- EFV, efavirenz
- Gapdh, glyceraldehyde-3-phosphate dehydrogenase
- HFD, high-fat diet
- HNF4α, hepatocyte nuclear factor 4 alpha
- High-fat diet
- LFD, low-fat diet
- MDZ, midazolam
- MPA, mobile phase A
- MPB, mobile phase B
- NADPH, nicotinamide adenine dinucleotide phosphate
- NAFLD, non-alcoholic fatty liver disease
- NRs, nuclear receptors
- Nuclear receptors
- O-18-HA, offspring from parental mice fed with 18 weeks HFD
- O-18-LA, offspring from parental mice fed with 18 weeks LFD
- O-4-HA, offspring from parental mice fed with 4 weeks HFD
- O-4-LA, offspring from parental mice fed with 4 weeks LFD
- O-8-HA, offspring from parental mice fed with 8 weeks HFD
- O-8-LA, offspring from parental mice fed with 8 weeks LFD
- Ontogenic expression
- Overweight
- PBS, phosphate-buffered saline
- PPARα, peroxisome proliferator-activated receptor alpha
- PXR, pregnane X receptor
- RSF, resorufin
- RT-qPCR, real-time quantitative PCR
- SD, standard deviation
- SULT1A1, sulfotransferase 1A1
- UGT1A1, uridine diphosphate glucuronosyltransferase 1A1
Collapse
Affiliation(s)
- Pei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Xueyan Shao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Yifan Bao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Junjie Zhu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Liming Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaochao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiao-bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
19
|
Gröschel C, Prinz-Wohlgenannt M, Mesteri I, Karuthedom George S, Trawnicek L, Heiden D, Aggarwal A, Tennakoon S, Baumgartner M, Gasche C, Lang M, Marculescu R, Manhardt T, Schepelmann M, Kallay E. Switching to a Healthy Diet Prevents the Detrimental Effects of Western Diet in a Colitis-Associated Colorectal Cancer Model. Nutrients 2019; 12:E45. [PMID: 31877961 PMCID: PMC7019913 DOI: 10.3390/nu12010045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 01/19/2023] Open
Abstract
Inflammatory bowel disease increases the odds of developing colitis-associated cancer. We hypothesized that Western-style diet (WD) aggravates azoxymethane (AOM)/dextran sulfate sodium salt (DSS)-induced colitis-associated tumorigenesis and that switching to the standard AIN93G diet will ameliorate disease symptoms even after cancer initiation. Female BALB/c mice received either WD (WD group) or standard AIN93G diet (AIN group) for the whole experimental period. After five weeks, the mice received 12.5 mg/kg AOM intraperitoneally, followed by three DSS cycles. In one group of mice, the WD was switched to AIN93G the day before starting the first DSS cycle (WD/AIN group). Feeding the WD during the whole experimental period aggravated colitis symptoms, shortened the colon (p < 0.05), changed microbiota composition and increased tumor promotion. On molecular level, the WD reduced proliferation (p < 0.05) and increased expression of the vitamin D catabolizing enzyme Cyp24a1 (p < 0.001). The switch to the AIN93G diet ameliorated this effect, reflected by longer colons, fewer (p < 0.05) and smaller (p < 0.01) aberrant colonic crypt foci, comparable with the AIN group. Our results show that switching to a healthy diet, even after cancer initiation is able to revert the deleterious effect of the WD and could be an effective preventive strategy to reduce colitis symptoms and prevent tumorigenesis.
Collapse
Affiliation(s)
- Charlotte Gröschel
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.-W.); (S.K.G.); (L.T.); (D.H.); (A.A.); (S.T.); (T.M.); (M.S.)
| | - Maximilian Prinz-Wohlgenannt
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.-W.); (S.K.G.); (L.T.); (D.H.); (A.A.); (S.T.); (T.M.); (M.S.)
| | - Ildiko Mesteri
- Institute of Pathology Überlingen, 88662 Überlingen, Germany;
| | - Sobha Karuthedom George
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.-W.); (S.K.G.); (L.T.); (D.H.); (A.A.); (S.T.); (T.M.); (M.S.)
| | - Lena Trawnicek
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.-W.); (S.K.G.); (L.T.); (D.H.); (A.A.); (S.T.); (T.M.); (M.S.)
| | - Denise Heiden
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.-W.); (S.K.G.); (L.T.); (D.H.); (A.A.); (S.T.); (T.M.); (M.S.)
| | - Abhishek Aggarwal
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.-W.); (S.K.G.); (L.T.); (D.H.); (A.A.); (S.T.); (T.M.); (M.S.)
| | - Samawansha Tennakoon
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.-W.); (S.K.G.); (L.T.); (D.H.); (A.A.); (S.T.); (T.M.); (M.S.)
| | - Maximilian Baumgartner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, 1090 Vienna, Austria; (M.B.); (C.G.); (M.L.)
| | - Christoph Gasche
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, 1090 Vienna, Austria; (M.B.); (C.G.); (M.L.)
| | - Michaela Lang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, 1090 Vienna, Austria; (M.B.); (C.G.); (M.L.)
| | - Rodrig Marculescu
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria;
| | - Teresa Manhardt
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.-W.); (S.K.G.); (L.T.); (D.H.); (A.A.); (S.T.); (T.M.); (M.S.)
| | - Martin Schepelmann
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.-W.); (S.K.G.); (L.T.); (D.H.); (A.A.); (S.T.); (T.M.); (M.S.)
| | - Enikö Kallay
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (C.G.); (M.P.-W.); (S.K.G.); (L.T.); (D.H.); (A.A.); (S.T.); (T.M.); (M.S.)
| |
Collapse
|
20
|
Emond C, DeVito MJ, Diliberto JJ, Birnbaum LS. The Influence of Obesity on the Pharmacokinetics of Dioxin in Mice: An Assessment Using Classical and PBPK Modeling. Toxicol Sci 2019; 164:218-228. [PMID: 29596651 DOI: 10.1093/toxsci/kfy078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The effects of body fat mass on the elimination of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) was examined in mice. When male C57BL/6J mice are fed a high-fat, simple carbohydrate diet (HFD) for 13 weeks, they develop an obese phenotype. In contrast, A/J mice fed an HFD do not become obese. After 13 weeks on a normal diet (ND) or HFD, male C57BL/6J and A/J mice received a single dose by gavage of 0.1 or 5.0 µg of 2,3,7,8-tetrachloro[1,6-3H] dibenzo-p-dioxin per kg body weight. Using classical pharmacokinetics, the blood elimination half-life of TCDD was approximately 10 and 2 times longer in the C57BL/6J on the HFD compared with the mice on the ND at 0.1 and 5.0 μg/kg doses, respectively. The diet did not increase the blood half-life of TCDD in the A/J mice, which did not get obese. Using a physiologically based pharmacokinetic model for TCDD that incorporated experimentally derived percent body fat mass and tissue partition coefficients, as well as data on hepatic sequestration, did not provide accurate predictions to the data and could not explain the increase in half-life of TCDD in the HFD groups. This work demonstrates that obesity influences the half-life of TCDD, but other undetermined factors are involved in its elimination because the increase in body fat mass, decreases in cytochrome P4501A2, and altered partition coefficients could not completely explain the prolonged half-life.
Collapse
Affiliation(s)
- Claude Emond
- BioSimulation Consulting Inc., Newark, DE, USA, 19713.,Department of Environmental and Occupational Health, University of Montreal, Quebec, Canada H3N 1X9
| | - Michael J DeVito
- National Institute of Environmental Health Sciences, National Toxicology Program, Research Triangle Park, NC, USA
| | - Janet J Diliberto
- National Health and Environmental Effects Research Laboratory, U.S. Environmental protection Agency, Research Triangle Park, NC, USA, 27711
| | - Linda S Birnbaum
- National Cancer Institute, Research Triangle Park, NC, USA, 27709
| |
Collapse
|
21
|
Naville D, Gaillard G, Julien B, Vega N, Pinteur C, Chanon S, Vidal H, Le Magueresse-Battistoni B. Chronic exposure to a pollutant mixture at low doses led to tissue-specific metabolic alterations in male mice fed standard and high-fat high-sucrose diet. CHEMOSPHERE 2019; 220:1187-1199. [PMID: 30722647 DOI: 10.1016/j.chemosphere.2018.12.177] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/26/2018] [Accepted: 12/23/2018] [Indexed: 06/09/2023]
Abstract
Excessive consumption of industrialized food and beverages is a major etiologic factor in the epidemics of obesity and associated metabolic diseases because these products are rich in fat and sugar. In addition, they contain food contact materials and environmental pollutants identified as metabolism disrupting chemicals. To evaluate the metabolic impact of these dietary threats (individually or combined), we used a male mouse model of chronic exposure to a mixture of low-dose archetypal food-contaminating chemicals that was added in standard or high-fat, high-sucrose (HFHS) diet. Specifically, the mixture contained bisphenol A, diethylhexylphthalate, 2,3,7,8-tetrachlorodibenzo-p-dioxine and polychlorinated biphenyl 153. Exposure lasted from 5 to 20 weeks of age. Metabolic exploration was conducted setting the basis of candidate gene expression mRNA analyses in liver, jejunum and adipose tissue depots from 20 week-old mice. Strong metabolic deleterious effects of the HFHS diet were demonstrated in line with obesity-associated metabolic features and insulin resistance. Pollutant exposure resulted in significant changes on plasma triglyceride levels and on the expression levels of genes mainly encoding xenobiotic processing in jejunum; estrogen receptors, regulators of lipoprotein lipase and inflammatory markers in jejunum and adipose tissues as well as adipogenesis markers. Importantly, the impact of pollutants was principally evidenced under standard diet. In addition, depending on nutritional conditions and on the metabolic tissue considered, the impact of pollutants could mimic or oppose the HFHS effects. Collectively, the present study extends the cocktail effect concept of a low-dosed pollutant mixture and originally points to tissue-specificity responsiveness especially in jejunum and adipose tissues.
Collapse
Affiliation(s)
- Danielle Naville
- Univ Lyon, CarMeN Laboratory, INSERM U1060, INRA U1397, INSA Lyon, Université Claude Bernard Lyon1, 69600, Oullins, France
| | - Guillain Gaillard
- Univ Lyon, CarMeN Laboratory, INSERM U1060, INRA U1397, INSA Lyon, Université Claude Bernard Lyon1, 69600, Oullins, France
| | - Benoit Julien
- Univ Lyon, CarMeN Laboratory, INSERM U1060, INRA U1397, INSA Lyon, Université Claude Bernard Lyon1, 69600, Oullins, France
| | - Nathalie Vega
- Univ Lyon, CarMeN Laboratory, INSERM U1060, INRA U1397, INSA Lyon, Université Claude Bernard Lyon1, 69600, Oullins, France
| | - Claudie Pinteur
- Univ Lyon, CarMeN Laboratory, INSERM U1060, INRA U1397, INSA Lyon, Université Claude Bernard Lyon1, 69600, Oullins, France
| | - Stéphanie Chanon
- Univ Lyon, CarMeN Laboratory, INSERM U1060, INRA U1397, INSA Lyon, Université Claude Bernard Lyon1, 69600, Oullins, France
| | - Hubert Vidal
- Univ Lyon, CarMeN Laboratory, INSERM U1060, INRA U1397, INSA Lyon, Université Claude Bernard Lyon1, 69600, Oullins, France
| | | |
Collapse
|
22
|
Knudsen JG, Bertholdt L, Gudiksen A, Gerbal-Chaloin S, Rasmussen MK. Skeletal Muscle Interleukin-6 Regulates Hepatic Cytochrome P450 Expression: Effects of 16-Week High-Fat Diet and Exercise. Toxicol Sci 2019; 162:309-317. [PMID: 29177473 DOI: 10.1093/toxsci/kfx258] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
High-fat diet (HFD) induces several changes to the pathways regulating energy homeostasis and changes the expression of the hepatic cytochrome p450 (Cyp) enzyme-system. Despite these pervious findings, it is still unclear how the effects of HFD and especially HFD in combination with treadmill running affect hepatic Cyp expression. In this study, we investigated the mRNA and protein expression of selected Cyp's in mice subjected to 16 weeks of HFD and treadmill running. To understand the regulatory mechanisms behind the exercise-induced reversion of the HFD-induced changes in Cyp expression, we used a model in which the exercise-induced myokine and known regulator of hepatic Cyp's, interleukin-6 (IL-6), were knocked out specifically in skeletal muscle. We found that HFD increased the mRNA expression of Cyp1a1 and Cyp4a10, and decreased the expression of Cyp2a4, Cyp2b10, Cyp2e1, and Cyp3a11. HFD in combination with treadmill running reversed the HFD increase in Cyp4a10 mRNA expression. In addition, we observed increased Cyp1a and Cyp3a protein expression as an effect of exercise, whereas Cyp2b expression was lowered as an effect of HFD. IL-6 effected the response in Cyp3a11 and Cyp1a expression. We observed no changes in the content of the aryl hydrocarbon receptor, constitutive androstane receptor, pregnane X receptor, or peroxisome proliferation activator receptor alpha. In conclusion, we show that both HFD and exercise in HFD-fed animals can regulate hepatic Cyp expression and that changes in Cyp3a in response to HFD and exercise are dependent on skeletal muscular IL-6.
Collapse
Affiliation(s)
- Jakob G Knudsen
- Department of Biology, Copenhagen University, DK-2200 Copenhagen, Denmark
| | - Lærke Bertholdt
- Department of Biology, Copenhagen University, DK-2200 Copenhagen, Denmark
| | - Anders Gudiksen
- Department of Biology, Copenhagen University, DK-2200 Copenhagen, Denmark
| | | | | |
Collapse
|
23
|
Li X, Wang Z, Klaunig JE. Modulation of xenobiotic nuclear receptors in high-fat diet induced non-alcoholic fatty liver disease. Toxicology 2018; 410:199-213. [DOI: 10.1016/j.tox.2018.08.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/06/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
|
24
|
Maejima Y, Yokota S, O'Hashi R, Aoyama M, Shimomura K. The effect of avertin anesthesia and a mixture of three types of anesthetic agents on food intakeand body weight in high fat-induced obese maleand female mice. Exp Anim 2018; 68:57-69. [PMID: 30210091 PMCID: PMC6389510 DOI: 10.1538/expanim.17-0145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Basic research on obesity is becoming more important due to an increasing number of obese
people. Experiments using obesity-model animals often require surgical interventions, such
as gastric operation, and proper selection of anesthesia is important. Avertin, an agent
mainly composed of 2,2,2-Tribromoethanol, has been used as general anesthesia for a long
time, without the use of narcotic drugs. In the current study, we found that a single
injection of avertin can decrease body weight (BW) in male and female C57BL/6J and ICR
mice with high fat-diet (HFD)-induced obesity, but not in standard diet-fed nonobese males
and females. Because the BW-reducing effect was more prominent in the female mice, we
compared the effects of avertin and a mixture of three types of anesthetic agents (3MIX),
which was developed in 2011, on BW reduction in HFD-induced obese female mice. Although
both avertin and 3MIX decreased food intake and BW, the effects of avertin were
significantly more potent than those of 3MIX. C-Fos expression, a neural activation
marker, was dramatically increased in the brain regions related to the regulation of both
food intake and the autonomic nervous system after avertin injection, but not after 3MIX
injection. This suggests that avertin strongly stimulates the center of feeding regulation
and the autonomic nervous system and therefore decreases BW. The current study suggests
the advantages of using 3MIX for surgical interventions in mice in obesity research, as it
is ideal to prevent anesthesia-induced BW decline.
Collapse
Affiliation(s)
- Yuko Maejima
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima-shi, Fukushima 960-1295, Japan
| | - Shoko Yokota
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima-shi, Fukushima 960-1295, Japan
| | - Rie O'Hashi
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima-shi, Fukushima 960-1295, Japan
| | - Masato Aoyama
- Department of Animal Science, Faculty of Agriculture, Utsunomiya University, 350 Minemachi, Utsunomiya-shi, Tochigi 321-8505, Japan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological Medicine, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima-shi, Fukushima 960-1295, Japan
| |
Collapse
|
25
|
Klaunig JE, Li X, Wang Z. Role of xenobiotics in the induction and progression of fatty liver disease. Toxicol Res (Camb) 2018; 7:664-680. [PMID: 30090613 PMCID: PMC6062016 DOI: 10.1039/c7tx00326a] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/09/2018] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease is a major cause of chronic liver pathology in humans. Fatty liver disease involves the accumulation of hepatocellular fat in hepatocytes that can progress to hepatitis. Steatohepatitis is categorized into alcoholic (ASH) or non-alcoholic (NASH) steatohepatitis based on the etiology of the insult. Both pathologies involve an initial steatosis followed by a progressive inflammation of the liver and eventual hepatic fibrosis (steatohepatitis) and cirrhosis. The involvement of pharmaceuticals and other chemicals in the initiation and progression of fatty liver disease has received increased study. This review will examine not only how xenobiotics initiate hepatic steatosis and steatohepatitis but also how the presence of fatty liver may modify the metabolism and pathologic effects of xenobiotics. The feeding of a high fat diet results in changes in the expression of nuclear receptors that are involved in adaptive and adverse liver effects following xenobiotic exposure. High fat diets also modulate cellular and molecular pathways involved in inflammation, metabolism, oxidative phosphorylation and cell growth. Understanding the role of hepatic steatosis and steatohepatitis on the sequelae of toxic and pathologic changes seen following xenobiotic exposure has importance in defining proper and meaningful human risk characterization of the drugs and other chemical agents.
Collapse
Affiliation(s)
- James E Klaunig
- Indiana University , School of Public Health , Bloomington , Indiana , USA .
| | - Xilin Li
- Indiana University , School of Public Health , Bloomington , Indiana , USA .
| | - Zemin Wang
- Indiana University , School of Public Health , Bloomington , Indiana , USA .
| |
Collapse
|