1
|
Lei JJ, Li S, Dong BX, Yang J, Ren Y. Acute intermittent porphyria: a disease with low penetrance and high heterogeneity. Front Genet 2024; 15:1374965. [PMID: 39188285 PMCID: PMC11345236 DOI: 10.3389/fgene.2024.1374965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024] Open
Abstract
Acute intermittent porphyria (AIP) is caused by mutations in the gene encoding hydroxymethylbilane synthase (HMBS), a key enzyme in the heme biosynthesis pathway. AIP is an autosomal dominant disorder characterized by low penetrance and a highly heterogenous clinical presentation. The estimated prevalence of AIP is 5-10 cases per 100,000 persons, with acute attacks manifesting in less than 1% of the at-risk population. This low frequency of attacks suggests significant roles for oligogenic inheritance and environmental factors in the pathogenesis of the disease. In recent years, identification of several modifier genes has advanced our understanding of the factors influencing AIP penetrance and disease severity. This review summarizes these factors including the impact of specific HMBS mutations, oligogenic inheritance, mitochondrial DNA copy number, age, sex, the influence of sex hormones, and the role of environmental factors. Further studies into the etiology of AIP disease penetrance should inform pathogenesis, potentially allowing for the development of more precise diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Jia-Jia Lei
- Department of First Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Shuang Li
- Department of First Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Bai-Xue Dong
- Department of First Clinical Medical School, Shanxi Medical University, Taiyuan, China
| | - Jing Yang
- Department of Endocrinology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yi Ren
- Department of Endocrinology, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Ko MY, Chon SH, Park H, Min E, Kim Y, Cha SW, Seo JW, Lee BS, Ka M, Hyun SA. Perfluorooctanoic acid induces cardiac dysfunction in human induced pluripotent stem cell-derived cardiomyocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 273:116170. [PMID: 38452704 DOI: 10.1016/j.ecoenv.2024.116170] [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: 11/24/2023] [Revised: 02/22/2024] [Accepted: 02/29/2024] [Indexed: 03/09/2024]
Abstract
Perfluorooctanoic acid (PFOA), commonly found in drinking water, leads to widespread exposure through skin contact, inhalation, and ingestion, resulting in detectable levels of PFOA in the bloodstream. In this study, we found that exposure to PFOA disrupts cardiac function in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). We observed reductions in field and action potentials in hiPSC-CMs exposed to PFOA. Furthermore, PFOA demonstrated a dose-dependent inhibitory effect on various ion channels, including the calcium, sodium, and potassium channels. Additionally, we noted dose-dependent inhibition of the expression of these ion channels in hiPSC-CMs following exposure to PFOA. These findings suggest that PFOA exposure can impair cardiac ion channel function and decrease the transcription of genes associated with these channels, potentially contributing to cardiac dysfunction such as arrhythmias. Our study sheds light on the electrophysiological and epigenetic consequences of PFOA-induced cardiac dysfunction, underscoring the importance of further research on the cardiovascular effects of perfluorinated compounds (PFCs).
Collapse
Affiliation(s)
- Moon Yi Ko
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Sun-Hwa Chon
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea; Graduate School of Pre-Clinical Laboratory Science, Konyang University, Daejeon 35365, Republic of Korea
| | - Heejin Park
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Euijun Min
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Younhee Kim
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Sin-Woo Cha
- Department of Nonclinical Studies, Korea Institute of Toxicology, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Joung-Wook Seo
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Byoung-Seok Lee
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea.
| | - Minhan Ka
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea.
| | - Sung-Ae Hyun
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea.
| |
Collapse
|
3
|
Pascale JV, Wolf A, Kadish Y, Diegisser D, Kulaprathazhe MM, Yemane D, Ali S, Kim N, Baruch DE, Yahaya MAF, Dirice E, Adebesin AM, Falck JR, Schwartzman ML, Garcia V. 20-Hydroxyeicosatetraenoic acid (20-HETE): Bioactions, receptors, vascular function, cardiometabolic disease and beyond. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 97:229-255. [PMID: 37236760 PMCID: PMC10683332 DOI: 10.1016/bs.apha.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
Vascular function is dynamically regulated and dependent on a bevy of cell types and factors that work in concert across the vasculature. The vasoactive eicosanoid, 20-Hydroxyeicosatetraenoic acid (20-HETE) is a key player in this system influencing the sensitivity of the vasculature to constrictor stimuli, regulating endothelial function, and influencing the renin angiotensin system (RAS), as well as being a driver of vascular remodeling independent of blood pressure elevations. Several of these bioactions are accomplished through the ligand-receptor pairing between 20-HETE and its high-affinity receptor, GPR75. This 20-HETE axis is at the root of various vascular pathologies and processes including ischemia induced angiogenesis, arteriogenesis, septic shock, hypertension, atherosclerosis, myocardial infarction and cardiometabolic diseases including diabetes and insulin resistance. Pharmacologically, several preclinical tools have been developed to disrupt the 20-HETE axis including 20-HETE synthesis inhibitors (DDMS and HET0016), synthetic 20-HETE agonist analogues (20-5,14-HEDE and 20-5,14-HEDGE) and 20-HETE receptor blockers (AAA and 20-SOLA). Systemic or cell-specific therapeutic targeting of the 20-HETE-GPR75 axis continues to be an invaluable approach as studies examine the molecular underpinnings activated by 20-HETE under various physiological settings. In particular, the development and characterization of 20-HETE receptor blockers look to be a promising new class of compounds that can provide a considerable benefit to patients suffering from these cardiovascular pathologies.
Collapse
Affiliation(s)
- Jonathan V Pascale
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Alexandra Wolf
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Yonaton Kadish
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - Danielle Diegisser
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | | | - Danait Yemane
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Samir Ali
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - Namhee Kim
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - David E Baruch
- School of Medicine, New York Medical College, Valhalla, NY, United States
| | - Muhamad Afiq Faisal Yahaya
- Department of Basic Sciences, MAHSA University, Selangor Darul Ehsan, Malaysia; Department of Human Anatomy, Universiti Putra Malaysia (UPM), Selangor Darul Ehsan, Malaysia
| | - Ercument Dirice
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Adeniyi M Adebesin
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Michal L Schwartzman
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States
| | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY, United States.
| |
Collapse
|
4
|
Shizu R, Ezaki K, Sato T, Sugawara A, Hosaka T, Sasaki T, Yoshinari K. PXR Suppresses PPARα-Dependent HMGCS2 Gene Transcription by Inhibiting the Interaction between PPARα and PGC1α. Cells 2021; 10:cells10123550. [PMID: 34944058 PMCID: PMC8700377 DOI: 10.3390/cells10123550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/13/2021] [Indexed: 01/11/2023] Open
Abstract
Background: PXR is a xenobiotic-responsive nuclear receptor that controls the expression of drug-metabolizing enzymes. Drug-induced activation of PXR sometimes causes drug–drug interactions due to the induced metabolism of co-administered drugs. Our group recently reported a possible drug–drug interaction mechanism via an interaction between the nuclear receptors CAR and PPARα. As CAR and PXR are structurally and functionally related receptors, we investigated possible crosstalk between PXR and PPARα. Methods: Human hepatocyte-like HepaRG cells were treated with various PXR ligands, and mRNA levels were determined by quantitative reverse transcription PCR. Reporter assays using the HMGCS2 promoter containing a PPARα-binding motif and mammalian two-hybrid assays were performed in HepG2 or COS-1 cells. Results: Treatment with PXR activators reduced the mRNA levels of PPARα target genes in HepaRG cells. In reporter assays, PXR suppressed PPARα-dependent gene expression in HepG2 cells. In COS-1 cells, co-expression of PGC1α, a common coactivator of PPARα and PXR, enhanced PPARα-dependent gene transcription, which was clearly suppressed by PXR. Consistently, in mammalian two-hybrid assays, the interaction between PGC1α and PPARα was attenuated by ligand-activated PXR. Conclusion: The present results suggest that ligand-activated PXR suppresses PPARα-dependent gene expression by inhibiting PGC1α recruitment.
Collapse
|
5
|
Pascale JV, Lucchesi PA, Garcia V. Unraveling the Role of 12- and 20- HETE in Cardiac Pathophysiology: G-Protein-Coupled Receptors, Pharmacological Inhibitors, and Transgenic Approaches. J Cardiovasc Pharmacol 2021; 77:707-717. [PMID: 34016841 PMCID: PMC8523029 DOI: 10.1097/fjc.0000000000001013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/03/2021] [Indexed: 12/17/2022]
Abstract
ABSTRACT Arachidonic acid-derived lipid mediators play crucial roles in the development and progression of cardiovascular diseases. Eicosanoid metabolites generated by lipoxygenases and cytochrome P450 enzymes produce several classes of molecules, including the epoxyeicosatrienoic acid (EET) and hydroxyeicosatetraenoic acids (HETE) family of bioactive lipids. In general, the cardioprotective effects of EETs have been documented across a number of cardiac diseases. In contrast, members of the HETE family have been shown to contribute to the pathogenesis of ischemic cardiac disease, maladaptive cardiac hypertrophy, and heart failure. The net effect of 12(S)- and 20-HETE depends upon the relative amounts generated, ratio of HETEs:EETs produced, timing of synthesis, as well as cellular and subcellular mechanisms activated by each respective metabolite. HETEs are synthesized by and affect multiple cell types within the myocardium. Moreover, cytochrome P450-derived and lipoxygenase- derived metabolites have been shown to directly influence cardiac myocyte growth and the regulation of cardiac fibroblasts. The mechanistic data uncovered thus far have employed the use of enzyme inhibitors, HETE antagonists, and the genetic manipulation of lipid-producing enzymes and their respective receptors, all of which influence a complex network of outcomes that complicate data interpretation. This review will summarize and integrate recent findings on the role of 12(S)-/20-HETE in cardiac diseases.
Collapse
Affiliation(s)
| | | | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY
| |
Collapse
|
6
|
Ma L, Tian Y, Peng C, Zhang Y, Zhang S. Recent advances in the epidemiology and genetics of acute intermittent porphyria. Intractable Rare Dis Res 2020; 9:196-204. [PMID: 33139978 PMCID: PMC7586877 DOI: 10.5582/irdr.2020.03082] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
Acute intermittent porphyria (AIP) is a dominant inherited disorder with a low penetrance that is caused by mutations in the gene coding for hydroxymethylbilane synthase (HMBS). Information about the epidemiology and molecular genetic features of this rare disorder is crucial to clinical research, and particularly to the evaluation of new treatments. Variations in the prevalence and penetrance of AIP in various studies may due to the different inclusion criteria and methods of assessment. Here, the prevalence and penetrance of AIP are analyzed systematically, and the genetic traits of different populations and findings regarding the genotype-phenotype correlation are summarized. In addition, quite a few studies have indicated that AIP susceptibility was affected by other factors, such as modifying genes. Findings regarding possible modifying genes are documented here, helping to reveal the pathogenesis of and treatments for AIP. The status of research on AIP in China reveals the lack of epidemiological and genetic studies of the Chinese population, a situation that needs to be promptly remedied.
Collapse
Affiliation(s)
- Liyan Ma
- Department of Endocrinology, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yu Tian
- Department of Endocrinology, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chenxing Peng
- Department of Endocrinology, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiran Zhang
- School of First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Songyun Zhang
- Department of Endocrinology, The second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
7
|
Moghadam ER, Ang HL, Asnaf SE, Zabolian A, Saleki H, Yavari M, Esmaeili H, Zarrabi A, Ashrafizadeh M, Kumar AP. Broad-Spectrum Preclinical Antitumor Activity of Chrysin: Current Trends and Future Perspectives. Biomolecules 2020; 10:E1374. [PMID: 32992587 PMCID: PMC7600196 DOI: 10.3390/biom10101374] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Pharmacological profile of phytochemicals has attracted much attention to their use in disease therapy. Since cancer is a major problem for public health with high mortality and morbidity worldwide, experiments have focused on revealing the anti-tumor activity of natural products. Flavonoids comprise a large family of natural products with different categories. Chrysin is a hydroxylated flavonoid belonging to the flavone category. Chrysin has demonstrated great potential in treating different disorders, due to possessing biological and therapeutic activities, such as antioxidant, anti-inflammatory, hepatoprotective, neuroprotective, etc. Over recent years, the anti-tumor activity of chrysin has been investigated, and in the present review, we provide a mechanistic discussion of the inhibitory effect of chrysin on proliferation and invasion of different cancer cells. Molecular pathways, such as Notch1, microRNAs, signal transducer and activator of transcription 3 (STAT3), nuclear factor-kappaB (NF-κB), PI3K/Akt, MAPK, etc., as targets of chrysin are discussed. The efficiency of chrysin in promoting anti-tumor activity of chemotherapeutic agents and suppressing drug resistance is described. Moreover, poor bioavailability, as one of the drawbacks of chrysin, is improved using various nanocarriers, such as micelles, polymeric nanoparticles, etc. This updated review will provide a direction for further studies in evaluating the anti-tumor activity of chrysin.
Collapse
Affiliation(s)
- Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran;
| | - Hui Li Ang
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
| | - Sholeh Etehad Asnaf
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, IslamicAzad University, Tehran 165115331, Iran;
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (H.S.); (H.E.)
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (H.S.); (H.E.)
| | - Mohammad Yavari
- Nursing and Midwifery Department, Islamic Azad University, Tehran Medical Sciences Branch, Tehran 1916893813, Iran;
| | - Hossein Esmaeili
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (H.S.); (H.E.)
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
| | - Milad Ashrafizadeh
- Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
| |
Collapse
|
8
|
Vacca M, Leslie J, Virtue S, Lam BYH, Govaere O, Tiniakos D, Snow S, Davies S, Petkevicius K, Tong Z, Peirce V, Nielsen MJ, Ament Z, Li W, Kostrzewski T, Leeming DJ, Ratziu V, Allison MED, Anstee QM, Griffin JL, Oakley F, Vidal-Puig A. Bone morphogenetic protein 8B promotes the progression of non-alcoholic steatohepatitis. Nat Metab 2020; 2:514-531. [PMID: 32694734 DOI: 10.1038/s42255-020-0214-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/24/2020] [Indexed: 12/15/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) is characterized by lipotoxicity, inflammation and fibrosis, ultimately leading to end-stage liver disease. The molecular mechanisms promoting NASH are poorly understood, and treatment options are limited. Here, we demonstrate that hepatic expression of bone morphogenetic protein 8B (BMP8B), a member of the transforming growth factor beta (TGFβ)-BMP superfamily, increases proportionally to disease stage in people and animal models with NASH. BMP8B signals via both SMAD2/3 and SMAD1/5/9 branches of the TGFβ-BMP pathway in hepatic stellate cells (HSCs), promoting their proinflammatory phenotype. In vivo, the absence of BMP8B prevents HSC activation, reduces inflammation and affects the wound-healing responses, thereby limiting NASH progression. Evidence is featured in primary human 3D microtissues modelling NASH, when challenged with recombinant BMP8. Our data show that BMP8B is a major contributor to NASH progression. Owing to the near absence of BMP8B in healthy livers, inhibition of BMP8B may represent a promising new therapeutic avenue for NASH treatment.
Collapse
Affiliation(s)
- Michele Vacca
- TVP Lab, WT/MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK.
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK.
| | - Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Samuel Virtue
- TVP Lab, WT/MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Brian Y H Lam
- Yeo Group and Genomics and Transcriptomics Core, WT/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Olivier Govaere
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Dina Tiniakos
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Department of Pathology, Aretaieion Hospital, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | | | - Susan Davies
- Liver Unit, Department of Medicine, Cambridge Biomedical Research Centre, Cambridge University Hospitals, Cambridge, UK
| | - Kasparas Petkevicius
- TVP Lab, WT/MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Zhen Tong
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Vivian Peirce
- TVP Lab, WT/MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | | | - Zsuzsanna Ament
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Wei Li
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | - Vlad Ratziu
- Sorbonne Université, Institute for Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, Paris, France
| | - Michael E D Allison
- Liver Unit, Department of Medicine, Cambridge Biomedical Research Centre, Cambridge University Hospitals, Cambridge, UK
| | - Quentin M Anstee
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- Biomolecular Medicine, Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Antonio Vidal-Puig
- TVP Lab, WT/MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit - Metabolic Research Laboratories, University of Cambridge, Cambridge, UK.
- Welcome Trust Sanger Institute, Hinxton, UK.
- Cambridge University Nanjing Centre of Technology and Innovation, Jiangbei Area, Nanjing, P R China.
| |
Collapse
|
9
|
The role of the microbiome and psychosocial stress in the expression and activity of drug metabolizing enzymes in mice. Sci Rep 2020; 10:8529. [PMID: 32444678 PMCID: PMC7244717 DOI: 10.1038/s41598-020-65595-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 05/06/2020] [Indexed: 12/21/2022] Open
Abstract
The gut microbiota is involved in a number of different metabolic processes of the host organism, including the metabolism of xenobiotics. In our study, we focused on liver cytochromes P450 (CYPs), which can metabolize a wide range of exo- and endogenous molecules. We studied changes in mRNA expression and CYP enzyme activities, as well as the mRNA expression of transcription factors that have an important role in CYP expression, all in stressed germ-free (GF) and stressed specific-pathogen-free (SPF) mice. Besides the presence of the gut microbiota, we looked at the difference between acute and chronic stress. Our results show that stress has an impact on CYP mRNA expression, but it is mainly chronic stress that has a significant effect on enzyme activities along with the gut microbiome. In acutely stressed mice, we observed significant changes at the mRNA level, however, the corresponding enzyme activities were not influenced. Our study suggests an important role of the gut microbiota along with chronic psychosocial stress in the expression and activity of CYPs, which can potentially lead to less effective drug metabolism and, as a result, a harmful impact on the organism.
Collapse
|
10
|
Khor CY, Khoo BY. PPARα plays an important role in the migration activity, and the expression of CYP2S1 and CYP1B1 in chrysin-treated HCT116 cells. Biotechnol Lett 2020; 42:1581-1595. [PMID: 32385743 DOI: 10.1007/s10529-020-02904-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/30/2020] [Indexed: 01/20/2023]
Abstract
OBJECTIVE This study aimed to examine the metabolising effect of chrysin by investigating the mRNA expression levels of PPARα and its related cellular mechanisms in HCT116 cells. RESULTS The mRNA expression of PPARα was significantly induced in HCT116 cells following treatment with chrysin for 36 h, but the mRNA expression of PPARα was inhibited, when the cells were treated with a combination of chrysin and MK886 (PPARα inhibitor). This phenomenon proved that the incorporation of MK886 lowers the expression levels of PPARα, thus enabling us to study the function of PPARα. The cell population of the G0/G1 phase significantly increased in chrysin-treated cells, which was accompanied by a decrease in the percentage of S phase cell population after 12 h of treatment. However, treatments of HCT116 cells with chrysin only or a combination of chrysin and MK886 did not show the opposite situation in the G0/G1 and S phase cell populations, indicating that the expression of PPARα may not be associated with the cell cycle in the treated cells. The migration rate in chrysin-treated HCT116 cells was reduced significantly after 24 and 36 h of treatments. However, the activity was revived, when the expression of PPARα was inhibited, indicating that the migration activity of chrysin-treated cells is likely correlated with the expression of PPARα. Comparison of the CYP2S1 and CYP1B1 mRNA expression in chrysin only treated, and a combination of chrysin and MK886-treated HCT116 cells for 24 and 36 h showed a significant difference in the expression levels, indicating that PPARα inhibitor could also modify the expression of CYP2S1 and CYP1B1. CONCLUSION The study indicates that PPARα may play an essential role in regulating the migration activity, and the expression of CYP2S1 and CYP1B1 in chrysin-treated colorectal cancer cells.
Collapse
Affiliation(s)
- Chin Yin Khor
- Institute for Research in Molecular Medicine (INFORMM), Universitit Sains Malaysia, 11800, Penang, Malaysia
| | - Boon Yin Khoo
- Institute for Research in Molecular Medicine (INFORMM), Universitit Sains Malaysia, 11800, Penang, Malaysia.
| |
Collapse
|
11
|
Li T, Hu S, Pang X, Wang J, Yin J, Li F, Wang J, Yang X, Xia B, Liu Y, Song W, Guo S. The marine-derived furanone reduces intracellular lipid accumulation in vitro by targeting LXRα and PPARα. J Cell Mol Med 2020; 24:3384-3398. [PMID: 31981312 PMCID: PMC7131916 DOI: 10.1111/jcmm.15012] [Citation(s) in RCA: 21] [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: 10/17/2019] [Revised: 12/30/2019] [Accepted: 01/10/2020] [Indexed: 12/22/2022] Open
Abstract
Recent studies have demonstrated that commercially available lipid-lowering drugs cause various side effects; therefore, searching for anti-hyperlipidaemic compounds with lower toxicity is a research hotspot. This study was designed to investigate whether the marine-derived compound, 5-hydroxy-3-methoxy-5-methyl-4-butylfuran-2(5H)-one, has an anti-hyperlipidaemic activity, and the potential underlying mechanism in vitro. Results showed that the furanone had weaker cytotoxicity compared to positive control drugs. In RAW 264.7 cells, the furanone significantly lowered ox-LDL-induced lipid accumulation (~50%), and its triglyceride (TG)-lowering effect was greater than that of liver X receptor (LXR) agonist T0901317. In addition, it significantly elevated the protein levels of peroxisome proliferator-activated receptors (PPARα) and ATP-binding cassette (ABC) transporters, which could be partially inhibited by LXR antagonists, GSK2033 and SR9243. In HepG2 cells, it significantly decreased oleic acid-induced lipid accumulation, enhanced the protein levels of low-density lipoprotein receptor (LDLR), ABCG5, ABCG8 and PPARα, and reduced the expression of sterol regulatory element-binding protein 2 (~32%). PPARα antagonists, GW6471 and MK886, could significantly inhibit the furanone-induced lipid-lowering effect. Furthermore, the furanone showed a significantly lower activity on the activation of the expression of lipogenic genes compared to T0901317. Taken together, the furanone exhibited a weak cytotoxicity but had powerful TC- and TG-lowering effects most likely through targeting LXRα and PPARα, respectively. These findings indicate that the furanone has a potential application for the treatment of dyslipidaemia.
Collapse
Affiliation(s)
- Ting Li
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Shu‐Mei Hu
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Xiao‐Yan Pang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Jun‐feng Wang
- CAS Key Laboratory of Tropical Marine Bio‐resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine MicrobiologySouth China Sea Institute of OceanologyChinese Academy of SciencesGuangzhouChina
| | - Jia‐Yu Yin
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Fa‐Hui Li
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Jin Wang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Xiao‐Qian Yang
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Bin Xia
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Yong‐Hong Liu
- CAS Key Laboratory of Tropical Marine Bio‐resources and Ecology/Guangdong Key Laboratory of Marine Materia Medica/RNAM Center for Marine MicrobiologySouth China Sea Institute of OceanologyChinese Academy of SciencesGuangzhouChina
| | - Wei‐Guo Song
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| | - Shou‐Dong Guo
- Institute of Lipid Metabolism and AtherosclerosisSchool of PharmacyInnovative Drug Research CentreWeifang Medical UniversityWeifangChina
| |
Collapse
|
12
|
Fu Y, Jia J, Yue L, Yang R, Guo Y, Ni X, Shi T. Systematically Analyzing the Pathogenic Variations for Acute Intermittent Porphyria. Front Pharmacol 2019; 10:1018. [PMID: 31572191 PMCID: PMC6753391 DOI: 10.3389/fphar.2019.01018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 08/09/2019] [Indexed: 12/18/2022] Open
Abstract
The rare autosomal dominant disorder acute intermittent porphyria (AIP) is caused by the deficient activity of hydroxymethylbilane synthase (HMBS). The symptoms of AIP are acute neurovisceral attacks which are induced by the dysfunction of heme biosynthesis. To better interpret the underlying mechanism of clinical phenotypes, we collected 117 HMBS gene mutations from reported individuals with AIP and evaluated the mutations' impacts on the corresponding protein structure and function. We found that several mutations with most severe clinical symptoms are located at dipyromethane cofactor (DPM) binding domain of HMBS. Mutations on these residues likely significantly influence the catalytic reaction. To infer new pathogenic mutations, we evaluated the pathogenicity for all the possible missense mutations of HMBS gene with different bioinformatic prediction algorithms, and identified 34 mutations with serious pathogenicity and low allele frequency. In addition, we found that gene PPARA may also play an important role in the mechanisms of AIP attacks. Our analysis about the distribution frequencies of the 23 variations revealed different distribution patterns among eight ethnic populations, which could help to explain the genetic basis that may contribute to population disparities in AIP prevalence. Our systematic analysis provides a better understanding for this disease and helps for the diagnosis and treatment of AIP.
Collapse
Affiliation(s)
- Yibao Fu
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Jinmeng Jia
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Lishu Yue
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Ruiying Yang
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yongli Guo
- Big Data and Engineering Research Center, Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Children’s Hospital, National Center for Children’s Health, Beijing Pediatric Research Institute, Capital Medical University, Beijing, China
- Biobank for Clinical Data and Samples in Pediatrics, Beijing Children’s Hospital, National Center for Children’s Health, Beijing Pediatric Research Institute, Capital Medical University, Beijing, China
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children’s Hospital, National Center for Children’s Health, Capital Medical University, Beijing, China
| | - Xin Ni
- Big Data and Engineering Research Center, Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, MOE Key Laboratory of Major Diseases in Children, Beijing Children’s Hospital, National Center for Children’s Health, Beijing Pediatric Research Institute, Capital Medical University, Beijing, China
- Biobank for Clinical Data and Samples in Pediatrics, Beijing Children’s Hospital, National Center for Children’s Health, Beijing Pediatric Research Institute, Capital Medical University, Beijing, China
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children’s Hospital, National Center for Children’s Health, Capital Medical University, Beijing, China
| | - Tieliu Shi
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| |
Collapse
|
13
|
Lv N, Yuan J, Ji A, Shi L, Gao M, Cui L, Jiang Q. Perfluorooctanoic acid-induced toxicities in chicken embryo primary cardiomyocytes: Roles of PPAR alpha and Wnt5a/Frizzled2. Toxicol Appl Pharmacol 2019; 381:114716. [PMID: 31445018 DOI: 10.1016/j.taap.2019.114716] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/25/2019] [Accepted: 08/18/2019] [Indexed: 01/06/2023]
Abstract
Perfluorooctanoic acid (PFOA) is a widespread persistent organic pollutant and may induce developmental toxicities, including developmental cardiotoxicity. To explore the potential mechanism of developmental cardiotoxicity induced by PFOA exposure, chicken embryo primary cardiomyocytes were extracted either from chicken embryos pretreated with PFOA (2 mg/kg), or from untreated embryos and then directly exposed cells to PFOA (1, 10, 30 or 100 μg/ml) in culture. Additionally, peroxisome proliferator activated receptor alpha (PPAR alpha) silencing lentivirus was applied to the embryos on embryonic day (ED2). Cell viability was measured with CCK-8 kit, morphology was assessed with hematoxylin and eosin staining, and intracellular Ca2+ concentrations were determined with Fluo-4 AM probe. Western blotting was utilized to confirm PPAR alpha silencing efficiency and the protein abundance of Wnt5a and Frizzled2. The results indicated that both PFOA pretreatment and direct exposure decreased primary cardiomyocyte viability, altered cell morphology and increased intracellular Ca2+ concentrations. While l-carnitine co-treatment effectively abolished such changes, PPAR alpha silencing only abolished most of the changes in PFOA pretreatment group, but not in cells directly exposed to relatively high doses of PFOA. The protein abundance of Wnt5a and Frizzled2 was increased by PFOA pretreatment, while direct exposure to PFOA increased Frizzled2 abundance but decreased Wnt5a abundance. PPAR alpha silencing resulted in over 50% decrease of PPAR alpha expression level, which abolished the Wnt5a/Frizzled2 expression alterations following PFOA exposure. In conclusion, PFOA-induced primary cardiomyocyte toxicity is associated with PPAR alpha and Wnt5a/Frizzled2, in which PPAR alpha seems to play regulatory roles towards Wnt5a/Frizzled2.
Collapse
Affiliation(s)
- Na Lv
- Department of Pharmacology, School of Pharmacy, Qingdao University, China
| | - Junhua Yuan
- Department of Special Medicine, School of Basic Medicine, Qingdao University, China
| | - Andong Ji
- Department of Toxicology, School of Public Health, Qingdao University, China
| | - Limei Shi
- Department of Toxicology, School of Public Health, Qingdao University, China
| | - Mengyu Gao
- Department of Toxicology, School of Public Health, Qingdao University, China
| | - Lianhua Cui
- Department of Toxicology, School of Public Health, Qingdao University, China
| | - Qixiao Jiang
- Department of Toxicology, School of Public Health, Qingdao University, China.
| |
Collapse
|
14
|
Klag T, Thomas M, Ehmann D, Courth L, Mailänder-Sanchez D, Weiss TS, Dayoub R, Abshagen K, Vollmar B, Thasler WE, Stange EF, Berg CP, Malek NP, Zanger UM, Wehkamp J. β-Defensin 1 Is Prominent in the Liver and Induced During Cholestasis by Bilirubin and Bile Acids via Farnesoid X Receptor and Constitutive Androstane Receptor. Front Immunol 2018; 9:1735. [PMID: 30100908 PMCID: PMC6072844 DOI: 10.3389/fimmu.2018.01735] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 07/13/2018] [Indexed: 12/12/2022] Open
Abstract
Background & aims Knowledge about innate antimicrobial defense of the liver is limited. We investigated hepatic expression and regulation of antimicrobial peptides with focus on the human beta defensin-1 (hBD-1). Methods Radial diffusion assay was used to analyze antimicrobial activity of liver tissue. Different defensins including hBD-1 and its activator thioredoxin-1 (TXN) were analyzed in healthy and cholestatic liver samples by qPCR and immunostaining. Regulation of hBD-1 expression was studied in vitro and in vivo using bile duct-ligated mice. Regulation of hBD-1 via bilirubin and bile acids (BAs) was studied using siRNA. Results We found strong antimicrobial activity of liver tissue against Escherichia coli. As a potential mediator of this antimicrobial activity we detected high expression of hBD-1 and TXN in hepatocytes, whereas other defensins were minimally expressed. Using a specific antibody for the reduced, antimicrobially active form of hBD-1 we found hBD-1 in co-localization with TXN within hepatocytes. hBD-1 was upregulated in cholestasis in a graded fashion. In cholestatic mice hepatic AMP expression (Defb-1 and Hamp) was enhanced. Bilirubin and BAs were able to induce hBD-1 in hepatic cell cultures in vitro. Treatment with siRNA and/or agonists demonstrated that the farnesoid X receptor (FXR) mediates basal expression of hBD-1, whereas both constitutive androstane receptor (CAR) and FXR seem to be responsible for the induction of hBD-1 by bilirubin. Conclusion hBD-1 is prominently expressed in hepatocytes. It is induced during cholestasis through bilirubin and BAs, mediated by CAR and especially FXR. Reduction by TXN activates hBD-1 to a potential key player in innate antimicrobial defense of the liver.
Collapse
Affiliation(s)
- Thomas Klag
- Department of Internal Medicine I, University of Tübingen, Tübingen, Germany
| | - Maria Thomas
- Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, Stuttgart and University of Tuebingen, Tuebingen, Germany
| | - Dirk Ehmann
- Department of Internal Medicine I, University of Tübingen, Tübingen, Germany
| | - Lioba Courth
- Department of Internal Medicine I, University of Tübingen, Tübingen, Germany
| | | | - Thomas S Weiss
- University Children Hospital (KUNO), Regensburg University Hospital, Regensburg, Germany
| | - Rania Dayoub
- University Children Hospital (KUNO), Regensburg University Hospital, Regensburg, Germany
| | - Kerstin Abshagen
- Rudolf-Zenker-Institute for Experimental Surgery, University Medicine Rostock, Rostock, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute for Experimental Surgery, University Medicine Rostock, Rostock, Germany
| | - Wolfgang E Thasler
- Department of Surgery, Grosshadern Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Eduard F Stange
- Department of Internal Medicine I, University of Tübingen, Tübingen, Germany
| | - Christoph P Berg
- Department of Internal Medicine I, University of Tübingen, Tübingen, Germany
| | - Nisar P Malek
- Department of Internal Medicine I, University of Tübingen, Tübingen, Germany
| | - Ulrich M Zanger
- Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, Stuttgart and University of Tuebingen, Tuebingen, Germany
| | - Jan Wehkamp
- Department of Internal Medicine I, University of Tübingen, Tübingen, Germany
| |
Collapse
|
15
|
Shi C, Min L, Yang J, Dai M, Song D, Hua H, Xu G, Gonzalez FJ, Liu A. Peroxisome Proliferator-Activated Receptor α Activation Suppresses Cytochrome P450 Induction Potential in Mice Treated with Gemfibrozil. Basic Clin Pharmacol Toxicol 2017; 121:169-174. [PMID: 28374976 DOI: 10.1111/bcpt.12794] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/29/2017] [Indexed: 12/19/2022]
Abstract
Gemfibrozil, a peroxisome proliferator-activated receptor α (PPARα) agonist, is widely used for hypertriglyceridaemia and mixed hyperlipidaemia. Drug-drug interaction of gemfibrozil and other PPARα agonists has been reported. However, the role of PPARα in cytochrome P450 (CYP) induction by fibrates is not well known. In this study, wild-type mice were first fed gemfibrozil-containing diets (0.375%, 0.75% and 1.5%) for 14 days to establish a dose-response relationship for CYP induction. Then, wild-type mice and Pparα-null mice were treated with a 0.75% gemfibrozil-containing diet for 7 days. CYP3a, CYP2b and CYP2c were induced in a dose-dependent manner by gemfibrozil. In Pparα-null mice, their mRNA level, protein level and activity were induced more than those in wild-type mice. So, gemfibrozil induced CYP, and this action was inhibited by activated PPARα. These data suggested that the induction potential of CYPs was suppressed by activated PPARα, showing a potential role of this receptor in drug-drug interactions and metabolic diseases treated with fibrates.
Collapse
Affiliation(s)
- Cunzhong Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Luo Min
- Medical School of Ningbo University, Ningbo, China
| | - Julin Yang
- Ningbo College of Health Sciences, Ningbo, China
| | - Manyun Dai
- Medical School of Ningbo University, Ningbo, China
| | - Danjun Song
- Medical School of Ningbo University, Ningbo, China
| | - Huiying Hua
- Medical School of Ningbo University, Ningbo, China
| | - Gangming Xu
- Medical School of Ningbo University, Ningbo, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, NIH, Bethesda, USA
| | - Aiming Liu
- Medical School of Ningbo University, Ningbo, China
| |
Collapse
|
16
|
Zhong S, Han W, Hou C, Liu J, Wu L, Liu M, Liang Z, Lin H, Zhou L, Liu S, Tang L. Relation of Transcriptional Factors to the Expression and Activity of Cytochrome P450 and UDP-Glucuronosyltransferases 1A in Human Liver: Co-Expression Network Analysis. AAPS JOURNAL 2016; 19:203-214. [PMID: 27681103 DOI: 10.1208/s12248-016-9990-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/08/2016] [Indexed: 12/31/2022]
Abstract
Cytochrome P450 (CYPs) and UDP-glucuronosyltransferases (UGTs) play important roles in the metabolism of exogenous and endogenous compounds. The gene transcription of CYPs and UGTs can be enhanced or reduced by transcription factors (TFs). This study aims to explore novel TFs involved in the regulatory network of human hepatic UGTs/CYPs. Correlations between the transcription levels of 683 key TFs and CYPs/UGTs in three different human liver expression profiles (n = 640) were calculated first. Supervised weighted correlation network analysis (sWGCNA) was employed to define hub genes among the selected TFs. The relationship among 17 defined TFs, CYPs/UGTs expression, and activity were evaluated in 30 liver samples from Chinese patients. The positive controls (e.g., PPARA, NR1I2, NR1I3) and hub TFs (NFIA, NR3C2, and AR) in the GreysWGCNA Module were significantly and positively associated with CYPs/UGTs expression. And the cancer- or inflammation-related TFs (TEAD4, NFKB2, and NFKB1) were negatively associated with mRNA expression of CYP2C9/CYP2E1/UGT1A9. Furthermore, the effect of NR1I2, NR1I3, AR, TEAD4, and NFKB2 on CYP450/UGT1A gene transcription translated into moderate influences on enzyme activities. To our knowledge, this is the first study to integrate Gene Expression Omnibus (GEO) datasets and supervised weighted correlation network analysis (sWGCNA) for defining TFs potentially related to CYPs/UGTs. We detected several novel TFs involved in the regulatory network of hepatic CYPs and UGTs in humans. Further validation and investigation may reveal their exact mechanism of CYPs/UGTs regulation.
Collapse
Affiliation(s)
- Shilong Zhong
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Southern Medical University, Guangzhou, China.,Medical Research Center of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510080, China
| | - Weichao Han
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, Department of Biopharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Chuqi Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, Department of Biopharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Junjin Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, Department of Biopharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lili Wu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, Department of Biopharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Menghua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, Department of Biopharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhi Liang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, Department of Biopharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Haoming Lin
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Southern Medical University, Guangzhou, China
| | - Shuwen Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Southern Medical University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of New Drug Screening, Department of Biopharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Lan Tang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Southern Medical University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of New Drug Screening, Department of Biopharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|