1
|
Choi YJ, Kim Y, Hwang S. Role of Neutrophils in the Development of Steatotic Liver Disease. Semin Liver Dis 2024; 44:300-318. [PMID: 39117322 DOI: 10.1055/s-0044-1789207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
This review explores the biological aspects of neutrophils, their contributions to the development of steatotic liver disease, and their potential as therapeutic targets for the disease. Although alcohol-associated and metabolic dysfunction-associated liver diseases originate from distinct etiological factors, the two diseases frequently share excessive lipid accumulation as a common contributor to their pathogenesis, thereby classifying them as types of steatotic liver disease. Dysregulated lipid deposition in the liver induces hepatic injury, triggering the activation of the innate immunity, partially through neutrophil recruitment. Traditionally recognized for their role in microbial clearance, neutrophils have recently garnered attention for their involvement in sterile inflammation, a pivotal component of steatotic liver disease pathogenesis. In conclusion, technological innovations, including single-cell RNA sequencing, have gradually disclosed the existence of various neutrophil subsets; however, how the distinct subsets of neutrophil population contribute differentially to the development of steatotic liver disease remains unclear.
Collapse
Affiliation(s)
- You-Jin Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan, Republic of Korea
| | - Yeonsoo Kim
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
2
|
Spathakis M, Dovrolis N, Filidou E, Kandilogiannakis L, Tarapatzi G, Valatas V, Drygiannakis I, Paspaliaris V, Arvanitidis K, Manolopoulos VG, Kolios G, Vradelis S. Exploring Microbial Metabolite Receptors in Inflammatory Bowel Disease: An In Silico Analysis of Their Potential Role in Inflammation and Fibrosis. Pharmaceuticals (Basel) 2024; 17:492. [PMID: 38675452 PMCID: PMC11054721 DOI: 10.3390/ph17040492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolites produced by dysbiotic intestinal microbiota can influence disease pathophysiology by participating in ligand-receptor interactions. Our aim was to investigate the differential expression of metabolite receptor (MR) genes between inflammatory bowel disease (IBD), healthy individuals (HIs), and disease controls in order to identify possible interactions with inflammatory and fibrotic pathways in the intestine. RNA-sequencing datasets containing 643 Crohn's disease (CD) patients, 467 ulcerative colitis (UC) patients and 295 HIs, and 4 Campylobacter jejuni-infected individuals were retrieved from the Sequence Read Archive, and differential expression was performed using the RaNA-seq online platform. The identified differentially expressed MR genes were used for correlation analysis with up- and downregulated genes in IBD, as well as functional enrichment analysis using a R based pipeline. Overall, 15 MR genes exhibited dysregulated expression in IBD. In inflamed CD, the hydroxycarboxylic acid receptors 2 and 3 (HCAR2, HCAR3) were upregulated and were associated with the recruitment of innate immune cells, while, in the non-inflamed CD ileum, the cannabinoid receptor 1 (CNR1) and the sphingosine-1-phospate receptor 4 (S1PR4) were downregulated and were involved in the regulation of B-cell activation. In inflamed UC, the upregulated receptors HCAR2 and HCAR3 were more closely associated with the process of TH-17 cell differentiation, while the pregnane X receptor (NR1I2) and the transient receptor potential vanilloid 1 (TRPV1) were downregulated and were involved in epithelial barrier maintenance. Our results elucidate the landscape of metabolite receptor expression in IBD, highlighting associations with disease-related functions that could guide the development of new targeted therapies.
Collapse
Affiliation(s)
- Michail Spathakis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.S.); (E.F.); (L.K.); (G.T.); (V.V.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Nikolas Dovrolis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.S.); (E.F.); (L.K.); (G.T.); (V.V.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Eirini Filidou
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.S.); (E.F.); (L.K.); (G.T.); (V.V.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Leonidas Kandilogiannakis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.S.); (E.F.); (L.K.); (G.T.); (V.V.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Gesthimani Tarapatzi
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.S.); (E.F.); (L.K.); (G.T.); (V.V.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Vassilis Valatas
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.S.); (E.F.); (L.K.); (G.T.); (V.V.); (K.A.); (V.G.M.); (G.K.)
- Gastroenterology and Hepatology Research Laboratory, Medical School, University of Crete, 71003 Heraklion, Greece;
| | - Ioannis Drygiannakis
- Gastroenterology and Hepatology Research Laboratory, Medical School, University of Crete, 71003 Heraklion, Greece;
| | | | - Konstantinos Arvanitidis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.S.); (E.F.); (L.K.); (G.T.); (V.V.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Vangelis G. Manolopoulos
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.S.); (E.F.); (L.K.); (G.T.); (V.V.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - George Kolios
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (M.S.); (E.F.); (L.K.); (G.T.); (V.V.); (K.A.); (V.G.M.); (G.K.)
- Individualised Medicine & Pharmacological Research Solutions Center (IMPReS), 68100 Alexandroupolis, Greece
| | - Stergios Vradelis
- Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| |
Collapse
|
3
|
Qian L, Zhu Y, Deng C, Liang Z, Chen J, Chen Y, Wang X, Liu Y, Tian Y, Yang Y. Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family in physiological and pathophysiological process and diseases. Signal Transduct Target Ther 2024; 9:50. [PMID: 38424050 PMCID: PMC10904817 DOI: 10.1038/s41392-024-01756-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family (PGC-1s), consisting of three members encompassing PGC-1α, PGC-1β, and PGC-1-related coactivator (PRC), was discovered more than a quarter-century ago. PGC-1s are essential coordinators of many vital cellular events, including mitochondrial functions, oxidative stress, endoplasmic reticulum homeostasis, and inflammation. Accumulating evidence has shown that PGC-1s are implicated in many diseases, such as cancers, cardiac diseases and cardiovascular diseases, neurological disorders, kidney diseases, motor system diseases, and metabolic disorders. Examining the upstream modulators and co-activated partners of PGC-1s and identifying critical biological events modulated by downstream effectors of PGC-1s contribute to the presentation of the elaborate network of PGC-1s. Furthermore, discussing the correlation between PGC-1s and diseases as well as summarizing the therapy targeting PGC-1s helps make individualized and precise intervention methods. In this review, we summarize basic knowledge regarding the PGC-1s family as well as the molecular regulatory network, discuss the physio-pathological roles of PGC-1s in human diseases, review the application of PGC-1s, including the diagnostic and prognostic value of PGC-1s and several therapies in pre-clinical studies, and suggest several directions for future investigations. This review presents the immense potential of targeting PGC-1s in the treatment of diseases and hopefully facilitates the promotion of PGC-1s as new therapeutic targets.
Collapse
Affiliation(s)
- Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, 450052, China
| | - Junmin Chen
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xue Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yanqing Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ye Tian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China.
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
4
|
Tumenbayar BI, Tutino VM, Brazzo JA, Yao P, Bae Y. FAK and p130Cas modulate stiffness-mediated early transcription and cellular metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575789. [PMID: 38293187 PMCID: PMC10827115 DOI: 10.1101/2024.01.15.575789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Cellular metabolism is influenced by the stiffness of the extracellular matrix. Focal adhesion kinase (FAK) and its binding partner, p130Cas, transmit biomechanical signals about substrate stiffness to the cell to regulate a variety of cellular responses, but their roles in early transcriptional and metabolic responses remain largely unexplored. We cultured mouse embryonic fibroblasts with or without siRNA-mediated FAK or p130Cas knockdown and assessed the early transcriptional responses of these cells to placement on soft and stiff substrates by RNA sequencing and bioinformatics analyses. Exposure to the stiff ECM altered the expression of genes important for metabolic and biosynthetic processes, and these responses were influenced by knockdown of FAK and p130Cas. Our findings reveal that FAK-p130Cas signaling mechanotransduces ECM stiffness to early transcriptional changes that alter cellular metabolism and biosynthesis.
Collapse
Affiliation(s)
- Bat-Ider Tumenbayar
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Vincent M. Tutino
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, NY 14260, USA
- Department of Neurosurgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Joseph A. Brazzo
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Peng Yao
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, NY 14260, USA
| |
Collapse
|
5
|
Guo Y, Liu B, Geng Y, Chen K, Li J, Yin X, Zhang S. Fisetin, a dietary flavonoid, promotes transintestinal cholesterol excretion through the activation of PPARδ. Food Res Int 2024; 175:113783. [PMID: 38129008 DOI: 10.1016/j.foodres.2023.113783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/14/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023]
Abstract
Fisetin, a dietary polyphenol abundantly found in strawberries, exhibits a broad spectrum of health-promoting activities, including antihyperlipidemic effects. This study aimed to investigate the regulatory effect of fisetin on cholesterol elimination through novel transintestinal cholesterol excretion (TICE) pathway. A hypercholesterolemic mouse model and human colon epithelial cancer cell line Caco-2 were utilized to conduct the study. In hypercholesterolemic mice, fisetin (25 mg/kg) treatment reduced serum total cholesterol by 46.48% and significantly decreased lipid accumulation in the liver. Furthermore, fisetin administration led to a substantial increase in the fecal neutral sterol contents, including coprostanol, coprostanone, dihydrocholesterol, and cholesterol. Specifically, these sterol contents increased by approximately 224.20%, 151.40%, 70.40% and 50.72% respectively. The fluorescence intensity of 22-NBD-cholesterol in intestinal perfusion increased by 95.94% in fisetin group (25 mg/kg), indicating that fisetin stimulated TICE. In high cholesterol-induced Caco-2 cells, fisetin at a concentration of 30 μM reduced total cholesterol and free cholesterol by 37.21% and 45.30% respectively, stimulated cholesterol excretion, and inhibited cholesterol accumulation. Additionally, fisetin upregulated the gene and protein expression of cholesterol efflux transporters ABCG5/G8 and ABCB1, while downregulating the cholesterol uptake regulator NPC1L1. Furthermore, fisetin increased LDLR protein expression and decreased PCSK9 expression. Notably, fisetin significantly activated nuclear receptor PPARδ in Caco-2 cells. PPARδ antagonist pretreatment counteracted the regulatory effects of fisetin on TICE regulators, suggesting fisetin lowered cholesterol through enhancing TICE by activation of intestinal PPARδ. Fisetin could be used as functional dietarysupplement for eliminating cholesterol and reducing the incidence of cardiovascular diseases.
Collapse
Affiliation(s)
- Yao Guo
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Bing Liu
- College of Public Health, Zhengzhou University, Zhengzhou, China; Food Laboratory of Zhongyuan, Luohe, China
| | - Yaping Geng
- Division of Infectious Disease Prevention and Control, Binzhou Center for Disease Control and Prevention, Shandong, China
| | - Ke Chen
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Junyan Li
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Xiangju Yin
- Institute of Resources and Environment, Henan Polytechnic University, Jiaozuo, China
| | - Shenshen Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, China; Food Laboratory of Zhongyuan, Luohe, China; Nutrition and Health Food Research Institute, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
6
|
Barragán R, Zuraikat FM, Cheng B, Scaccia SE, Cochran J, Aggarwal B, Jelic S, St‐Onge M. Paradoxical Effects of Prolonged Insufficient Sleep on Lipid Profile: A Pooled Analysis of 2 Randomized Trials. J Am Heart Assoc 2023; 12:e032078. [PMID: 37815115 PMCID: PMC10757551 DOI: 10.1161/jaha.123.032078] [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: 08/04/2023] [Accepted: 08/15/2023] [Indexed: 10/11/2023]
Abstract
Background Insufficient sleep is associated with increased cardiovascular disease risk, but causality is unclear. We investigated the impact of prolonged mild sleep restriction (SR) on lipid and inflammatory profiles. Methods and Results Seventy-eight participants (56 women [12 postmenopausal]; age, 34.3±12.5 years; body mass index, 25.8±3.5 kg/m2) with habitual sleep duration 7 to 9 h/night (adequate sleep [AS]) underwent two 6-week conditions in a randomized crossover design: AS versus SR (AS-1.5 h/night). Total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol, triglycerides, and inflammatory markers (CRP [C-reactive protein], interleukin 6, and tumor necrosis factor-α) were assessed. Linear models tested effects of SR on outcomes in the full sample and by sex+menopausal status (premenopausal versus postmenopausal women+men). In the full sample, SR increased high-density lipoprotein cholesterol compared with AS (β=1.2±0.5 mg/dL; P=0.03). Sex+menopausal status influenced the effects of SR on change in total cholesterol (P-interaction=0.04), LDL-C (P-interaction=0.03), and interleukin 6 (P-interaction=0.07). Total cholesterol and LDL-C decreased in SR versus AS in premenopausal women (total cholesterol: β=-4.2±1.9 mg/dL; P=0.03; LDL-C: β=-6.3±2.0 mg/dL; P=0.002). Given paradoxical effects of SR on cholesterol concentrations, we explored associations between changes in inflammation and end point lipids under each condition. Increases in interleukin 6 and tumor necrosis factor-α during SR tended to relate to lower LDL-C in premenopausal women (interleukin 6: β=-5.3±2.6 mg/dL; P=0.051; tumor necrosis factor-α: β=-32.8±14.2 mg/dL; P=0.027). Conclusions Among healthy adults, prolonged insufficient sleep does not increase atherogenic lipids. However, increased inflammation in SR tends to predict lower LDL-C in premenopausal women, resembling the "lipid paradox" in which low cholesterol associates with increased cardiovascular disease risk in proinflammatory conditions. Registration URL: https://www.clinicaltrials.gov; Unique identifiers: NCT02835261, NCT02960776.
Collapse
Affiliation(s)
- Rocío Barragán
- Department of Preventive Medicine and Public HealthUniversity of ValenciaValenciaSpain
- Centro de Investigación Biomédica En Red Fisiopatología de la Obesidad y NutriciónInstituto de Salud Carlos IIIMadridSpain
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
| | - Faris M. Zuraikat
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of General Medicine, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
- New York Nutrition Obesity Research CenterColumbia University Irving Medical CenterNew YorkNY
| | - Bin Cheng
- Department of Biostatistics, Mailman School of Public HealthColumbia University Irving Medical CenterNew YorkNY
| | - Samantha E. Scaccia
- Division of Cardiology, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
| | - Justin Cochran
- Department of SurgeryColumbia University Irving Medical CenterNew YorkNY
| | - Brooke Aggarwal
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of Cardiology, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
| | - Sanja Jelic
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
| | - Marie‐Pierre St‐Onge
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of General Medicine, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
- New York Nutrition Obesity Research CenterColumbia University Irving Medical CenterNew YorkNY
| |
Collapse
|
7
|
Iyer DR, Venkatraman J, Tanguy E, Vitale N, Mahapatra NR. Chromogranin A and its derived peptides: potential regulators of cholesterol homeostasis. Cell Mol Life Sci 2023; 80:271. [PMID: 37642733 PMCID: PMC11072126 DOI: 10.1007/s00018-023-04908-3] [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: 03/03/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/31/2023]
Abstract
Chromogranin A (CHGA), a member of the granin family of proteins, has been an attractive therapeutic target and candidate biomarker for several cardiovascular, neurological, and inflammatory disorders. The prominence of CHGA stems from the pleiotropic roles of several bioactive peptides (e.g., catestatin, pancreastatin, vasostatins) generated by its proteolytic cleavage and by their wide anatomical distribution. These peptides are emerging as novel modulators of cardiometabolic diseases that are often linked to high blood cholesterol levels. However, their impact on cholesterol homeostasis is poorly understood. The dynamic nature of cholesterol and its multitudinous roles in almost every aspect of normal body function makes it an integral component of metabolic physiology. A tightly regulated coordination of cholesterol homeostasis is imperative for proper functioning of cellular and metabolic processes. The deregulation of cholesterol levels can result in several pathophysiological states. Although studies till date suggest regulatory roles for CHGA and its derived peptides on cholesterol levels, the mechanisms by which this is achieved still remain unclear. This review aims to aggregate and consolidate the available evidence linking CHGA with cholesterol homeostasis in health and disease. In addition, we also look at common molecular regulatory factors (viz., transcription factors and microRNAs) which could govern the expression of CHGA and genes involved in cholesterol homeostasis under basal and pathological conditions. In order to gain further insights into the pathways mediating cholesterol regulation by CHGA/its derived peptides, a few prospective signaling pathways are explored, which could act as primers for future studies.
Collapse
Affiliation(s)
- Dhanya R Iyer
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Janani Venkatraman
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| | - Emeline Tanguy
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives, CNRS UPR 3212 and Université de Strasbourg, 5 Rue Blaise Pascal, 67000, Strasbourg, France.
| | - Nitish R Mahapatra
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India.
| |
Collapse
|
8
|
Abstract
COVID-19 infections decrease total cholesterol, LDL-C, HDL-C, and apolipoprotein A-I, A-II, and B levels while triglyceride levels may be increased or inappropriately normal for the poor nutritional status. The degree of reduction in total cholesterol, LDL-C, HDL-C, and apolipoprotein A-I are predictive of mortality. With recovery lipid/lipoprotein levels return towards pre-infection levels and studies have even suggested an increased risk of dyslipidemia post-COVID-19 infection. The potential mechanisms for these changes in lipid and lipoprotein levels are discussed. Decreased HDL-C and apolipoprotein A-I levels measured many years prior to COVID-19 infections are associated with an increased risk of severe COVID-19 infections while LDL-C, apolipoprotein B, Lp (a), and triglyceride levels were not consistently associated with an increased risk. Finally, data suggest that omega-3-fatty acids and PCSK9 inhibitors may reduce the severity of COVID-19 infections. Thus, COVID-19 infections alter lipid/lipoprotein levels and HDL-C levels may affect the risk of developing COVID-19 infections.
Collapse
|
9
|
Guo X, Hu J, He G, Chen J, Yang Y, Qin D, Li C, Huang Z, Hu D, Wei C, Wang F, Yu B. Loss of APOO (MIC26) aggravates obesity-related whitening of brown adipose tissue via PPARα-mediated functional interplay between mitochondria and peroxisomes. Metabolism 2023; 144:155564. [PMID: 37088120 DOI: 10.1016/j.metabol.2023.155564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND Mitochondrial dysfunction and aberrant structure in adipose tissue occur in obesity and obesity-linked brown adipose tissue (BAT) whitening; however, whether this aberrant architecture contributes to or is the result of obesity is unknown. Apolipoprotein O (APOO) is a constitutive protein of the mitochondrial cristae organizing system complex. This study aimed to characterize the physiological consequences of APOO deficiency in vivo. METHODS APOO expression was analyzed in different human and murine adipose depots, and mice lacking APOO in adipocytes (ApooACKO) are developed to examine the metabolic consequences of adipocyte-specific APOO ablation in vitro and in vivo. RESULTS Results showed that APOO expression is reduced in BAT from both diet-induced and leptin-deficient obese mice. APOO-knockout mice showed increased adiposity, BAT dysfunction and whitening, reduced non-shivering thermogenesis, and blunted responses to cold stimuli. APOO deficiency disrupted mitochondrial structure in brown adipocytes and impaired oxidative phosphorylation, thereby inducing a shift from oxidative to glycolytic metabolism, increasing lipogenic enzyme levels and BAT whitening. APOO inactivation inhibited thermogenesis in BAT by reducing mitochondrial long-chain fatty acid oxidation. It also disturbed peroxisomal biogenesis and very long-chain fatty acid oxidation via peroxisome proliferator-activated receptor α. CONCLUSIONS Altogether, APOO deficiency in adipocytes aggravates BAT whitening and diet-induced obesity; thus, APOO could be a therapeutic target for obesity.
Collapse
Affiliation(s)
- Xin Guo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China
| | - Jiarui Hu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China
| | - Guangxu He
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China
| | - Jin Chen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China
| | - Yang Yang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China
| | - Donglu Qin
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China
| | - Chenyu Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China
| | - Zhijie Huang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China
| | - Die Hu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China
| | - Cheng Wei
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China
| | - Fengjiao Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China
| | - Bilian Yu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Research Institute of Blood Lipid and Atherosclerosis, Central South University, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China; Hunan Key Laboratory of Cardiometabolic Medicine, No. 139 Middle Renmin Road, Changsha 410011, Hunan, China.
| |
Collapse
|
10
|
Nhoek P, Ahn S, Pel P, Kim YM, Huh J, Kim HW, Noh M, Chin YW. Alkaloids and Coumarins with Adiponectin-Secretion-Promoting Activities from the Leaves of Orixa japonica. JOURNAL OF NATURAL PRODUCTS 2023; 86:138-148. [PMID: 36529937 DOI: 10.1021/acs.jnatprod.2c00844] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Fractionation of a methanol extract of Orixa japonica leaves led to the identification of five new quinoline alkaloids (1, 2, 4, 8, and 9), three new coumarins (15, 17, and 19), and 20 known compounds. The structures were determined by analysis of 1D and 2D NMR spectroscopic data. The absolute configuration of 19 was proposed by electronic circular dichroism calculation. Among the compounds tested in the phenotypic screening to measure adiponectin secretion in human bone marrow mesenchymal stem cells, metabolites 4 and 12 stimulated adiponectin secretions with EC50 values of 13.8 and 25.8 μM, respectively. Further PPARγ binding assay and molecular modeling suggested that compounds 4 and 12 are selective PPARγ agonists.
Collapse
Affiliation(s)
- Piseth Nhoek
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Sungjin Ahn
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Pisey Pel
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Mi Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jungmoo Huh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyun Woo Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Gyeonggi-do 10326, Republic of Korea
| | - Minsoo Noh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Won Chin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
11
|
Tetramethylpyrazine and Paeoniflorin Synergistically Attenuate Cholesterol Efflux in Macrophage Cells via Enhancing ABCA1 and ABCG1 Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4304790. [PMID: 36387364 PMCID: PMC9653297 DOI: 10.1155/2022/4304790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/28/2022] [Accepted: 10/29/2022] [Indexed: 11/08/2022]
Abstract
The formation of foam cells is a characteristic of the occurrence and development of atherosclerosis. ATP-binding cassette subfamily A1 and G1 (ABCA1 and ABCG1) and scavenger receptor B1 (SR-B1) play critical roles in promoting intracellular cholesterol efflux to high-density lipoprotein (HDL) or apolipoprotein A1 (apoA1). We attempted to test the effect of the tetramethylpyrazine-paeoniflorin pair (TP) on cholesterol outflow in foam cells derived from macrophages. In this study, RAW264.7 macrophages were treated with 80 mg/L oxidized low-density lipoprotein (ox-LDL) for 24 h to obtain foam cells. Then they were intervened with TP (tetramethylpyrazine 40 ug/ml plus paeoniflorin 80 ug/ml) for additional 24 h. The distribution of cholesterol in foam cells was evaluated by oil red O staining. The contents of total cholesterol (TC) and free cholesterol (FC) were assessed with commercial kits. Fluorescent imaging was observed with a fluorescent inverted microscope. The capacity of cholesterol efflux was measured with a fluorescent plate reader, and the transcript and protein levels of ABCA1, ABCG1, and SR-B1 were detected by Western blot and quantitative polymerase chain reactions (Q-PCRs). Cytokines in the medium were detected by ELISA and adjusted by total cellular proteins. The results showed that TP decreased ox-LDL-induced cholesterol deposition and foam cell formation by promoting cholesterol efflux to apoA1, which was related to the upregulation of ABCA1 and ABCG1. Moreover, TP decreased the secretion of ox-LDL-induced tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), and monocyte chemotactic protein-1 (MCP-1), an important profoam cell cytokine in atherosclerosis.
Collapse
|
12
|
Griesler B, Schuelke C, Uhlig C, Gadasheva Y, Grossmann C. Importance of Micromilieu for Pathophysiologic Mineralocorticoid Receptor Activity-When the Mineralocorticoid Receptor Resides in the Wrong Neighborhood. Int J Mol Sci 2022; 23:12592. [PMID: 36293446 PMCID: PMC9603863 DOI: 10.3390/ijms232012592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/06/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
The mineralocorticoid receptor (MR) is a member of the steroid receptor family and acts as a ligand-dependent transcription factor. In addition to its classical effects on water and electrolyte balance, its involvement in the pathogenesis of cardiovascular and renal diseases has been the subject of research for several years. The molecular basis of the latter has not been fully elucidated, but an isolated increase in the concentration of the MR ligand aldosterone or MR expression does not suffice to explain long-term pathologic actions of the receptor. Several studies suggest that MR activity and signal transduction are modulated by the surrounding microenvironment, which therefore plays an important role in MR pathophysiological effects. Local changes in micromilieu, including hypoxia, ischemia/reperfusion, inflammation, radical stress, and aberrant salt or glucose concentrations affect MR activation and therefore may influence the probability of unphysiological MR actions. The surrounding micromilieu may modulate genomic MR activity either by causing changes in MR expression or MR activity; for example, by inducing posttranslational modifications of the MR or novel interaction with coregulators, DNA-binding sites, or non-classical pathways. This should be considered when developing treatment options and strategies for prevention of MR-associated diseases.
Collapse
Affiliation(s)
| | | | | | | | - Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| |
Collapse
|
13
|
Hines IN, Milton J, Kremer M, Wheeler MD. Ablation of Tumor Necrosis Factor Alpha Receptor 1 Signaling Blunts Steatohepatitis in Peroxisome Proliferator Activated Receptor α-Deficient Mice. MEDICAL RESEARCH ARCHIVES 2022; 10:3082. [PMID: 36865784 PMCID: PMC9977327 DOI: 10.18103/mra.v10i9.3082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Tumor necrosis factor -alpha (TNFα) is strongly associated with fatty liver disease (i.e, hepatosteatosis). Cytokine production has been thought of as a consequence of hepatic lipid accumulation which becomes a critical factor in the development of chronic liver pathologies as well as insulin resistance. The purpose of this study was to test the hypothesis that TNFα directly regulates lipid metabolism in liver in the mutant peroxisome-proliferator activated receptor-alpha (PPARα-/-) mouse model with robust hepatic lipid accumulation. At 10 weeks of age, TNFα and TNF receptor 1 expression are increased in livers of PPARα-/- mice compared to wild type. PPARα-/- mice were then crossed with mice lacking the receptor for TNFα receptor 1 (TNFR1-/-). Wild type, PPARα-/-, TNFR1-/-, PPARα-/- x TNFR1-/- mice were housed on ad-libitum standard chow diet for up to 40 weeks. Increases in hepatic lipid and liver injury and metabolic disruption associated with PPARα ablation were largely blunted when PPARα-/- mice were crossed with TNFR1-/- mice. These data support the hypothesis that TNFR1 signaling is critical for accumulation of lipid in liver. Therapies that reduce pro-inflammatory responses, namely TNFα, could have important clinical implications to reduce hepatosteatosis and progression of severe liver disease.
Collapse
Affiliation(s)
- Ian N. Hines
- Department of Nutrition Science, East Carolina University, North Carolina, USA
| | - Jamie Milton
- Department of Nutrition Science, East Carolina University, North Carolina, USA
| | - Michael Kremer
- Department of General and Visceral Surgery, Hospital of Aarau, Aarau, Switzerland
| | - Michael D. Wheeler
- Department of Nutrition Science, East Carolina University, North Carolina, USA,
| |
Collapse
|
14
|
Gehrke N, Hofmann LJ, Straub BK, Rühle F, Waisman A, Galle PR, Schattenberg JM. Hepatic interleukin-1 receptor type 1 signalling regulates insulin sensitivity in the early phases of nonalcoholic fatty liver disease. Clin Transl Med 2022; 12:e1048. [PMID: 36101976 PMCID: PMC9471277 DOI: 10.1002/ctm2.1048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/15/2022] [Accepted: 08/23/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is associated with hepatic as well as systemic insulin resistance even in the absence of type 2 diabetes. The extent and pathways through which hepatic inflammation modulates insulin sensitivity in NAFLD are only partially understood. We explored the contribution of hepatic interleukin (IL)-1 signalling in a novel conditional knockout mouse model and expand the knowledge on this signalling pathway with regard to its liver-specific functions. METHODS A high-fat, high-carbohydrate diet (HFD) over 12 weeks was used in male hepatocyte-specific IL-1 receptor type 1 (IL-1R1) knockout mice (Il1r1Hep-/- ) and wild-type (WT) littermates. RESULTS Both genotypes developed an obese phenotype and accompanying macrovesicular hepatic steatosis. In contrast to WT mice, microvesicular steatosis and ballooning injury was less pronounced in HFD-fed Il1r1Hep-/- mice, and alanine aminotransferase remained in the normal range. This was paralleled by the suppression of injurious and proinflammatory hepatic c-Jun N-terminal kinases and extracellular signal-regulated kinases signalling, stable peroxisome proliferator activated receptor gamma coactivator-1alpha and farnesoid X receptor-alpha expression and preservation of mitochondrial function. Strikingly, despite HFD-feeding Il1r1Hep-/- mice remained highly insulin sensitive as indicated by lower insulin levels, homeostatic model assessment for insulin resistance, higher glucose tolerance, more stable hepatic insulin signalling cascade, and less adipose tissue inflammation compared to the WT. CONCLUSIONS The current data highlights that hepatocyte IL-1R1 contributes to hepatic and extrahepatic insulin resistance. Future liver-directed therapies in NAFLD could have effects on insulin sensitivity when improving hepatic inflammation and IL-1R1 signalling.
Collapse
Affiliation(s)
- Nadine Gehrke
- I. Department of MedicineUniversity Medical Center of the Johannes Gutenberg University MainzMainz55131Germany
| | - Lea J. Hofmann
- I. Department of MedicineUniversity Medical Center of the Johannes Gutenberg University MainzMainz55131Germany
| | - Beate K. Straub
- Institute of PathologyUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - Frank Rühle
- Bioinformatics Core FacilityInstitute of Molecular Biology (IMB)MainzGermany
| | - Ari Waisman
- Institute for Molecular MedicineUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
- Research Center for ImmunotherapyUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - Peter R. Galle
- I. Department of MedicineUniversity Medical Center of the Johannes Gutenberg University MainzMainz55131Germany
- Research Center for ImmunotherapyUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - Jörn M. Schattenberg
- I. Department of MedicineUniversity Medical Center of the Johannes Gutenberg University MainzMainz55131Germany
| |
Collapse
|
15
|
Janovick NA, Dann HM, Loor JJ, Drackley JK. Prepartum dietary energy intake alters hepatic expression of genes related to peroxisome proliferator-activated receptor and inflammation in peripartal dairy cows. J Dairy Sci 2022; 105:8069-8086. [PMID: 36028348 DOI: 10.3168/jds.2021-21669] [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: 12/05/2021] [Accepted: 05/18/2022] [Indexed: 11/19/2022]
Abstract
We determined the effect of prepartum plane of energy intake on liver function and metabolism pre- and postpartum by combining in vivo and in vitro data with mRNA expression data. A subset of multiparous prepartal Holsteins (n = 18) from a previously conducted experiment consumed 1 of 3 amounts of dietary energy intake, relative to their requirements. A diet formulated to allow consumption of ≥150% of net energy requirements during the far-off dry period and the close-up dry period was fed for ad libitum intake (150E) or in restricted amounts so that cows consumed 80% of requirements for energy (80E). A second diet was formulated to include wheat straw (26.1% of dry matter) to limit energy intake to 100% of NRC (2001) requirements for energy when fed ad libitum during the far-off period (100E). In the close-up period, 100E was fed the 150E diet for ad libitum intake. Expression of mRNA for genes related to fatty acid oxidation (PPARA, CPT1A, ACOX1) was greater for 100E cows than 150E cows on d 14 postpartum. These expression patterns were related to in vitro data for conversion of palmitate to CO2, acid-soluble products, and esterified products by liver slices. Abundance of mRNA for PC displayed a sharp peak for all groups on d 1 postpartum, but serum glucose did not reflect this peak. The mRNA expression of SREBF1 was greater for 150E and 100E cows prepartum compared with 80E, and was positively related to rate of palmitate esterification postpartum. Expression of NR1H3 (LXRA) mRNA was greater for 100E cows on d 14 postpartum compared with 150E cows, which corresponded to expression of PPARA. An inflammatory response occurred in the liver around the time of parturition for 150E cows, as expression of IL1B was elevated both pre- and postpartum compared with 100E cows. The spike in IL1B expression for 150E cows on d 14 postpartum corresponded to the peak concentration of total lipids in liver tissue for all groups in this experiment. Overconsumption of energy prepartum was detrimental to the expression of important genes related to PPAR and liver function, especially postpartum. Furthermore, results provide evidence for inflammation related to accumulation of lipids in liver and overnutrition prepartum.
Collapse
Affiliation(s)
- N A Janovick
- Department of Animal Sciences, University of Illinois, Urbana-Champaign 61801
| | - H M Dann
- Department of Animal Sciences, University of Illinois, Urbana-Champaign 61801
| | - J J Loor
- Department of Animal Sciences, University of Illinois, Urbana-Champaign 61801
| | - J K Drackley
- Department of Animal Sciences, University of Illinois, Urbana-Champaign 61801.
| |
Collapse
|
16
|
PPARβ/δ Augments IL-1β-Induced COX-2 Expression and PGE2 Biosynthesis in Human Mesangial Cells via the Activation of SIRT1. Metabolites 2022; 12:metabo12070595. [PMID: 35888719 PMCID: PMC9320509 DOI: 10.3390/metabo12070595] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/11/2022] [Accepted: 06/22/2022] [Indexed: 12/10/2022] Open
Abstract
Peroxisome proliferator-activated receptor β/δ (PPARβ/δ), a ligand-activated nuclear receptor, regulates lipid and glucose metabolism and inflammation. PPARβ/δ can exert an anti-inflammatory effect by suppressing proinflammatory cytokine production. Cyclooxygenase-2 (COX-2)-triggered inflammation plays a crucial role in the development of many inflammatory diseases, including glomerulonephritis. However, the effect of PPARβ/δ on the expression of COX-2 in the kidney has not been fully elucidated. The present study showed that PPARβ/δ was functionally expressed in human mesangial cells (hMCs), where its expression was increased by interleukin-1β (IL-1β) treatment concomitant with enhanced COX-2 expression and prostaglandin E2 (PGE2) biosynthesis. The treatment of hMCs with GW0742, a selective agonist of PPARβ/δ, or the overexpression of PPARβ/δ via an adenovirus-mediated approach significantly increased COX-2 expression and PGE2 production. PPARβ/δ could further augment the IL-1β-induced COX-2 expression and PGE2 production in hMCs. Moreover, both PPARβ/δ activation and overexpression markedly increased sirtuin 1 (SIRT1) expression. The inhibition or knockdown of SIRT1 significantly attenuated the effects of PPARβ/δ on the IL-1β-induced expression of COX-2 and PGE2 biosynthesis. Taken together, PPARβ/δ could augment the IL-1β-induced COX-2 expression and PGE2 production in hMCs via the SIRT1 pathway. Given the critical role of COX-2 in glomerulonephritis, PPARβ/δ may represent a novel target for the treatment of renal inflammatory diseases.
Collapse
|
17
|
Sæterstad S, Østvik AE, Røyset ES, Bakke I, Sandvik AK, Granlund AVB. Profound gene expression changes in the epithelial monolayer of active ulcerative colitis and Crohn's disease. PLoS One 2022; 17:e0265189. [PMID: 35275975 PMCID: PMC8916644 DOI: 10.1371/journal.pone.0265189] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 02/25/2022] [Indexed: 12/21/2022] Open
Abstract
In recent years it has become apparent that the epithelium is highly involved in inflammatory bowel disease (IBD) pathophysiology. The majority of gene expression studies of IBD are generated from heterogeneous biopsies, providing no distinction between immune cells, the epithelium and other mucosal cells. By using laser capture microdissection (LCM) coupled with RNA sequencing, we aimed to characterize the expressional changes of the isolated colonic epithelial monolayer from ulcerative colitis (UC) and Crohn’s disease (CD) patients compared to healthy controls (HC). The analysis identified 3706 genes as differentially expressed between active IBD epithelium and HC. Weighted gene co-expression network analysis was used to stratify genes into modules, which were subsequently characterized using enrichment analysis. Our data show a distinct upregulation of the antigen presentation machinery during inflammation, including major histocompatibility complex class II molecules (e.g. HLA-DPA1, HLA-DPB1, HLA-DRA) and key transcription factors/activators (STAT1, IRF1, CIITA). We also see an epithelial downregulation of retinoic acid-responsive nuclear receptors (RARA, RARB, RXRA), but upregulation of retinoid-metabolizing enzymes (RDH11, ALDH1A2, ALDH1A3), which together suggest a perturbation of epithelial vitamin A signaling during active IBD. Lastly, we identified a cluster of stress-related genes, including activator protein 1 components JUNB and ATF3, as significantly upregulated in active UC but not in CD, revealing an interesting aspect of IBD heterogeneity. The results represent a unique resource for enhanced understanding of epithelial involvement in IBD inflammation and is a valuable tool for further studies on these processes.
Collapse
Affiliation(s)
- Siri Sæterstad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ann Elisabet Østvik
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav’s University Hospital, Trondheim, Norway
| | - Elin Synnøve Røyset
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Pathology, St. Olav’s University Hospital, Trondheim, Norway
| | - Ingunn Bakke
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Medicine, St Olav’s University Hospital, Trondheim, Norway
| | - Arne Kristian Sandvik
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav’s University Hospital, Trondheim, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Atle van Beelen Granlund
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Medicine, St Olav’s University Hospital, Trondheim, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- * E-mail:
| |
Collapse
|
18
|
Allegra A, Petrarca C, Di Gioacchino M, Casciaro M, Musolino C, Gangemi S. Modulation of Cellular Redox Parameters for Improving Therapeutic Responses in Multiple Myeloma. Antioxidants (Basel) 2022; 11:antiox11030455. [PMID: 35326105 PMCID: PMC8944660 DOI: 10.3390/antiox11030455] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 01/25/2023] Open
Abstract
Raised oxidative stress and abnormal redox status are typical features of multiple myeloma cells, and the identification of the intimate mechanisms that regulate the relationships between neoplastic cells and redox homeostasis may reveal possible new anti-myeloma therapeutic targets to increase the effectiveness of anti-myeloma drugs synergistically or to eradicate drug-resistant clones while reducing toxicity toward normal cells. An alteration of the oxidative state is not only responsible for the onset of multiple myeloma and its progression, but it also appears essential for the therapeutic response and for developing any chemoresistance. Our review aimed to evaluate the literature’s current data on the effects of oxidative stress on the response to drugs generally employed in the therapy of multiple myeloma, such as proteasome inhibitors, immunomodulators, and autologous transplantation. In the second part of the review, we analyzed the possibility of using other substances, often of natural origin, to modulate the oxidative stress to interfere with the progression of myelomatous disease.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
- Correspondence: (A.A.); (M.D.G.)
| | - Claudia Petrarca
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
| | - Mario Di Gioacchino
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
- Correspondence: (A.A.); (M.D.G.)
| | - Marco Casciaro
- Unit and School of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Unit and School of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| |
Collapse
|
19
|
ALSuhaymi N, Darwish AM, Khattab AEN. Assessment of Two Potential Probiotic Strains As Anti-Obesity Supplements Under High-Fat Feeding Conditions. Probiotics Antimicrob Proteins 2022:10.1007/s12602-022-09912-w. [PMID: 35088380 DOI: 10.1007/s12602-022-09912-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2022] [Indexed: 01/19/2023]
Abstract
Obesity is one of the chronic diseases that increase annually and cause cardiovascular disease, which is the main cause of death worldwide. So, this study aims to evaluate the role of the two potential probiotics: Lactiplantibacillus plantarum Pro1 and Lacticaseibacillus rhamnosus Pro2, isolated from the fermented milk and corn silage as anti-obesity supplements. Seventy-five male BALB/c mice were distributed to five groups (control, obesity, obesity plus L. plantarum (OLP), obesity plus L. rhamnosus (OLR) and obesity plus mixture of two strains (OM)). The body weight, lipid profile, histopathology and enzymes of liver were assessed. RT-PCR was used to determine the expression of CYP7A1, ALTG4, TNFα and ROR genes.The findings show that the obesity group recorded the significant highest value of the body weight, TC, TG, LDL, AST and ALT, while OLP group recorded the significant lowest value. Liver tissue of obesity group has necrosis and fatty changes, while the OLP group was related to the control group. The findings of RT-PCR show non-significant differences between the control group and the OLP group, with significant differences between the control group and the set groups in expression of CYP7A1, ALTG4, TNFα and ROR genes. L. plantarum Pro1 reduced the expression of inflammation genes (TNFα and ROR), and increase the expression of the lipid metabolism genes (CYP7A1, ALTG4) to reduce the inflammatory effects of obesity in the liver, and decrease the cholesterol level in serum. Therefore, L. plantarum Pro1 is useful as anti-obesity supplements and an eliminator of the relevant diseases.
Collapse
Affiliation(s)
- Naif ALSuhaymi
- Department of Emergency Medical Services, College of Health Sciences in AlQunfudah, Umm Al-Qura University, Mekkah, Saudi Arabia
| | - Ahmed Mohamed Darwish
- Cell Biology Department, Biotechnology Research Institute, National Research Centre, Dokki, Giza, Egypt.
| | - Abd El-Nasser Khattab
- Genetics and Cytology Department, Biotechnology Research Institute, National Research Centre, Dokki, Giza, Egypt
| |
Collapse
|
20
|
Blunder S, Krimbacher T, Moosbrugger‐Martinz V, Gruber R, Schmuth M, Dubrac S. Keratinocyte-derived IL-1β induces PPARG downregulation and PPARD upregulation in human reconstructed epidermis following barrier impairment. Exp Dermatol 2021; 30:1298-1308. [PMID: 33683743 PMCID: PMC8451818 DOI: 10.1111/exd.14323] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/15/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear hormone receptors. In skin, PPARs modulate inflammation, lipid synthesis, keratinocyte differentiation and proliferation and thus are important for skin barrier homeostasis. Accordingly, PPAR expression is altered in various skin conditions that entail epidermal barrier impairment, that is atopic dermatitis (AD) and psoriasis. Using human epidermal equivalents (HEEs), we established models of acute epidermal barrier impairment devoid of immune cells. We assessed PPAR and cytokine expression after barrier perturbation and examined effects of keratinocyte-derived cytokines on PPAR expression. We show that acetone or SDS treatment causes graded impairment of epidermal barrier function. Furthermore, we demonstrate that besides IL-1β and TNFα, IL-33 and TSLP are highly relevant markers for acute epidermal barrier impairment. Both SDS- and acetone-mediated epidermal barrier impairment reduce PPARG expression levels, whereas only SDS enhances PPARD expression. In line with findings in IL-1β and TNFα-treated HEEs, abrogation of IL-1 signalling restores PPARG expression and limits the increase of PPARD expression in SDS-induced epidermal barrier impairment. Thus, following epidermal barrier perturbation, keratinocyte-derived IL-1β and partly TNFα modulate PPARG and PPARD expression. These results emphasize a role for PPARγ and PPARβ/δ in acute epidermal barrier impairment with possible implications for diseases such as AD and psoriasis.
Collapse
Affiliation(s)
- Stefan Blunder
- Department of Dermatology, Venereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Thomas Krimbacher
- Department of Dermatology, Venereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | | | - Robert Gruber
- Department of Dermatology, Venereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Matthias Schmuth
- Department of Dermatology, Venereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and AllergologyMedical University of InnsbruckInnsbruckAustria
| |
Collapse
|
21
|
Wang H, Wang L, Li Y, Luo S, Ye J, Lu Z, Li X, Lu H. The HIF-2α/PPARα pathway is essential for liraglutide-alleviated, lipid-induced hepatic steatosis. Biomed Pharmacother 2021; 140:111778. [PMID: 34062416 DOI: 10.1016/j.biopha.2021.111778] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Liraglutide has been demonstrated to alleviate hepatic steatosis in clinical practice, but the underlying mechanism remains unclear. Our previous study indicated that the HIF-2α/PPARα pathway was involved in hepatic lipid accumulation induced by hypoxia.We aimed to investigate whether liraglutide could alleviate lipid-induced hepatic steatosis via the HIF-2α/PPARα pathway. Whole-body HIF-2α heterozygous knockout (HIF-2α+/-) mice and littermate wild-type (WT) mice were successfully established. Male mice challenged with a high-fat diet were treated with liraglutide (0.6 mg/kg/d) or normal saline by intraperitoneal injection for 4 weeks. We observed that, compared with WT mice, many indicators of HIF-2α+/- mice improved, including GTT, ITT, fasting blood glucose, body weight, liver weight, and lipid profile in serum or liver lipid deposition, and the expression level of PPARα, mitochondrial function genes, and fatty acid oxidation genes were upregulated, while those of HIF-2α and lipogenesis genes were downregulated significantly. After liraglutide treatment in WT mice, we found that significant improvements were observed in the fat mass, GTT, ITT, fasting blood glucose, body weight, liver weight, lipid profile in serum or liver lipid deposition; the β-oxidation genes were upregulated and the lipogenesis genes were downregulated; and the abundance of intestinal Akkermansia muciniphila increased significantly. However, the effects of liraglutide on WT mice were not observed in HIF-2α+/- mice. In addition, in the HepG2 steatotic hepatocyte model, liraglutide alleviated lipid deposits by repressing lipid synthesis and enhancing fatty acid β-oxidation, which were substantially suppressed by the HIF-2α modulators. Therefore, the HIF-2α/PPARα pathway is essential for liraglutide-alleviated lipid-induced hepatic steatosis.
Collapse
Affiliation(s)
- Hou Wang
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Lingling Wang
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Yun Li
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Shunkui Luo
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Jianfang Ye
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Zhanjin Lu
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Xiaobin Li
- Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai 519000, Guangdong, China.
| | - Hongyun Lu
- Department of Endocrinology & Metabolism, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China; Department of Endocrinology & Metabolism, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai 519000, Guangdong, China.
| |
Collapse
|
22
|
Uekita H, Yamamoto H, Niinaga R, Yamane N, Yoshii M, Yamauchi-Takihara K, Kihara S. Reciprocal association of serum Mac-2 binding protein and HDL-cholesterol concentrations. Clin Chim Acta 2021; 516:142-148. [PMID: 33571485 DOI: 10.1016/j.cca.2021.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Mac-2 binding protein (Mac-2BP) is used as a serum biomarker of nonalcoholic steatohepatitis, considered to be a liver phenotype of metabolic syndrome (MetS). In this study, we investigated the serum Mac-2BP concentrations-correlated MetS-related clinical parameters in vivo, and the underlying mechanism in vitro. MATERIALS & METHODS We enrolled 54 healthy Japanese men who underwent health examination at Osaka University Health Care Center in this study. Physical and serum biochemical parameters were obtained from all the subjects. In the cultured HepG2 cells, the effects of interferon (IFN)-γ on the expression of Mac-2BP, apolipoprotein (apo) A-I, and ATP binding cassette transporter A1 (ABCA1) were studied. RESULTS Serum Mac-2BP concentrations correlated negatively with HDL-C, and positively with body mass index and systolic blood pressure in univariate analysis. These results suggested the association between Mac-2BP and MetS, although none of these 3 parameters had significant correlation with serum Mac-2BP concentrations in multivariate analysis. In HepG2 cells, IFN-γ stimulation resulted in the increased Mac-2BP and the decreased ABCA1 and apo A-I mRNA concentrations, while Mac-2BP had no effects on ABCA1 and apo A-I concentrations. CONCLUSIONS The serum Mac-2BP concentrations are negatively correlated with HDL-C concentrations in healthy subjects, as a result of chronic inflammation.
Collapse
Affiliation(s)
- Hiromi Uekita
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan; Department of Medical Technology, Faculty of Health Sciences, Kansai University of Health Sciences, Osaka, Japan
| | - Hiroyasu Yamamoto
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Ryu Niinaga
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Noriko Yamane
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Manami Yoshii
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | - Shinji Kihara
- Department of Biomedical Informatics, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
23
|
Frambach SJCM, de Haas R, Smeitink JAM, Rongen GA, Russel FGM, Schirris TJJ. Brothers in Arms: ABCA1- and ABCG1-Mediated Cholesterol Efflux as Promising Targets in Cardiovascular Disease Treatment. Pharmacol Rev 2020; 72:152-190. [PMID: 31831519 DOI: 10.1124/pr.119.017897] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a leading cause of cardiovascular disease worldwide, and hypercholesterolemia is a major risk factor. Preventive treatments mainly focus on the effective reduction of low-density lipoprotein cholesterol, but their therapeutic value is limited by the inability to completely normalize atherosclerotic risk, probably due to the disease complexity and multifactorial pathogenesis. Consequently, high-density lipoprotein cholesterol gained much interest, as it appeared to be cardioprotective due to its major role in reverse cholesterol transport (RCT). RCT facilitates removal of cholesterol from peripheral tissues, including atherosclerotic plaques, and its subsequent hepatic clearance into bile. Therefore, RCT is expected to limit plaque formation and progression. Cellular cholesterol efflux is initiated and propagated by the ATP-binding cassette (ABC) transporters ABCA1 and ABCG1. Their expression and function are expected to be rate-limiting for cholesterol efflux, which makes them interesting targets to stimulate RCT and lower atherosclerotic risk. This systematic review discusses the molecular mechanisms relevant for RCT and ABCA1 and ABCG1 function, followed by a critical overview of potential pharmacological strategies with small molecules to enhance cellular cholesterol efflux and RCT. These strategies include regulation of ABCA1 and ABCG1 expression, degradation, and mRNA stability. Various small molecules have been demonstrated to increase RCT, but the underlying mechanisms are often not completely understood and are rather unspecific, potentially causing adverse effects. Better understanding of these mechanisms could enable the development of safer drugs to increase RCT and provide more insight into its relation with atherosclerotic risk. SIGNIFICANCE STATEMENT: Hypercholesterolemia is an important risk factor of atherosclerosis, which is a leading pathological mechanism underlying cardiovascular disease. Cholesterol is removed from atherosclerotic plaques and subsequently cleared by the liver into bile. This transport is mediated by high-density lipoprotein particles, to which cholesterol is transferred via ATP-binding cassette transporters ABCA1 and ABCG1. Small-molecule pharmacological strategies stimulating these transporters may provide promising options for cardiovascular disease treatment.
Collapse
Affiliation(s)
- Sanne J C M Frambach
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ria de Haas
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan A M Smeitink
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerard A Rongen
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tom J J Schirris
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences (S.J.C.M.F., G.A.R., F.G.M.R., T.J.J.S.), Radboud Center for Mitochondrial Medicine (S.J.C.M.F., R.d.H., J.A.M.S., F.G.M.R., T.J.J.S.), Department of Pediatrics (R.d.H., J.A.M.S.), and Department of Internal Medicine, Radboud Institute for Health Sciences (G.A.R.), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Yang X, Wu F, Li L, Lynch EC, Xie L, Zhao Y, Fang K, Li J, Luo J, Xu L, Zou X, Lu F, Chen G. Celastrol alleviates metabolic disturbance in high-fat diet-induced obese mice through increasing energy expenditure by ameliorating metabolic inflammation. Phytother Res 2020; 35:297-310. [PMID: 32776627 DOI: 10.1002/ptr.6800] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022]
Abstract
Celastrol, a natural triterpene, has been shown to treat obesity and its related metabolic disorders. In this study, we first assessed the relationship between the antiobesity effects of celastrol and its antiinflammatory activities. Our results showed that celastrol can reduce weight gain, ameliorate glucose intolerance, insulin resistance, and dyslipidemia without affecting food intake in high-fat diet-induced obese mice. A CLAMS was used to clarify the improvement of metabolic profiles was attribute to increased adipose thermogenesis after celastrol treatment. Further studies found that celastrol decreased the infiltration of macrophage as well as its inflammatory products (IL-1β, IL-18, MCP-1α, and TNF-α) in liver and adipose tissues, which also displayed an obvious inhibition of TLR3/NLRP3 inflammasome molecules. This study demonstrated that celastrol could be a potential drug for treating metabolic disorders, the underlying mechanism is related to ameliorating metabolic inflammation, thus increasing body energy expenditure.
Collapse
Affiliation(s)
- Xueping Yang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Wu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingli Li
- Department of Traditional Chinese Medicine, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ernest C Lynch
- Department of Nutrition & Food Science, Texas A&M University, College Station, Texas, USA
| | - Linglin Xie
- Department of Nutrition & Food Science, Texas A&M University, College Station, Texas, USA
| | - Yan Zhao
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Fang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingbin Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinlong Luo
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijun Xu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zou
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuer Lu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guang Chen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Li P, Wang Y, Luo J, Zeng Q, Wang M, Bai M, Zhou H, Wang J, Jiang H. Downregulation of OCTN2 by cytokines plays an important role in the progression of inflammatory bowel disease. Biochem Pharmacol 2020; 178:114115. [PMID: 32579962 DOI: 10.1016/j.bcp.2020.114115] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022]
Abstract
Inflammatory bowel diseases (IBD) are characterized by chronic relapsing disorders of the gastrointestinal tract. OCTN2 (SLC22A5) and its substrate l-carnitine (l-Car) play crucial roles in maintaining normal intestinal function. An aim of this study was to delineate the expression alteration of OCTN2 in IBD and its underlying mechanism. We also investigated the impact of OCTN2 on IBD progression and the possibility of improving IBD through OCTN2 regulation. Our results showed decreased OCTN2 expression levels and l-Car content in inflamed colon tissues of IBD patients and mice, which negatively correlated with the degree of colonic inflammation in IBD mice. Mixed proinflammatory cytokines TNF-α, IL-1β and IFNγ downregulated the expression of OCTN2 and subsequently reduced the l-Car content through PPARγ/RXRα pathways in FHC cells. OCTN2 silencing reduced the proliferation rate of the colon cells, whereas OCTN2 overexpression increased the proliferation rate. Furthermore, the ability of PPARγ agonist, luteolin, to increase OCTN2 expression resulted in the alleviation of colonic inflammatory responses. In conclusion, OCTN2 was downregulated in IBD by proinflammatory cytokines via the PPARγ/RXRα pathways, which reduced l-Car concentration and subsequently induced IBD deterioration. Upregulation of OCTN2 by the PPARγ agonist alleviated colonic inflammation. Our findings suggest that, OCTN2 may serve as a therapeutic target for IBD therapy.
Collapse
Affiliation(s)
- Ping Li
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yuqing Wang
- Hangzhou Translational Medicine Research Center, Hangzhou First People's Hospital, Hangzhou, China; Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jun Luo
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qingquan Zeng
- College of Medicine, Zhejiang University, Hangzhou, China; Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Miaojuan Wang
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Mengru Bai
- Hangzhou Translational Medicine Research Center, Hangzhou First People's Hospital, Hangzhou, China; Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hui Zhou
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jinhai Wang
- College of Medicine, Zhejiang University, Hangzhou, China.
| | - Huidi Jiang
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
26
|
Fontecha-Barriuso M, Martin-Sanchez D, Martinez-Moreno JM, Monsalve M, Ramos AM, Sanchez-Niño MD, Ruiz-Ortega M, Ortiz A, Sanz AB. The Role of PGC-1α and Mitochondrial Biogenesis in Kidney Diseases. Biomolecules 2020; 10:biom10020347. [PMID: 32102312 PMCID: PMC7072614 DOI: 10.3390/biom10020347] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is one of the fastest growing causes of death worldwide, emphasizing the need to develop novel therapeutic approaches. CKD predisposes to acute kidney injury (AKI) and AKI favors CKD progression. Mitochondrial derangements are common features of both AKI and CKD and mitochondria-targeting therapies are under study as nephroprotective agents. PGC-1α is a master regulator of mitochondrial biogenesis and an attractive therapeutic target. Low PGC-1α levels and decreased transcription of its gene targets have been observed in both preclinical AKI (nephrotoxic, endotoxemia, and ischemia-reperfusion) and in experimental and human CKD, most notably diabetic nephropathy. In mice, PGC-1α deficiency was associated with subclinical CKD and predisposition to AKI while PGC-1α overexpression in tubular cells protected from AKI of diverse causes. Several therapeutic strategies may increase kidney PGC-1α activity and have been successfully tested in animal models. These include AMP-activated protein kinase (AMPK) activators, phosphodiesterase (PDE) inhibitors, and anti-TWEAK antibodies. In conclusion, low PGC-1α activity appears to be a common feature of AKI and CKD and recent characterization of nephroprotective approaches that increase PGC-1α activity may pave the way for nephroprotective strategies potentially effective in both AKI and CKD.
Collapse
Affiliation(s)
- Miguel Fontecha-Barriuso
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
| | - Diego Martin-Sanchez
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
| | - Julio Manuel Martinez-Moreno
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
| | - Maria Monsalve
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), 28029 Madrid, Spain;
| | - Adrian Mario Ramos
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
| | - Maria Dolores Sanchez-Niño
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
| | - Marta Ruiz-Ortega
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
- School of Medicine, UAM, 28029 Madrid, Spain
| | - Alberto Ortiz
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
- School of Medicine, UAM, 28029 Madrid, Spain
- IRSIN, 28040 Madrid, Spain
| | - Ana Belen Sanz
- Research Institute-Fundacion Jimenez Diaz, Autonoma University, 28040 Madrid, Spain; (M.F.-B.); (D.M.-S.); (J.M.M.-M.); (A.M.R.); (M.D.S.-N.); (M.R.-O.); (A.O.)
- REDINREN, 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-91-550-48-00
| |
Collapse
|
27
|
Lecoutre S, Montel V, Vallez E, Pourpe C, Delmont A, Eury E, Verbanck M, Dickes-Coopman A, Daubersies P, Lesage J, Laborie C, Tailleux A, Staels B, Froguel P, Breton C, Vieau D. Transcription profiling in the liver of undernourished male rat offspring reveals altered lipid metabolism pathways and predisposition to hepatic steatosis. Am J Physiol Endocrinol Metab 2019; 317:E1094-E1107. [PMID: 31638854 DOI: 10.1152/ajpendo.00291.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Clinical and animal studies have reported an association between low birth weight and the development of nonalcoholic fatty liver disease (NAFLD) in offspring. Using a model of prenatal maternal 70% food restriction diet (FR30) in the rat, we previously showed that maternal undernutrition predisposes offspring to altered lipid metabolism in adipose tissue, especially on a high-fat (HF) diet. Here, using microarray-based expression profiling combined with metabolic, endocrine, biochemical, histological, and lipidomic approaches, we assessed whether FR30 procedure sensitizes adult male offspring to impaired lipid metabolism in the liver. No obvious differences were noted in the concentrations of triglycerides, cholesterol, and bile acids in the liver of 4-mo-old FR30 rats whichever postweaning diet was used. However, several clues suggest that offspring's lipid metabolism and steatosis are modified by maternal undernutrition. First, lipid composition was changed (i.e., higher total saturated fatty acids and lower elaidic acid) in the liver, whereas larger triglyceride droplets were observed in hepatocytes of undernourished rats. Second, FR30 offspring exhibited long-term impact on hepatic gene expression and lipid metabolism pathways on a chow diet. Although the transcriptome profile was globally modified by maternal undernutrition, cholesterol and bile acid biosynthesis pathways appear to be key targets, indicating that FR30 animals were predisposed to impaired hepatic cholesterol metabolism. Third, the FR30 protocol markedly modifies hepatic gene transcription profiles in undernourished offspring in response to postweaning HF. Overall, FR30 offspring may exhibit impaired metabolic flexibility, which does not enable them to properly cope with postweaning nutritional challenges influencing the development of nonalcoholic fatty liver.
Collapse
Affiliation(s)
- Simon Lecoutre
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | - Valérie Montel
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | - Emmanuelle Vallez
- Université Lille, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, INSERM U1011-European Genomic Institute for Diabetes, Lille, France
| | - Charlène Pourpe
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | | | - Elodie Eury
- Université Lille, UMR 8199, European Genomic Institute for Diabetes, Lille, France
| | - Marie Verbanck
- Université Lille, UMR 8199, European Genomic Institute for Diabetes, Lille, France
| | - Anne Dickes-Coopman
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | | | - Jean Lesage
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | - Christine Laborie
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | - Anne Tailleux
- Université Lille, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, INSERM U1011-European Genomic Institute for Diabetes, Lille, France
| | - Bart Staels
- Université Lille, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, INSERM U1011-European Genomic Institute for Diabetes, Lille, France
| | - Philippe Froguel
- Université Lille, UMR 8199, European Genomic Institute for Diabetes, Lille, France
| | - Christophe Breton
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | - Didier Vieau
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| |
Collapse
|
28
|
Kasimanickam R, Kasimanickam V, Grende K. Endometrial expression of various genes (ISGs, PPARs, RXRs and MUC1) on day 16 post-ovulation in repeat breeder cows, with or without subclinical endometritis. Theriogenology 2019; 142:251-259. [PMID: 31711690 DOI: 10.1016/j.theriogenology.2019.10.036] [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: 03/05/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 12/01/2022]
Abstract
Our objective was to elucidate differences in endometrial mRNA expressions of interferon-stimulated genes (ISG15, CTSL1, RSAD2, SLC2A1, CXCL10, and SLC27A6), peroxisome proliferator activated receptors (PPARA, PPARD, and PPARG), retinoic acid receptors (RXRA, RXRB, and RXRG), and mucin 1 (MUC1) in repeat breeder cows, with or without subclinical endometritis (RB + SE and RB, respectively) and normal cows on day 16 post-ovulation (n = 4 cows per group). The CXCL10 and SLC27A6 mRNA abundances were greater for normal cows compared to RB and RB + SE cows (P < 0.05 and P < 0.01 respectively) whereas ISG15 and SLC2A1 mRNA abundances were greater for normal cows compared to RB + SE (P < 0.05). The SLC27A6 mRNA abundances were greater for RB versus RB + SE (P < 0.01). Similarly, PPARD, PPARG, RXRA and RXRG mRNA abundances were greater for normal cows compared to RB and RB + SE (P < 0.01 and P < 0.05, respectively). Abundances of PPARD, PPARG, RXRA and RXRG mRNA were greater for RB versus RB + SE (P < 0.05) and MUC1 was lower in abundance in normal cows compared to RB or RB + SE (P < 0.05). Key predicted molecular functions were binding, signal transducer and transporter; key biological processes were cellular, localization and metabolic; key cellular components were cell part, membrane and organelle components; and key protein classes were nucleic acid binding, receptor, and transcription factors. Gene networking analysis highlighted interactions and pathways involving PAPRs, RXRs, and MUC1, notably among PPARD, PPARG, and MUC1. In conclusion, endometrial mRNA expressions of ISGs (CXCL10 and SLC27A6), PPAR isomers (PPARD and PPARG), and RXRs (RXRA and RXRG) were in lower abundances, whereas MUC1 expression was more abundant in RB or RB + SE compared to normal cows on day 16. In addition, ISG15 and SLC2A1 genes were less abundant in RB + SE versus RB or normal cows. Altered expression of these uterine genes and associated potential impairment in embryo elongation and implantation may promote embryonic loss in repeat breeder cows. Furthermore, interactions among PPARD, PPARG and MUC1 may be therapeutically exploitable.
Collapse
Affiliation(s)
- R Kasimanickam
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
| | - V Kasimanickam
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - K Grende
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
29
|
Ryan KM, Patterson I, McLoughlin DM. Peroxisome proliferator-activated receptor gamma co-activator-1 alpha in depression and the response to electroconvulsive therapy. Psychol Med 2019; 49:1859-1868. [PMID: 30191781 DOI: 10.1017/s0033291718002556] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The transcriptional coactivator peroxisome proliferator-activated receptor-γ coactivator (PGC-1α), termed the 'master regulator of mitochondrial biogenesis', has been implicated in stress and resilience to stress-induced depressive-like behaviours in animal models. However, there has been no study conducted to date to examine PGC-1α levels in patients with depression or in response to antidepressant treatment. Our aim was to assess PGC-1α mRNA levels in blood from healthy controls and patients with depression pre-/post-electroconvulsive therapy (ECT), and to examine the relationship between blood PGC-1α mRNA levels and clinical symptoms and outcomes with ECT. METHODS Whole blood PGC-1α mRNA levels were analysed in samples from 67 patients with a major depressive episode and 70 healthy controls, and in patient samples following a course of ECT using quantitative real-time polymerase chain reaction (qRT-PCR). Exploratory subgroup correlational analyses were carried out to determine the relationship between PGC-1α and mood scores. RESULTS PGC-1α levels were lower in patients with depression compared with healthy controls (p = 0.03). This lower level was predominantly accounted for by patients with psychotic unipolar depression (p = 0.004). ECT did not alter PGC-1α levels in the depressed group as a whole, though exploratory analyses revealed a significant increase in PGC-1α in patients with psychotic unipolar depression post-ECT (p = 0.045). We found no relationship between PGC-1α mRNA levels and depression severity or the clinical response to ECT. CONCLUSIONS PGC-1α may represent a novel therapeutic target for the treatment of depression, and be a common link between various pathophysiological processes implicated in depression.
Collapse
Affiliation(s)
- Karen M Ryan
- Trinity College Institute of Neuroscience, Trinity College Dublin,Dublin,Ireland
| | - Ian Patterson
- Trinity College Institute of Neuroscience, Trinity College Dublin,Dublin,Ireland
| | - Declan M McLoughlin
- Trinity College Institute of Neuroscience, Trinity College Dublin,Dublin,Ireland
| |
Collapse
|
30
|
He Y, Zhou X, Li X, Jin X, Wang X, Pan X, Bi D. Relationship between CYP3A29 and pregnane X receptor in landrace pigs: Pig CYP3A29 has a similar mechanism of regulation to human CYP3A4. Comp Biochem Physiol C Toxicol Pharmacol 2018; 214:9-16. [PMID: 30153482 DOI: 10.1016/j.cbpc.2018.08.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/21/2018] [Accepted: 08/21/2018] [Indexed: 11/30/2022]
Abstract
The objective of this study was to provide evidence of the validity of utilizing pigs as a model to study the regulation of human CYP3A4, with special emphasis on drug-drug interactions. We determined the mRNA expression and distribution of CYP3A and metabolic nuclear receptors in different tissues isolated from landrace pigs. Our results showed that CYP3A and metabolic nuclear receptor mRNAs were most highly expressed in liver tissues. The expression of the metabolic nuclear receptor pregnane X receptor (PXR) had a significant correlation with expression of CYP3A29, an analog of human CYP3A4. The correlation between their transcriptional levels was further demonstrated using LPS and TNF-α. The mRNA and protein expression of CYP3A29 and PXR in HepLi cells was significantly reduced by LPS and TNF-α treatment. CYP3A29 promoter activity was dramatically elevated by PXR over expression, whereas LPS and TNF-α treatment inhibited the enhanced CYP3A29 promoter activity that was induced by PXR; presumably through inhibition of PXR promoter activity. Furthermore, the inhibition of CYP3A29 promoter activity by LPS and TNF-α treatment was blocked by knockdown of PXR or retinoid X receptor (RXR). These data suggest high similarity in the regulation mechanism of pig CYP3A29 and human CYP3A4. Our research provided a significant evaluation to determine whether pigs are suitable as an experimental animal model.
Collapse
Affiliation(s)
- Yucheng He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Xiaoqiao Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Xiaowen Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Xiue Jin
- Hubei Provincial Institute of Veterinary Drug Control, Wuhan, PR China
| | - Xiliang Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China.
| | - Xiaoping Pan
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, PR China
| | - Dingren Bi
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| |
Collapse
|
31
|
Saadane A, Mast N, Trichonas G, Chakraborty D, Hammer S, Busik JV, Grant MB, Pikuleva IA. Retinal Vascular Abnormalities and Microglia Activation in Mice with Deficiency in Cytochrome P450 46A1-Mediated Cholesterol Removal. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 189:405-425. [PMID: 30448403 DOI: 10.1016/j.ajpath.2018.10.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/27/2018] [Accepted: 10/15/2018] [Indexed: 12/11/2022]
Abstract
CYP46A1 is the cytochrome P450 enzyme that converts cholesterol to 24-hydroxycholesterol, a cholesterol elimination product and a potent liver X receptor (LXR) ligand. We conducted retinal characterizations of Cyp46a1-/- mice that had normal fasting blood glucose levels but up to a 1.8-fold increase in retinal cholesterol. The retina of Cyp46a1-/- mice exhibited venous beading and tortuosity, microglia/macrophage activation, and increased vascular permeability, features commonly associated with diabetic retinopathy. The expression of Lxrα and Lxrβ was increased in both the whole Cyp46a1-/- retina and retinal macroglia/macrophages. The LXR-target genes were affected as well, primarily in activated microglial cells and macrophages. In the latter, the LXR-transactivated genes (Abca1, Abcg1, Apod, Apoe, Mylip, and Arg2) were up-regulated; similarly, there was an up-regulation of the LXR-transrepressed genes (Ccl2, Ptgs2, Cxcl1, Il1b, Il6, Nos2, and Tnfa). For comparison, gene expression was investigated in bone marrow-derived macrophages from Cyp46a1-/- mice as well as retinal and bone marrow-derived macrophages from Cyp27a1-/- and Cyp27a1-/-Cyp46a1-/- mice. CYP46A1 expression was detected in retinal endothelial cells, and this expression was increased in the proinflammatory environment. Retinal Cyp46a1-/- phosphoproteome revealed altered phosphorylation of 30 different proteins, including tight junction protein zonula occludens 1 and aquaporin 4. Collectively, the data obtained establish metabolic and regulatory significance of CYP46A1 for the retina and suggest pharmacologic activation of CYP46A1 as a potential therapeutic approach to dyslipidemia-induced retinal damage.
Collapse
Affiliation(s)
- Aicha Saadane
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio
| | - Natalia Mast
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio
| | - George Trichonas
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio
| | | | - Sandra Hammer
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Julia V Busik
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Maria B Grant
- Department of Ophthalmology, University of Alabama, Birmingham, Alabama
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, the University Hospitals, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
32
|
Socha BM, Łada P, Szczepańska AA, Łupicka M, Korzekwa AJ. The influence of experimentally induced endometritis on the PPAR expression profile in the bovine endometrium. Theriogenology 2018; 122:74-83. [PMID: 30243137 DOI: 10.1016/j.theriogenology.2018.09.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/06/2018] [Accepted: 09/12/2018] [Indexed: 02/06/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors of the nuclear receptor superfamily. The PPARs activity can be modified e.g. by arachidonic acid metabolites. Escherichia coli (E. coli) is one of the main infectious agent of endometritis in dairy cows. We hypothesized that PPAR expression profile change in the bovine endometrium under the influence of LPS or E. coli. In experiment 1, endometrial explants were obtained post mortem from heifers and incubated without (control) or with LPS for 12, 24, 48, 72 and 96 h. In experiment 2, heifers were intrauterine infused with 0.9% NaCl (control) or with E. coli suspension in 0.9% NaCl. Endometrial biopsies were performed before (0 h) and 12, 24, 48, 72, 96 h after the infusions. In experiment 1, the increase in protein expression was observed for PPARα 48 h, for PPARβ/δ 24, 72 and 96 h, whereas for PPARγ 12, 24 and 96 h after LPS treatment relative to the control groups. In experiment 2, the up-regulation in protein expression was observed for PPARα 48 and 72 h, for PPARβ/δ 72 and 96 h, for PPARγ1 and PPARγ2 12 and 96 h after the intrauterine infusion with E. coli suspension compared to the control group. Changes in mRNA and protein PPAR expression profile in endometrial explants under the exposure of LPS indicate participation of these nuclear receptors in signal transduction during stimulation with LPS. The patterns of mRNA and protein PPAR expression in endometrial bioptates suggest that during experimentally induced endometritis in vivo, PPARs role may be connected both with enhancement of inflammation as well restoring physiological conditions in uterus.
Collapse
Affiliation(s)
- B M Socha
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences (IARFR PAS), Tuwima Str. 10, 10-747, Olsztyn, Poland
| | - P Łada
- Veterinary Clinic 3VET, Ludowa Str. 78/3, 18-200 Wysokie Mazowieckie, Poland
| | - A A Szczepańska
- Department of Biodiversity Protection, IARFR PAS, Tuwima Str. 10, 10-747, Olsztyn, Poland
| | - M Łupicka
- Department of Biodiversity Protection, IARFR PAS, Tuwima Str. 10, 10-747, Olsztyn, Poland
| | - A J Korzekwa
- Department of Biodiversity Protection, IARFR PAS, Tuwima Str. 10, 10-747, Olsztyn, Poland.
| |
Collapse
|
33
|
Zhang Y, Gupta S, Ilstad-Minnihan A, Ayyangar S, Hay AD, Pascual V, Ilowite NT, Macaubas C, Mellins ED. Interleukin-1 in monocyte activation phenotypes in systemic juvenile idiopathic arthritis: Observations from a clinical trial of rilonacept, an interleukin-1 inhibitor. Clin Immunol 2018; 194:9-18. [PMID: 29928998 DOI: 10.1016/j.clim.2018.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 05/18/2018] [Accepted: 06/16/2018] [Indexed: 01/14/2023]
Abstract
Systemic juvenile idiopathic arthritis (sJIA) is a childhood rheumatic disease of unknown origin. Dysregulated innate immunity is implicated in disease pathology. We investigated if IL-1 inhibition affects circulating cytokines and monocyte gene expression. CD14+ monocytes from patients in the RAPPORT trial were analyzed by RT-PCR for expression of IL1B and transcription factors associated with monocyte activation. Serum IL-1ra decreased with treatment, and IL-18BP transiently increased. Serum levels of IL-1β, IL-6, IL-10 and IL-18 were unchanged. IRF5 and STAT6 were decreased, and PPARG was increased, independent of clinical response, and may represent a skew toward a PPARG-driven M2-like phenotype. IL1B expression was decreased in early clinical responders. A transient increase in STAT1, and a decrease in SOCS1 preceded the reduction in IL1B in early clinical responders. Changes in IL1B/STAT1/SOCS1 could be associated with crosstalk between IL-1 and IFN pathways in sJIA. These transcriptional changes might be useful as drug response biomarkers.
Collapse
Affiliation(s)
- Yujuan Zhang
- Department of Pediatrics, Program in Immunology, Stanford University, Stanford, CA, USA
| | - Saloni Gupta
- Department of Pediatrics, Program in Immunology, Stanford University, Stanford, CA, USA
| | | | - Sashi Ayyangar
- Department of Pediatrics, Program in Immunology, Stanford University, Stanford, CA, USA
| | - Arielle D Hay
- Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Norman T Ilowite
- Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Claudia Macaubas
- Department of Pediatrics, Program in Immunology, Stanford University, Stanford, CA, USA.
| | - Elizabeth D Mellins
- Department of Pediatrics, Program in Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
34
|
Cheung KT, Sze DMY, Chan KH, Leung PHM. Involvement of caspase-4 in IL-1 beta production and pyroptosis in human macrophages during dengue virus infection. Immunobiology 2018; 223:356-364. [DOI: 10.1016/j.imbio.2017.10.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 10/19/2017] [Indexed: 12/23/2022]
|
35
|
Al-Quraishy S, Dkhil MA, Abdel-Baki AAS, Delic D, Wunderlich F. Protective vaccination alters gene expression of the liver of Balb/c mice in response to early prepatent blood-stage malaria of Plasmodium chabaudi. Parasitol Res 2018; 117:1115-1129. [DOI: 10.1007/s00436-018-5789-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/25/2018] [Indexed: 12/19/2022]
|
36
|
Tsai SY, Catts VS, Fullerton JM, Corley SM, Fillman SG, Weickert CS. Nuclear Receptors and Neuroinflammation in Schizophrenia. MOLECULAR NEUROPSYCHIATRY 2018; 3:181-191. [PMID: 29888229 DOI: 10.1159/000485565] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/21/2017] [Indexed: 01/23/2023]
Abstract
Introduction Several nuclear receptor family members have been associated with schizophrenia and inflammation. Vitamins A and D exert anti-inflammatory actions, but their receptors (mainly nuclear receptors) have not been extensively studied in either schizophrenia brains or in association with neuroinflammation. We examined the expression of vitamin A (RARs and RXRs) and vitamin D and protein disulphide-isomerase A3 (PDIA3) receptors, as well as nuclear orphan receptors (NR4As), in the context of elevated cytokine expression in the dorsolateral prefrontal cortex (DLPFC). Methods mRNA levels of nuclear receptors were measured in DLPFC tissues via RT-qPCR. ANCOVAs comparing high inflammation schizophrenia, low inflammation schizophrenia and low inflammation control groups were performed. Results RARG, RXRB, NR4A1 and NR4A3 transcripts showed significant differential expression across the three groups (ANCOVA p = 0.02-0.001). Post hoc testing revealed significant reductions in RARG expression in schizophrenia with low inflammation compared to schizophrenia with high inflammation and to controls, and RXRB mRNA was significantly reduced in schizophrenia with low inflammation compared to controls. NR4A1 and NR4A3 mRNAs were decreased in schizophrenia with high inflammation compared to schizophrenia with low inflammation, with NR4A1 also significantly different to controls. Conclusion In schizophrenia, changes in nuclear receptor mRNA levels involved with mediating actions of vitamin A derivatives vary according to the inflammatory state of brains.
Collapse
Affiliation(s)
- Shan-Yuan Tsai
- Schizophrenia Research Institute, Sydney, New South Wales, Australia.,Neuroscience Research Australia, Sydney, New South Wales, Australia.,School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Vibeke S Catts
- Schizophrenia Research Institute, Sydney, New South Wales, Australia.,Neuroscience Research Australia, Sydney, New South Wales, Australia.,School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Janice M Fullerton
- Schizophrenia Research Institute, Sydney, New South Wales, Australia.,Neuroscience Research Australia, Sydney, New South Wales, Australia.,School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Susan M Corley
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Stuart G Fillman
- Schizophrenia Research Institute, Sydney, New South Wales, Australia.,Neuroscience Research Australia, Sydney, New South Wales, Australia.,School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney, New South Wales, Australia.,Neuroscience Research Australia, Sydney, New South Wales, Australia.,School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
37
|
Xiao J, Zhang J, Zhao Y, Huang W, Guo Z, Su B, Guo Q. Sex differences of steroid receptor coactivator-1 expression after spinal cord injury in mice. Neurol Res 2017; 39:1022-1027. [DOI: 10.1080/01616412.2017.1367077] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Jiayu Xiao
- Student Brigade, Third Military Medical University, Chongqing, China
| | - Jiqiang Zhang
- Chongqing Key Laboratory of Neurobiology, Department of Neurobiology, Third Military Medical University, Chongqing, China
| | - Yangang Zhao
- Chongqing Key Laboratory of Neurobiology, Department of Neurobiology, Third Military Medical University, Chongqing, China
| | - Wenjie Huang
- Student Brigade, Third Military Medical University, Chongqing, China
| | - Zhikai Guo
- Student Brigade, Third Military Medical University, Chongqing, China
| | - Bingyin Su
- Development and Regeneration Key Lab of Sichuan Province, Department of Anatomy and Histology and Embryology, Department of Pathology, Chengdu Medical College, Chengdu, China
| | - Qiang Guo
- Chongqing Key Laboratory of Neurobiology, Department of Neurobiology, Third Military Medical University, Chongqing, China
| |
Collapse
|
38
|
Luo M, Yeruva S, Liu Y, Chodisetti G, Riederer B, Menon MB, Tachibana K, Doi T, Seidler UE. IL-1β-Induced Downregulation of the Multifunctional PDZ Adaptor PDZK1 Is Attenuated by ERK Inhibition, RXRα, or PPARα Stimulation in Enterocytes. Front Physiol 2017; 8:61. [PMID: 28223944 PMCID: PMC5293818 DOI: 10.3389/fphys.2017.00061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 01/23/2017] [Indexed: 12/21/2022] Open
Abstract
Background: The PDZ adaptor protein PDZK1 modulates the membrane expression and function of a variety of intestinal receptors and ion/nutrient transporters. Its expression is strongly decreased in inflamed intestinal mucosa of mice and IBD patients. Aim and Methods: We investigated whether the inflammation-associated PDZK1 downregulation is a direct consequence of proinflammatory cytokine release by treating intestinal Caco-2BBE cells with TNF-α, IFN-γ, and IL-1β, and analysing PDZK1 promotor activity, mRNA and protein expression. Results: IL-1β was found to significantly decrease PDZK1 promoter activity, mRNA and protein expression in Caco-2BBE cells. A distal region of the hPDZK1 promoter was identified to be important for basal expression and IL-1β-responsiveness. This region harbors the retinoid acid response element RARE as well as binding sites for transcription factors involved in IL-β downstream signaling. ERK1/2 inhibition by the specific MEK1/2 inhibitors PD98059/U0126 significantly attenuated the IL-1β mediated downregulation of PDZK1, while NF-κB, p38 MAPK, and JNK inhibition did not. Expression of the nuclear receptors RXRα and PPARα was decreased in inflamed colonic-mucosa of ulcerative colitis patients and in IL-1β-treated Caco2-BBE cells. Moreover, the RAR/RXR ligand 9-cis retinoic acid and the PPARα-agonist GW7647 stimulated PDZK1 mRNA and protein expression and attenuated IL-1β-mediated inhibition. Conclusions: The strong decrease in PDZK1 expression during intestinal inflammation may be in part a consequence of IL-1β-mediated RXRα and PPARα repression and can be attenuated by agonists for either nuclear receptor, or by ERK1/2 inhibition. The negative consequences of inflammation-induced PDZK1 downregulation on epithelial transport-function may thus be amenable to pharmacological therapy.
Collapse
Affiliation(s)
- Min Luo
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical SchoolHannover, Germany; Department of Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Sunil Yeruva
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School Hannover, Germany
| | - Yongjian Liu
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical SchoolHannover, Germany; Department of Endocrinology, the Second Affiliated Hospital of Chongqing Medical UniversityChongqing, China
| | - Giriprakash Chodisetti
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School Hannover, Germany
| | - Brigitte Riederer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School Hannover, Germany
| | - Manoj B Menon
- Department of Biochemistry, Hannover Medical School Hannover, Germany
| | - Keisuke Tachibana
- Laboratory of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Osaka University Osaka, Japan
| | - Takefumi Doi
- Laboratory of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Osaka University Osaka, Japan
| | - Ursula E Seidler
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School Hannover, Germany
| |
Collapse
|
39
|
Niu Z, Tang J, Zhang W, Chen Y, Huang Y, Chen B, Li J, Shen P. Caspase-1 promotes monocyte-macrophage differentiation by repressing PPARγ. FEBS J 2017; 284:568-585. [PMID: 28052562 DOI: 10.1111/febs.13998] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 12/19/2016] [Accepted: 01/03/2017] [Indexed: 12/24/2022]
Abstract
Monocyte-to-macrophage differentiation is tightly controlled in vivo, as disruption of the normal differentiation program can lead to diverse disorders. Caspase-1, the first identified member of the caspase family, regulates differentiation in various cell types such as Th17 cells and adipocytes. However, the contribution of caspase-1 in monocyte-macrophage differentiation remains elusive. Here we report that caspase-1 is significantly downregulated in leukemia cells from patients with acute monocytic leukemia. By using the phorbol 12-myristate 13-acetate-induced cell differentiation model, we found that caspase-1 activation was required for the differentiation of human monocytes to macrophages. Further analysis of peroxisome proliferator-activated receptor γ (PPARγ) protein levels revealed that the monocyte-macrophage differentiation program could be divided into two stages. Caspase-1-mediated downregulation of PPARγ was important in the late stage of monocyte-macrophage differentiation; however, PPARγ protein levels had little effect on the early stage differentiation. Accumulation of PPARγ protein by troglitazone treatment potently suppressed the late stage of macrophage differentiation, which might be linked to inhibition of nuclear factor-κB activity. The data provide a plausible mechanistic basis by which caspase-1 promotes the differentiation of macrophages from monocytes.
Collapse
Affiliation(s)
- Zhiyuan Niu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, China
| | - Jiajin Tang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, China
| | - Wenlong Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, China
| | - Yongjun Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, China
| | - Yahong Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, China
| | - Bing Chen
- Department of Hematology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, China
| | - Jiahong Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, China
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, China
| |
Collapse
|
40
|
Hatami M, Saidijam M, Yadegarzari R, Borzuei S, Soltanian A, Arian MS, Goodarzi MT. Peroxisome Proliferator-Activated Receptor-γGene Expression and Its Association with Oxidative Stress in Patients with Metabolic Syndrome. Chonnam Med J 2016; 52:201-6. [PMID: 27689030 PMCID: PMC5040769 DOI: 10.4068/cmj.2016.52.3.201] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 05/02/2016] [Accepted: 05/10/2016] [Indexed: 12/11/2022] Open
Abstract
Regulation of the peroxisome proliferator-activated receptor-γ (PPAR-γ) gene plays an important role in controlling the metabolism of lipids and inflammatory processes. Therefore, it can be associated with the pathogenesis of metabolic syndrome (MetS). The purpose of this study was to determine the expression of this gene in peripheral blood mononuclear cells (PBMC) in patients with metabolic syndrome. Using real-time polymerase chain reaction (PCR), mRNA expression of PPAR-γ was found in PBMC from 37 subjects with MetS and 30 healthy controls. Serum levels of glucose and lipid profiles were measured. The total antioxidant capacity (TAC) was measured using the ferric reducing ability of plasma (FRAP) test. Malondialdehyde (MDA) was determined using a fluorimetric method. Total oxidant status (TOS) in serum was assayed according to oxidation of ferric to ferrous in the presence of methyl orange. Super oxide dismutase (SOD) activity was measured using a Randox kit. Expression of PPAR-γ gene was significantly increased in patients with MetS compared to the control subjects (p=0.002). There was no difference in serum levels of TAC, MDA and SOD between the two study groups, but a significant difference was observed in the TOS (p=0.03). Serum levels of triglycerides and glucose were significantly higher in subjects with MetS. According to the results of our study, an increase in the expression of PPAR-γ in subjects with MetS indicated a possible role of PPAR-γ in the pathogenesis of this disease.
Collapse
Affiliation(s)
- Mehdi Hatami
- Department of Clinical Biochemistry, Medical School, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Reza Yadegarzari
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shiva Borzuei
- Department of Internal Medicine, Medical School, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Soltanian
- Department of Biostatistics, Health School, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marzieh Safi Arian
- Department of Clinical Biochemistry, Medical School, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Taghi Goodarzi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
41
|
Scharping NE, Menk AV, Moreci RS, Whetstone RD, Dadey RE, Watkins SC, Ferris RL, Delgoffe GM. The Tumor Microenvironment Represses T Cell Mitochondrial Biogenesis to Drive Intratumoral T Cell Metabolic Insufficiency and Dysfunction. Immunity 2016; 45:374-88. [PMID: 27496732 PMCID: PMC5207350 DOI: 10.1016/j.immuni.2016.07.009] [Citation(s) in RCA: 513] [Impact Index Per Article: 64.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/16/2016] [Accepted: 06/08/2016] [Indexed: 02/06/2023]
Abstract
Although tumor-specific T cells recognize cancer cells, they are often rendered dysfunctional due to an immunosuppressive microenvironment. Here we showed that T cells demonstrated persistent loss of mitochondrial function and mass when infiltrating murine and human tumors, an effect specific to the tumor microenvironment and not merely caused by activation. Tumor-infiltrating T cells showed a progressive loss of PPAR-gamma coactivator 1α (PGC1α), which programs mitochondrial biogenesis, induced by chronic Akt signaling in tumor-specific T cells. Reprogramming tumor-specific T cells through enforced expression of PGC1α resulted in superior intratumoral metabolic and effector function. Our data support a model in which signals in the tumor microenvironment repress T cell oxidative metabolism, resulting in effector cells with metabolic needs that cannot be met. Our studies also suggest that modulation or reprogramming of the altered metabolism of tumor-infiltrating T cells might represent a potential strategy to reinvigorate dysfunctional T cells for cancer treatment.
Collapse
Affiliation(s)
- Nicole E Scharping
- Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ashley V Menk
- Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15261, USA
| | - Rebecca S Moreci
- Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15261, USA
| | - Ryan D Whetstone
- Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Rebekah E Dadey
- Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Simon C Watkins
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Robert L Ferris
- Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Greg M Delgoffe
- Tumor Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15261, USA; Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
42
|
Zhang L, Tian F, Gao X, Wang X, Wu C, Li N, Li J. N-3 Polyunsaturated Fatty Acids Improve Liver Lipid Oxidation-Related Enzyme Levels and Increased the Peroxisome Proliferator-Activated Receptor α Expression Level in Mice Subjected to Hemorrhagic Shock/Resuscitation. Nutrients 2016; 8:237. [PMID: 27110821 PMCID: PMC4848705 DOI: 10.3390/nu8040237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/09/2016] [Accepted: 04/15/2016] [Indexed: 01/02/2023] Open
Abstract
Appropriate metabolic interventions after hemorrhagic shock/resuscitation injury have not yet been identified. We aimed to examine the effects of fish oil on lipid metabolic intervention after hemorrhagic shock/resuscitation. Firstly, 48 C57BL/6 mice were assigned to six groups (n = 8 per group). The sham group did not undergo surgery, while mice in the remaining groups were sacrificed 1–5 days after hemorrhagic shock/resuscitation. In the second part, mice were treated with saline or fish oil (n = 8 per group) five days after injury. We determined serum triglyceride levels and liver tissues were collected and prepared for qRT-PCR or Western blot analysis. We found that triglyceride levels were increased five days after hemorrhagic shock/resuscitation, but decreased after addition of fish oil. After injury, the protein and gene expression of carnitine palmitoyltransferase 1A, fatty acid transport protein 1, and peroxisome proliferator-activated receptor-α decreased significantly in liver tissue. In contrast, after treatment with fish oil, the expression levels of these targets increased compared with those in the saline group. The present results suggest n-3 polyunsaturated fatty acids could improve lipid oxidation-related enzymes in liver subjected to hemorrhagic shock/resuscitation. This function is possibly accomplished through activating the peroxisome proliferator-activated receptor-α pathway.
Collapse
Affiliation(s)
- Li Zhang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Feng Tian
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Xuejin Gao
- Department of General Surgery, Jinling Hospital, Clinical College of Southern Medical University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Xinying Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Chao Wu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Ning Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| | - Jieshou Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Zhongshan East Road 305, Nanjing 210002, China.
| |
Collapse
|
43
|
Aho V, Ollila HM, Kronholm E, Bondia-Pons I, Soininen P, Kangas AJ, Hilvo M, Seppälä I, Kettunen J, Oikonen M, Raitoharju E, Hyötyläinen T, Kähönen M, Viikari JSA, Härmä M, Sallinen M, Olkkonen VM, Alenius H, Jauhiainen M, Paunio T, Lehtimäki T, Salomaa V, Orešič M, Raitakari OT, Ala-Korpela M, Porkka-Heiskanen T. Prolonged sleep restriction induces changes in pathways involved in cholesterol metabolism and inflammatory responses. Sci Rep 2016; 6:24828. [PMID: 27102866 PMCID: PMC4840329 DOI: 10.1038/srep24828] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 04/05/2016] [Indexed: 12/22/2022] Open
Abstract
Sleep loss and insufficient sleep are risk factors for cardiometabolic diseases, but data on how insufficient sleep contributes to these diseases are scarce. These questions were addressed using two approaches: an experimental, partial sleep restriction study (14 cases and 7 control subjects) with objective verification of sleep amount, and two independent epidemiological cohorts (altogether 2739 individuals) with questions of sleep insufficiency. In both approaches, blood transcriptome and serum metabolome were analysed. Sleep loss decreased the expression of genes encoding cholesterol transporters and increased expression in pathways involved in inflammatory responses in both paradigms. Metabolomic analyses revealed lower circulating large HDL in the population cohorts among subjects reporting insufficient sleep, while circulating LDL decreased in the experimental sleep restriction study. These findings suggest that prolonged sleep deprivation modifies inflammatory and cholesterol pathways at the level of gene expression and serum lipoproteins, inducing changes toward potentially higher risk for cardiometabolic diseases.
Collapse
Affiliation(s)
- Vilma Aho
- Department of Physiology, Faculty of Medicine, University of Helsinki, Finland
| | - Hanna M Ollila
- Department of Physiology, Faculty of Medicine, University of Helsinki, Finland
- Genomics and Biomarkers unit and Institute for Molecular Medicine FIMM, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry, University of Helsinki and Helsinki University Hospital, Finland
- Stanford University Center for Sleep Sciences, Palo Alto, CA, USA
| | - Erkki Kronholm
- Department of Chronic Disease Prevention, Population Studies Unit, National Institute for Health and Welfare, Turku, Finland
| | - Isabel Bondia-Pons
- VTT Technical Research Centre of Finland, Espoo, Finland
- Steno Diabetes Center A/S, Gentofte, Denmark
| | - Pasi Soininen
- Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Antti J Kangas
- Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland
| | - Mika Hilvo
- VTT Technical Research Centre of Finland, Espoo, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories, and University of Tampere, School of Medicine, Tampere, Finland
| | - Johannes Kettunen
- Genomics and Biomarkers unit and Institute for Molecular Medicine FIMM, National Institute for Health and Welfare, Helsinki, Finland
- Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Mervi Oikonen
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Fimlab Laboratories, and University of Tampere, School of Medicine, Tampere, Finland
| | - Tuulia Hyötyläinen
- VTT Technical Research Centre of Finland, Espoo, Finland
- Steno Diabetes Center A/S, Gentofte, Denmark
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Jorma S A Viikari
- Department of Medicine, University of Turku, and Division of Medicine, Turku University Hospital, Turku, Finland
| | - Mikko Härmä
- Brain and Work Research Centre, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Mikael Sallinen
- Brain and Work Research Centre, Finnish Institute of Occupational Health, Helsinki, Finland
- Agora Center, University of Jyväskylä, Jyväskylä, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Institute of Biomedicine, Anatomy, University of Helsinki, Finland
| | - Harri Alenius
- Unit of Excellence for Immunotoxicology, Finnish Institute of Occupational Health, Helsinki, Finland
| | - Matti Jauhiainen
- Genomics and Biomarkers unit and Institute for Molecular Medicine FIMM, National Institute for Health and Welfare, Helsinki, Finland
| | - Tiina Paunio
- Genomics and Biomarkers unit and Institute for Molecular Medicine FIMM, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry, University of Helsinki and Helsinki University Hospital, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and University of Tampere, School of Medicine, Tampere, Finland
| | - Veikko Salomaa
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland
| | - Matej Orešič
- VTT Technical Research Centre of Finland, Espoo, Finland
- Steno Diabetes Center A/S, Gentofte, Denmark
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Mika Ala-Korpela
- Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Oulu University Hospital, Oulu, Finland
- Computational Medicine, School of Social and Community Medicine &Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | | |
Collapse
|
44
|
Bertoni G, Trevisi E, Lombardelli R. Some new aspects of nutrition, health conditions and fertility of intensively reared dairy cows. ITALIAN JOURNAL OF ANIMAL SCIENCE 2016. [DOI: 10.4081/ijas.2009.491] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Shavva VS, Mogilenko DA, Bogomolova AM, Nikitin AA, Dizhe EB, Efremov AM, Oleinikova GN, Perevozchikov AP, Orlov SV. PPARγ Represses Apolipoprotein A-I Gene but Impedes TNFα-Mediated ApoA-I Downregulation in HepG2 Cells. J Cell Biochem 2016; 117:2010-22. [DOI: 10.1002/jcb.25498] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 01/25/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Vladimir S. Shavva
- Department of Biochemistry; Institute of Experimental Medicine; Russian Academy of Medical Sciences; St. Petersburg Russia
- Department of Embryology; St. Petersburg State University; St. Petersburg Russia
| | - Denis A. Mogilenko
- Department of Biochemistry; Institute of Experimental Medicine; Russian Academy of Medical Sciences; St. Petersburg Russia
- Department of Embryology; St. Petersburg State University; St. Petersburg Russia
| | | | - Artemy A. Nikitin
- Department of Biochemistry; Institute of Experimental Medicine; Russian Academy of Medical Sciences; St. Petersburg Russia
- Department of Biochemistry; St. Petersburg State University; St. Petersburg Russia
| | - Ella B. Dizhe
- Department of Biochemistry; Institute of Experimental Medicine; Russian Academy of Medical Sciences; St. Petersburg Russia
| | - Alexander M. Efremov
- Department of Biochemistry; Institute of Experimental Medicine; Russian Academy of Medical Sciences; St. Petersburg Russia
- Department of Embryology; St. Petersburg State University; St. Petersburg Russia
| | - Galina N. Oleinikova
- Department of Biochemistry; Institute of Experimental Medicine; Russian Academy of Medical Sciences; St. Petersburg Russia
| | - Andrej P. Perevozchikov
- Department of Biochemistry; Institute of Experimental Medicine; Russian Academy of Medical Sciences; St. Petersburg Russia
- Department of Embryology; St. Petersburg State University; St. Petersburg Russia
| | - Sergey V. Orlov
- Department of Biochemistry; Institute of Experimental Medicine; Russian Academy of Medical Sciences; St. Petersburg Russia
- Department of Embryology; St. Petersburg State University; St. Petersburg Russia
| |
Collapse
|
46
|
Keller R, Klein M, Thomas M, Dräger A, Metzger U, Templin MF, Joos TO, Thasler WE, Zell A, Zanger UM. Coordinating Role of RXRα in Downregulating Hepatic Detoxification during Inflammation Revealed by Fuzzy-Logic Modeling. PLoS Comput Biol 2016; 12:e1004431. [PMID: 26727233 PMCID: PMC4699813 DOI: 10.1371/journal.pcbi.1004431] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 07/05/2015] [Indexed: 12/31/2022] Open
Abstract
During various inflammatory processes circulating cytokines including IL-6, IL-1β, and TNFα elicit a broad and clinically relevant impairment of hepatic detoxification that is based on the simultaneous downregulation of many drug metabolizing enzymes and transporter genes. To address the question whether a common mechanism is involved we treated human primary hepatocytes with IL-6, the major mediator of the acute phase response in liver, and characterized acute phase and detoxification responses in quantitative gene expression and (phospho-)proteomics data sets. Selective inhibitors were used to disentangle the roles of JAK/STAT, MAPK, and PI3K signaling pathways. A prior knowledge-based fuzzy logic model comprising signal transduction and gene regulation was established and trained with perturbation-derived gene expression data from five hepatocyte donors. Our model suggests a greater role of MAPK/PI3K compared to JAK/STAT with the orphan nuclear receptor RXRα playing a central role in mediating transcriptional downregulation. Validation experiments revealed a striking similarity of RXRα gene silencing versus IL-6 induced negative gene regulation (rs = 0.79; P<0.0001). These results concur with RXRα functioning as obligatory heterodimerization partner for several nuclear receptors that regulate drug and lipid metabolism. During inflammation, circulating proinflammatory cytokines such as TNFα, IL-1ß, and IL-6, which are produced by, e.g., Kupffer cells, macrophages, or tumor cells, play important roles in hepatocellular signaling pathways and in the regulation of cellular homeostasis. In particular, these cytokines are responsible for the acute phase response (APR) but also for a dramatic reduction of drug detoxification capacity due to impaired expression of numerous genes coding for drug metabolic enzymes and transporters. Here we used high-throughput (phospho-)proteomic and gene expression data to investigate the impact of canonical signaling pathways in mediating IL-6-induced downregulation of drug metabolism related genes. We performed chemical inhibition perturbations to show that most of the IL-6 effects on gene expression are mediated through the MAPK and PI3K/AKT pathways. We constructed a prior knowledge network as basis for a fuzzy logic model that was trained with extensive gene expression data to identify critical regulatory nodes. Our results suggest that the nuclear receptor RXRα plays a central role, which was convincingly validated by RXRα gene silencing experiments. This work shows how computational modeling can support identifying decisive regulatory events from large-scale experimental data.
Collapse
Affiliation(s)
- Roland Keller
- Center for Bioinformatics Tuebingen (ZBIT), University of Tuebingen, Tuebingen, Germany
| | - Marcus Klein
- Dr. Margarete Fischer Bosch-Institute of Clinical Pharmacology, Stuttgart
- University of Tuebingen, Tuebingen, Germany
| | - Maria Thomas
- Dr. Margarete Fischer Bosch-Institute of Clinical Pharmacology, Stuttgart
- University of Tuebingen, Tuebingen, Germany
| | - Andreas Dräger
- Center for Bioinformatics Tuebingen (ZBIT), University of Tuebingen, Tuebingen, Germany
- Systems Biology Research Group, University of California, San Diego, La Jolla, California, United States of America
| | - Ute Metzger
- NMI Institute of Natural and Medical Sciences, Reutlingen, Germany
| | | | - Thomas O. Joos
- NMI Institute of Natural and Medical Sciences, Reutlingen, Germany
| | - Wolfgang E. Thasler
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the University of Munich, Munich, Germany
| | - Andreas Zell
- Center for Bioinformatics Tuebingen (ZBIT), University of Tuebingen, Tuebingen, Germany
| | - Ulrich M. Zanger
- Dr. Margarete Fischer Bosch-Institute of Clinical Pharmacology, Stuttgart
- University of Tuebingen, Tuebingen, Germany
- * E-mail:
| |
Collapse
|
47
|
Fan A, Wang Q, Yuan Y, Cheng J, Chen L, Guo X, Li Q, Chen B, Huang X, Huang Q. Liver X receptor-α and miR-130a-3p regulate expression of sphingosine 1-phosphate receptor 2 in human umbilical vein endothelial cells. Am J Physiol Cell Physiol 2015; 310:C216-26. [PMID: 26669941 DOI: 10.1152/ajpcell.00102.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 11/23/2015] [Indexed: 02/07/2023]
Abstract
Recent studies have shown that activation of liver X receptors (LXRs) attenuates the development of atherosclerosis, not only by regulating lipid metabolism but also by suppressing inflammatory signaling. Sphingosine 1-phosphate receptor 2 (S1PR2), an important inflammatory gene product, plays a role in the development of various inflammatory diseases. It was proposed that S1PR2 might be regulated by LXR-α. In the present study, the effect of LXR-α on tumor necrosis factor-α (TNF-α)-induced S1PR2 expression in human umbilical vein endothelial cells (HUVECs) was investigated and the underlying mechanism was explored. The results demonstrated that TNF-α led to an increase in S1PR2 expression and triggered a downregulation of LXR-α expression in HUVECs as well. Downregulation of LXR-α with specific small interfering RNA (siRNA) remarkably enhanced the primary as well as TNF-α-induced expression of S1PR2 in HUVECs. Activation of LXR-α by agonist GW3965 inhibited both primary and TNF-α-induced S1PR2 expression. GW3965 also attenuated S1PR2-induced endothelial barrier dysfunction. The data further showed that TNF-α induced a significant decrease in miR-130a-3p expression. Overexpression of miR-130a-3p with mimic product reduced S1PR2 protein expression, and inhibition of miR-130a-3p by specific inhibitor resulted in an increase in S1PR2 protein expression. Furthermore, activation of LXRs with agonist enhanced the expression of miR-130a-3p, and knockdown of LXR-α by siRNA suppressed miR-130a-3p expression. These results suggest that LXR-α might downregulate S1PR2 expression via miR-130a-3p in quiescent HUVECs. Stimulation of TNF-α attenuates the activity of LXR-α and results in enhanced S1PR2 expression.
Collapse
Affiliation(s)
- Aihui Fan
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China; Department of Physiology, Guangdong Medical College, Dongguan, People's Republic of China; and
| | - Qian Wang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Yongjun Yuan
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Jilun Cheng
- Department of Pharmacology, Guangdong Medical College, Dongguan, People's Republic of China
| | - Lixian Chen
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiaohua Guo
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Qiang Li
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Bo Chen
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| | - Xuliang Huang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China;
| | - Qiaobing Huang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
48
|
Szalowska E, Pronk TE, Peijnenburg AA. Cyclosporin A induced toxicity in mouse liver slices is only slightly aggravated by Fxr-deficiency and co-occurs with upregulation of pro-inflammatory genes and downregulation of genes involved in mitochondrial functions. BMC Genomics 2015; 16:822. [PMID: 26482353 PMCID: PMC4617705 DOI: 10.1186/s12864-015-2054-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 10/12/2015] [Indexed: 12/27/2022] Open
Abstract
Background The transcription factor farnesoid X receptor (FXR) governs bile acid and energy homeostasis, is involved in inflammation, and has protective functions in the liver. In the present study we investigated the effect of Fxr deficiency in mouse precision cut liver slices (PCLS) exposed to a model hepatotoxicant cyclosporin A (CsA). It was anticipated that Fxr deficiency could aggravate toxicity of CsA in PCLS and pinpoint to novel genes/processes regulated by FXR. Methods To test this hypothesis, PCLS obtained from livers of wild type mice (WT-PCLS) and Fxr-knockout mice (FXRKO-PCLS) were treated with 40 μM CsA for 24 h and 48 h. ATP and histological assays were applied to assess the viability of PCLS. DNA microarrays combined with bioinformatics analysis were used to identify genes and processes that were affected by CsA in WT-PCLS and/or FXRKO-PCLS. In addition, WT-PCLS and FXRKO-PCLS were exposed to the endogenous FXR ligand chenodeoxycholic acid (CDCA) and subjected to q-PCR to determine whether subsets of known FXR-targets and the identified genes were regulated upon FXR activation in an FXR-dependent manner. Results No difference in viability was observed between WT-PCLS and FXRKO-PCLS upon CsA treatment. Transcriptomics data analysis revealed that CsA significantly upregulated stress-response and inflammation and significantly downregulated processes involved in lipid and glucose metabolism in WT-PCLS and FXRKO-PCLS. However, only in FXRKO-PCLS, CsA upregulated additional pro-inflammatory genes and downregulated genes related to mitochondrial functions. Furthermore, only in WT-PCLS, CDCA upregulated a subset of known FXR-target genes as well as the regulator of inflammation and mitochondrial functions peroxisome proliferator- activated receptor delta (Ppar delta). Conclusions Although FXR governs energy metabolism, no major differences in response to CsA could be observed between WT-PCLS and FXRKO-PCLS in regulation of processes involved in lipid and glucose metabolism. This finding indicates that CsA does not directly affect FXR functions in relation to the above mentioned processes. However, the more pronounced induction of pro-inflammatory genes and the downregulation of genes involved in mitochondrial functions only in FXRKO-PCLS suggest that FXR deficiency aggravates CsA-induced inflammation and impairs mitochondrial functions. Therefore, FXR can exert its hepatoprotective functions by controlling inflammation and mitochondrial functions, possibly involving an FXR-PPAR delta cross-talk. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2054-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ewa Szalowska
- RIKILT - Institute of Food Safety/Wageningen UR, Akkermaalsbos 2, P.O. Box 230, 6700 AE, Wageningen, The Netherlands. .,RIKILT-Institute of Food Safety/Wageningen UR, Akkermaalsbos 2, 6708 WB, Wageningen, The Netherlands.
| | - Tessa E Pronk
- Centre for Health Protection, National Institute for Public Health and the Environment (GZB, RIVM), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands.
| | - Ad Acm Peijnenburg
- RIKILT - Institute of Food Safety/Wageningen UR, Akkermaalsbos 2, P.O. Box 230, 6700 AE, Wageningen, The Netherlands.
| |
Collapse
|
49
|
Akbar H, Batistel F, Drackley JK, Loor JJ. Alterations in Hepatic FGF21, Co-Regulated Genes, and Upstream Metabolic Genes in Response to Nutrition, Ketosis and Inflammation in Peripartal Holstein Cows. PLoS One 2015; 10:e0139963. [PMID: 26451842 PMCID: PMC4599736 DOI: 10.1371/journal.pone.0139963] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/18/2015] [Indexed: 12/29/2022] Open
Abstract
In rodents, fibroblast growth factor 21 (FGF21) has emerged as a key metabolic regulator produced by liver. To gather preliminary data on the potential importance of FGF1, co-regulated genes, and upstream metabolic genes, we examined the hepatic mRNA expression in response to nutrition and inflammation in dairy cows. In experiment 1, induction of ketosis through feed restriction on d 5 postpartum upregulated FGF21, its co-receptor KLB, and PPARA but only elicited a numerical increase in serum FGF21 concentration. In experiment 2, cows in control (CON) or receiving 50 g/d of L-carnitine (C50) from -14 through 21 d had increased FGF21, PPARA, and NFIL3 on d 10 compared with d 2 postpartum. In contrast, compared with CON and C50, 100 g/d L-carnitine (C100) resulted in lower FGF21, KLB, ANGPTL4, and ARNTL expression on d 10. In experiment 3, cows were fed during the dry period either a higher-energy (OVE; 1.62 Mcal/kg DM) or lower-energy (CON; 1.34 Mcal/kg DM) diet and received 0 (OVE:N, CON:N) or 200 μg of LPS (OVE:Y, CON:Y) into the mammary gland at d 7 postpartum. For FGF21 mRNA expression in CON, the LPS challenge (CON:Y) prevented a decrease in expression between d 7 and 14 postpartum such that cows in CON:N had a 4-fold lower expression on d 14 compared with d 7. The inflammatory stimulus induced by LPS in CON:Y resulted in upregulation of PPARA on d 14 to a similar level as cows in OVE:N. In OVE:Y, expression of PPARA was lower than CON:N on d 7 and remained unchanged on d 14. On d 7, LPS led to a 4-fold greater serum FGF21 only in OVE but not in CON cows. In fact, OVE:Y reached the same serum FGF21 concentration as CON:N, suggesting a carryover effect of dietary energy level on signaling mechanisms within liver. Overall, results indicate that nutrition, ketosis, and inflammation during the peripartal period can alter hepatic FGF21, co-regulated genes, and upstream metabolic genes to various extents. The functional outcome of these changes merits further study, and in particular the mechanisms regulating transcription in response to changes in energy balance and feed intake.
Collapse
Affiliation(s)
- Haji Akbar
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - Fernanda Batistel
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - James K. Drackley
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, Illinois, United States of America
| | - Juan J. Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
50
|
Tarasenko TN, Singh LN, Chatterji-Len M, Zerfas PM, Cusmano-Ozog K, McGuire PJ. Kupffer cells modulate hepatic fatty acid oxidation during infection with PR8 influenza. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2391-401. [PMID: 26319418 DOI: 10.1016/j.bbadis.2015.08.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 08/20/2015] [Accepted: 08/25/2015] [Indexed: 12/30/2022]
Abstract
In response to infection, patients with inborn errors of metabolism may develop a functional deterioration termed metabolic decompensation. The biochemical hallmarks of this disruption of metabolic homeostasis are disease specific and may include acidosis, hyperammonemia or hypoglycemia. In a model system previously published by our group, we noted that during influenza infection, mice displayed a depression in hepatic mitochondrial enzymes involved in nitrogen metabolism. Based on these findings, we hypothesized that this normal adaptation may extend to other metabolic pathways, and as such, may impact various inborn errors of metabolism. Since the liver is a critical organ in inborn errors of metabolism, we carried out untargeted metabolomic profiling of livers using mass spectrometry in C57Bl/6 mice infected with influenza to characterize metabolic adaptation. Pathway analysis of metabolomic data revealed reductions in CoA synthesis, and long chain fatty acyl CoA and carnitine species. These metabolic adaptations coincided with a depression in hepatic long chain β-oxidation mRNA and protein. To our surprise, the metabolic changes observed occurred in conjunction with a hepatic innate immune response, as demonstrated by transcriptional profiling and flow cytometry. By employing an immunomodulation strategy to deplete Kupffer cells, we were able to improve the expression of multiple genes involved in β-oxidation. Based on these findings, we are the first to suggest that the role of the liver as an immunologic organ is central in the pathophysiology of hepatic metabolic decompensation in inborn errors of metabolism due to respiratory viral infection.
Collapse
Affiliation(s)
- Tatyana N Tarasenko
- Metabolism, Infection and Immunity Unit, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Larry N Singh
- Center for Mitochondrial and Epigenomic Medicine, Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Milani Chatterji-Len
- Metabolism, Infection and Immunity Unit, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Patricia M Zerfas
- Office of Research Services, Division of Veterinary Resources, National Institutes of Health, Bethesda, MD, USA
| | - Kristina Cusmano-Ozog
- Biochemical Genetics and Metabolism Laboratory, Children's National Medical Center, Washington, DC, USA
| | - Peter J McGuire
- Metabolism, Infection and Immunity Unit, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|