1
|
Gupta H, Bhandari U. Molecular Insight into Obesity-Associated Nephropathy: Clinical Implications and Possible Strategies for its Management. Curr Drug Targets 2025; 26:188-202. [PMID: 39411934 DOI: 10.2174/0113894501314788241008115712] [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: 04/29/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 04/11/2025]
Abstract
Obesity is a significant health concern due to its rapid increase worldwide. It has been linked to the pathogenic factors of renal diseases, cancer, cardiovascular diseases, hypertension, dyslipidemia, and type 2 diabetes. Notably, obesity raises the likelihood of developing chronic kidney disease (CKD), leading to higher adult mortality and morbidity rates. This study explores the molecular mechanisms that underlie obesity-associated nephropathy and its clinical implications. Obesity-Associated Nephropathy (OAN) develops and worsens due to insulin resistance and hyperinsulinemia, which promote renal sodium reabsorption, glomerular hyperfiltration, and hypertension, leading to progressive kidney damage. Renal damage is further aggravated by persistent inflammation and redox damage, mediated by adipokines and proinflammatory cytokines, such as TNF-α and IL-6. Furthermore, stimulation of the sympathetic nervous system and the renin-angiotensin- aldosterone system (RAAS) intensifies glomerular hypertension and fibrosis. These elements cause glomerular hyperfiltration, renal hypertrophy, and progressive kidney damage. Clinical manifestations of obesity-associated nephropathy include proteinuria, reduced glomerular filtration rate (GFR), and ultimately, CKD. Management strategies currently focus on lifestyle modifications, such as weight loss through diet and exercise, which have been effective in reducing proteinuria and improving GFR. Pharmacological treatments targeting metabolic pathways, including GLP-1 receptor agonists and SGLT2 inhibitors, have shown renoprotective properties. Additionally, traditional RAAS inhibitors offer therapeutic benefits. Early detection and comprehensive management of OAN are essential to prevent its progression and lessen the burden of CKD.
Collapse
Affiliation(s)
- Himani Gupta
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110062, India
| | - Uma Bhandari
- Department of Pharmacology, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
2
|
Wang X, Cheng Z, Tai W, Shi M, Ayazi M, Liu Y, Sun L, Yu C, Fan Z, Guo B, He X, Sun D, Young W, Ren Y. Targeting foamy macrophages by manipulating ABCA1 expression to facilitate lesion healing in the injured spinal cord. Brain Behav Immun 2024; 119:431-453. [PMID: 38636566 DOI: 10.1016/j.bbi.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
Spinal cord injury (SCI) triggers a complex cascade of events, including myelin loss, neuronal damage, neuroinflammation, and the accumulation of damaged cells and debris at the injury site. Infiltrating bone marrow derived macrophages (BMDMϕ) migrate to the epicenter of the SCI lesion, where they engulf cell debris including abundant myelin debris to become pro-inflammatory foamy macrophages (foamy Mϕ), participate neuroinflammation, and facilitate the progression of SCI. This study aimed to elucidate the cellular and molecular mechanisms underlying the functional changes in foamy Mϕ and their potential implications for SCI. Contusion at T10 level of the spinal cord was induced using a New York University (NYU) impactor (5 g rod from a height of 6.25 mm) in male mice. ABCA1, an ATP-binding cassette transporter expressed by Mϕ, plays a crucial role in lipid efflux from foamy cells. We observed that foamy Mϕ lacking ABCA1 exhibited increased lipid accumulation and a higher presence of lipid-accumulated foamy Mϕ as well as elevated pro-inflammatory response in vitro and in injured spinal cord. We also found that both genetic and pharmacological enhancement of ABCA1 expression accelerated lipid efflux from foamy Mϕ, reduced lipid accumulation and inhibited the pro-inflammatory response of foamy Mϕ, and accelerated clearance of cell debris and necrotic cells, which resulted in functional recovery. Our study highlights the importance of understanding the pathologic role of foamy Mϕ in SCI progression and the potential of ABCA1 as a therapeutic target for modulating the inflammatory response, promoting lipid metabolism, and facilitating functional recovery in SCI.
Collapse
Affiliation(s)
- Xi Wang
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA; Institute of Neurosciences, Fourth Military Medical University, Xi'an 710032, China; College of Life Sciences and Medicine, Northwest University, Xi'an 710069, China.
| | - Zhijian Cheng
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Wenjiao Tai
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Mingjun Shi
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Maryam Ayazi
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Yang Liu
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Li Sun
- Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Caiyong Yu
- Institute of Neurosciences, Fourth Military Medical University, Xi'an 710032, China
| | - Zhongmin Fan
- Department of Critical Care Medicine and Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Xi'an 710032, China
| | - Bin Guo
- Department of Pathology, Guizhou Medical University, Guiyang 550025, China
| | - Xijing He
- Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Dongming Sun
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA
| | - Wise Young
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA
| | - Yi Ren
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, New Brunswick, NJ 08854, USA; Department of Biomedical Sciences, Florida State University College of Medicine, 1115 West Call Street, Tallahassee, FL 32306, USA.
| |
Collapse
|
3
|
Kim J, Kim JY, Byeon HE, Kim JW, Kim HA, Suh CH, Choi S, Linton MF, Jung JY. Inhibition of Toll-like Receptors Alters Macrophage Cholesterol Efflux and Foam Cell Formation. Int J Mol Sci 2024; 25:6808. [PMID: 38928513 PMCID: PMC11203583 DOI: 10.3390/ijms25126808] [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: 05/23/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Arterial macrophage cholesterol accumulation and impaired cholesterol efflux lead to foam cell formation and the development of atherosclerosis. Modified lipoproteins interact with toll-like receptors (TLR), causing an increased inflammatory response and altered cholesterol homeostasis. We aimed to determine the effects of TLR antagonists on cholesterol efflux and foam cell formation in human macrophages. Stimulated monocytes were treated with TLR antagonists (MIP2), and the cholesterol efflux transporter expression and foam cell formation were analyzed. The administration of MIP2 attenuated the foam cell formation induced by lipopolysaccharides (LPS) and oxidized low-density lipoproteins (ox-LDL) in stimulated THP-1 cells (p < 0.001). The expression of ATP-binding cassette transporters A (ABCA)-1, ABCG-1, scavenger receptor (SR)-B1, liver X receptor (LXR)-α, and peroxisome proliferator-activated receptor (PPAR)-γ mRNA and proteins were increased (p < 0.001) following MIP2 administration. A concentration-dependent decrease in the phosphorylation of p65, p38, and JNK was also observed following MIP2 administration. Moreover, an inhibition of p65 phosphorylation enhanced the expression of ABCA1, ABCG1, SR-B1, and LXR-α. TLR inhibition promoted the cholesterol efflux pathway by increasing the expression of ABCA-1, ABCG-1, and SR-B1, thereby reducing foam cell formation. Our results suggest a potential role of the p65/NF-kB/LXR-α/ABCA1 axis in TLR-mediated cholesterol homeostasis.
Collapse
Affiliation(s)
- Jaemi Kim
- Department of Rheumatology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.K.); (C.-H.S.)
| | - Ji-Yun Kim
- Institute of Medical Science, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.-Y.K.)
| | - Hye-Eun Byeon
- Institute of Medical Science, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.-Y.K.)
| | - Ji-Won Kim
- Department of Rheumatology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.K.); (C.-H.S.)
| | - Hyoun-Ah Kim
- Department of Rheumatology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.K.); (C.-H.S.)
| | - Chang-Hee Suh
- Department of Rheumatology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.K.); (C.-H.S.)
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea;
| | - MacRae F. Linton
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
| | - Ju-Yang Jung
- Department of Rheumatology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea; (J.K.); (C.-H.S.)
| |
Collapse
|
4
|
Ding J, Nguyen AT, Lohman K, Hensley MT, Parker D, Hou L, Taylor J, Voora D, Sawyer JK, Boudyguina E, Bancks MP, Bertoni A, Pankow JS, Rotter JI, Goodarzi MO, Tracy RP, Murdoch DM, Duprez D, Rich SS, Psaty BM, Siscovick D, Newgard CB, Herrington D, Hoeschele I, Shea S, Stein JH, Patel M, Post W, Jacobs D, Parks JS, Liu Y. LXR signaling pathways link cholesterol metabolism with risk for prediabetes and diabetes. J Clin Invest 2024; 134:e173278. [PMID: 38747290 PMCID: PMC11093600 DOI: 10.1172/jci173278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 03/20/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUNDPreclinical studies suggest that cholesterol accumulation leads to insulin resistance. We previously reported that alterations in a monocyte cholesterol metabolism transcriptional network (CMTN) - suggestive of cellular cholesterol accumulation - were cross-sectionally associated with obesity and type 2 diabetes (T2D). Here, we sought to determine whether the CMTN alterations independently predict incident prediabetes/T2D risk, and correlate with cellular cholesterol accumulation.METHODSMonocyte mRNA expression of 11 CMTN genes was quantified among 934 Multi-Ethnic Study of Atherosclerosis (MESA) participants free of prediabetes/T2D; cellular cholesterol was measured in a subset of 24 monocyte samples.RESULTSDuring a median 6-year follow-up, lower expression of 3 highly correlated LXR target genes - ABCG1 and ABCA1 (cholesterol efflux) and MYLIP (cholesterol uptake suppression) - and not other CMTN genes, was significantly associated with higher risk of incident prediabetes/T2D. Lower expression of the LXR target genes correlated with higher cellular cholesterol levels (e.g., 47% of variance in cellular total cholesterol explained by ABCG1 expression). Further, adding the LXR target genes to overweight/obesity and other known predictors significantly improved prediction of incident prediabetes/T2D.CONCLUSIONThese data suggest that the aberrant LXR/ABCG1-ABCA1-MYLIP pathway (LAAMP) is a major T2D risk factor and support a potential role for aberrant LAAMP and cellular cholesterol accumulation in diabetogenesis.FUNDINGThe MESA Epigenomics and Transcriptomics Studies were funded by NIH grants 1R01HL101250, 1RF1AG054474, R01HL126477, R01DK101921, and R01HL135009. This work was supported by funding from NIDDK R01DK103531 and NHLBI R01HL119962.
Collapse
Affiliation(s)
- Jingzhong Ding
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Kurt Lohman
- Department of Medicine, Division of Cardiology, and
| | | | - Daniel Parker
- Department of Medicine, Division of Geriatrics, Duke University, Durham, North Carolina, USA
| | - Li Hou
- Department of Medicine, Division of Cardiology, and
| | - Jackson Taylor
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, Ohio, USA
| | - Deepak Voora
- Department of Medicine, Division of Cardiology, and
| | - Janet K. Sawyer
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Elena Boudyguina
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Michael P. Bancks
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Alain Bertoni
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - David M. Murdoch
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University, Durham, North Carolina, USA
| | - Daniel Duprez
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Systems and Population Health, University of Washington, Seattle, Washington, USA
| | | | - Christopher B. Newgard
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - David Herrington
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Ina Hoeschele
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, Virginia, USA
| | - Steven Shea
- Department of Medicine, Columbia University, New York, New York, USA
| | - James H. Stein
- School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Manesh Patel
- Department of Medicine, Division of Cardiology, and
| | - Wendy Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - David Jacobs
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - John S. Parks
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Yongmei Liu
- Department of Medicine, Division of Cardiology, and
| |
Collapse
|
5
|
Stefik D, Vranic V, Ivkovic N, Bozic-Nedeljkovic B, Supic G. Genetic variants in the retinoid X receptor gene contribute to osteoarthritis susceptibility. ARCH BIOL SCI 2024; 76:151-160. [DOI: 10.2298/abs240306010s] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Osteoarthritis (OA) is a progressive disease of the joints that causes a gradual loss of function, resulting in limited mobility. Chronic inflammation is the main molecular process that triggers and propagates this disease. The retinoid X receptor (RXR), a member of the nuclear receptor family, is involved in modulating inflammatory pathways by influencing key procatabolic inflammatory cytokines, chemokines, and enzymes responsible for instigating and sustaining chronic joint inflammation. We evaluated the association between OA risk and genetic variants in the RXR? isoform. Compared to control individuals, a statistically significant difference in genotype distribution was detected for the rs7864987 polymorphism (P=0.008), while a positive inclination toward association was noted for rs3118523 (P=0.077). According to our findings based on the additive model, it appears that RXR? rs7864987 is linked to a higher risk of OA (adjusted odds ratio (OR)=1.846, P=0.012), whereas rs3118523 is associated with decreased risk of OA (adjusted OR=0.569, P=0.030). These results suggest that RXR? could be a significant inflammation-related gene involved in the complex network underlying the immunopathology of osteoarthritis. RXR? polymorphisms could potentially drive individualized retinoid therapy for OA based on genetic profile.
Collapse
Affiliation(s)
- Debora Stefik
- Institute for Medical Research, Military Medical Academy, Belgrade, Serbia
| | - Vladimir Vranic
- Clinic for Orthopedic Surgery and Traumatology, Military Medical Academy, Belgrade, Serbia
| | - Nemanja Ivkovic
- Institute for Medical Research, Military Medical Academy, Belgrade, Serbia
| | | | - Gordana Supic
- Institute for Medical Research, Military Medical Academy, Belgrade, Serbia + Medical Faculty of Military Medical Academy, University of Defense, Belgrade, Serbia
| |
Collapse
|
6
|
Wen Y, Su E, Xu L, Menez S, Moledina DG, Obeid W, Palevsky PM, Mansour SG, Devarajan P, Cantley LG, Cahan P, Parikh CR. Analysis of the human kidney transcriptome and plasma proteome identifies markers of proximal tubule maladaptation to injury. Sci Transl Med 2023; 15:eade7287. [PMID: 38091407 PMCID: PMC11405121 DOI: 10.1126/scitranslmed.ade7287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/20/2023] [Indexed: 12/18/2023]
Abstract
Acute kidney injury (AKI) is a major risk factor for long-term adverse outcomes, including chronic kidney disease. In mouse models of AKI, maladaptive repair of the injured proximal tubule (PT) prevents complete tissue recovery. However, evidence for PT maladaptation and its etiological relationship with complications of AKI is lacking in humans. We performed single-nucleus RNA sequencing of 120,985 nuclei in kidneys from 17 participants with AKI and seven healthy controls from the Kidney Precision Medicine Project. Maladaptive PT cells, which exhibited transcriptomic features of dedifferentiation and enrichment in pro-inflammatory and profibrotic pathways, were present in participants with AKI of diverse etiologies. To develop plasma markers of PT maladaptation, we analyzed the plasma proteome in two independent cohorts of patients undergoing cardiac surgery and a cohort of marathon runners, linked it to the transcriptomic signatures associated with maladaptive PT, and identified nine proteins whose genes were specifically up- or down-regulated by maladaptive PT. After cardiac surgery, both cohorts of patients had increased transforming growth factor-β2 (TGFB2), collagen type XXIII-α1 (COL23A1), and X-linked neuroligin 4 (NLGN4X) and had decreased plasminogen (PLG), ectonucleotide pyrophosphatase/phosphodiesterase 6 (ENPP6), and protein C (PROC). Similar changes were observed in marathon runners with exercise-associated kidney injury. Postoperative changes in these markers were associated with AKI progression in adults after cardiac surgery and post-AKI kidney atrophy in mouse models of ischemia-reperfusion injury and toxic injury. Our results demonstrate the feasibility of a multiomics approach to discovering noninvasive markers and associating PT maladaptation with adverse clinical outcomes.
Collapse
Affiliation(s)
- Yumeng Wen
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily Su
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Leyuan Xu
- Section of Nephrology, Department of Medicine, Yale School of Medicine, New Haven, CT 06504, USA
| | - Steven Menez
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dennis G Moledina
- Section of Nephrology, Department of Medicine, Yale School of Medicine, New Haven, CT 06504, USA
| | - Wassim Obeid
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paul M Palevsky
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Kidney Medicine Section, Medical Service, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Sherry G Mansour
- Section of Nephrology, Department of Medicine, Yale School of Medicine, New Haven, CT 06504, USA
| | - Prasad Devarajan
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lloyd G Cantley
- Section of Nephrology, Department of Medicine, Yale School of Medicine, New Haven, CT 06504, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chirag R Parikh
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
7
|
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
|
8
|
Sharma I, Liao Y, Zheng X, Kanwar YS. New Pandemic: Obesity and Associated Nephropathy. Front Med (Lausanne) 2021; 8:673556. [PMID: 34268323 PMCID: PMC8275856 DOI: 10.3389/fmed.2021.673556] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
Incidence of obesity related renal disorders have increased 10-folds in recent years. One of the consequences of obesity is an increased glomerular filtration rate (GFR) that leads to the enlargement of the renal glomerulus, i.e., glomerulomegaly. This heightened hyper-filtration in the setting of type 2 diabetes irreparably damages the kidney and leads to progression of end stage renal disease (ESRD). The patients suffering from type 2 diabetes have progressive proteinuria, and eventually one third of them develop chronic kidney disease (CKD) and ESRD. For ameliorating the progression of CKD, inhibitors of renin angiotensin aldosterone system (RAAS) seemed to be effective, but on a short-term basis only. Long term and stable treatment strategies like weight loss via restricted or hypo-caloric diet or bariatric surgery have yielded better promising results in terms of amelioration of proteinuria and maintenance of normal GFR. Body mass index (BMI) is considered as a traditional marker for the onset of obesity, but apparently, it is not a reliable indicator, and thus there is a need for more precise evaluation of regional fat distribution and amount of muscle mass. With respect to the pathogenesis, recent investigations have suggested perturbation in fatty acid and cholesterol metabolism as the critical mediators in ectopic renal lipid accumulation associated with inflammation, increased generation of ROS, RAAS activation and consequential tubulo-interstitial injury. This review summarizes the renewed approaches for the obesity assessment and evaluation of the pathogenesis of CKD, altered renal hemodynamics and potential therapeutic targets.
Collapse
Affiliation(s)
- Isha Sharma
- Departments of Pathology and Medicine, Northwestern University, Chicago, IL, United States
| | - Yingjun Liao
- Departments of Pathology and Medicine, Northwestern University, Chicago, IL, United States.,Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoping Zheng
- Departments of Pathology and Medicine, Northwestern University, Chicago, IL, United States.,Department of Urology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yashpal S Kanwar
- Departments of Pathology and Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
9
|
Han D, Fang R, Shi R, Jin Y, Wang Q. LncRNA NKILA knockdown promotes cell viability and represses cell apoptosis, autophagy and inflammation in lipopolysaccharide-induced sepsis model by regulating miR-140-5p/CLDN2 axis. Biochem Biophys Res Commun 2021; 559:8-14. [PMID: 33932903 DOI: 10.1016/j.bbrc.2021.04.074] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/17/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play vital roles in human diseases, including sepsis-induced acute kidney injury (AKI). Here, we aimed to investigate the functions of lncRNA NKILA in sepsis-engendered AKI. METHODS HK2 cells stimulated with LPS were used to mimic sepsis-induced AKI in vitro. qRT-PCR was conducted for lncRNA NKILA and miR-140-5p levels. Cell Counting Kit-8 (CCK-8) assay and flow cytometry analysis were employed to analyze cell viability and apoptosis. Western blot assay was utilized to measured protein levels. ELISA kits were used to examine the concentrations of IL-6, IL-1β and TNF-α. Dual-luciferase reporter assay was utilized to analyze the relationships among lncRNA NKILA, miR-140-5p and claudin 2 (CLDN2). RESULTS LPS restrained HK2 cell viability and accelerated cell apoptosis and autophagy. LncRNA NKILA was increased in LPS-treated HK2 cells. LncRNA NKILA silencing reversed the promotional influence of LPS on cell progression in HK2 cells. miR-140-5p inhibition ameliorated lncRNA NKILA knockdown-mediated cell injury in LPS-mediated HK2 cells. CLDN2 was the target of miR-140-5p. MiR-140-5p elevation promoted cell viability and suppressed cell apoptosis, autophagy and inflammation in LPS-induced HK2 cells, with CLDN2 elevation overturned the effects. CONCLUSION LncRNA NKILA silencing protected HK2 cells from LPS-induced impairments by reducing CLDN2 through sponging miR-140-5p.
Collapse
Affiliation(s)
- Dan Han
- Department of Emergency Internal Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai City, China
| | - Rong Fang
- Department of Emergency Internal Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai City, China
| | - Rong Shi
- Department of Emergency Internal Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai City, China
| | - Yuanyuan Jin
- Department of Emergency Internal Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai City, China
| | - Qian Wang
- Department of Emergency Internal Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai City, China.
| |
Collapse
|
10
|
Dai W, Wang M, Wang P, Wen J, Wang J, Cha S, Xiao X, He Y, Shu R, Bai D. lncRNA NEAT1 ameliorates LPS‑induced inflammation in MG63 cells by activating autophagy and suppressing the NLRP3 inflammasome. Int J Mol Med 2021; 47:607-620. [PMID: 33416115 PMCID: PMC7797466 DOI: 10.3892/ijmm.2020.4827] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 11/19/2020] [Indexed: 02/05/2023] Open
Abstract
The mechanisms of inflammation in bone and joint tissue are complex and involve long non‑coding RNAs (lncRNAs), which play an important role in this process. The aim of the present study was to screen out differentially expressed genes in human osteoblasts stimulated by inflammation, and to further explore the mechanisms underlying inflammatory responses and the functional activity of human osteoblasts through bioinformatics methods and in vitro experiments. For this purpose, MG63 cells were stimulated with various concentrations of lipopolysaccharide (LPS) for different periods of time to construct an optimal inflammatory model and RNA sequencing was then performed on these cells. The levels of nuclear enriched abundant transcript 1 (NEAT1), various inflammatory factors, Nod‑like receptor protein 3 (NLRP3) protein and osteogenesis‑related proteins, as well as the levels of cell apoptosis‑ and cell cycle‑related markers were measured in MG63 cells stimulated with LPS, transfected with NEAT1 overexpression plasmid and treated with bexarotene by western blot analysis, RT‑qPCR, immunofluorescence, FISH, TEM and flow cytometry. There were 427 differentially expressed genes in the LPS‑stimulated MG63 cells, in which NEAT1 was significantly downregulated. LPS upregulated the expression of inflammatory cytokines and NLRP3, inhibited the expression of autophagy‑related and osteogenesis‑related proteins, promoted apoptosis and altered the cell cycle, which was partially inhibited by NEAT1 overexpression and promoted by bexarotene. LPS stimulated inflammation in the MG63 cells and inhibited the retinoid X receptor (RXR)‑α to downregulate the expression of NEAT1 and decrease levels of autophagy, which promoted the activation of NLRP3 and the release of inflammatory factors, and impaired the functional activity of osteoblasts, thus promoting the development of inflammation.
Collapse
Affiliation(s)
- Wenyu Dai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, Department of Orthodontics and Pediatrics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041
| | - Manyi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, Department of Orthodontics and Pediatrics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041
- Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510599, P.R. China
| | - Peiqi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, Department of Orthodontics and Pediatrics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041
| | - Ji Wen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, Department of Orthodontics and Pediatrics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041
| | - Jiangyue Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, Department of Orthodontics and Pediatrics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041
| | - Sa Cha
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, Department of Orthodontics and Pediatrics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041
| | - Xueling Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, Department of Orthodontics and Pediatrics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041
| | - Yiruo He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, Department of Orthodontics and Pediatrics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041
| | - Rui Shu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, Department of Orthodontics and Pediatrics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, Department of Orthodontics and Pediatrics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041
| |
Collapse
|
11
|
Chen M, You G, Xie C, Yang R, Hu W, Zheng Z, Liu S, Ye L. Pharmacokinetics of metformin in collagen-induced arthritis rats. Biochem Pharmacol 2021; 185:114413. [PMID: 33434538 DOI: 10.1016/j.bcp.2021.114413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/22/2022]
Abstract
Due to the elevated presence of cytokines, the expressions of metabolic enzymes and drug transporters are altered in rheumatoid arthritis (RA). Given the high incidence of diabetes in patients with RA, the aim of the present study was to investigate the metformin pharmacokinetics of a single oral dose in rats with collagen-induced arthritis (CIA). Blood and urine samples were collected at different timepoints, and analyzed by ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). Tissue samples were also collected to investigate the expression of metabolic enzymes and drug transporters by quantitative reverse transcription-polymerase chain reaction (RT-qPCR) and western blot. The results indicated that the bioavailability of metformin was markedly decreased in the CIA rats. Moreover, metformin was not metabolized by enzymes of rat liver microsomes, suggesting that the decreased bioavailability of metformin was independent of the liver metabolism. In addition, the mRNA, protein expression level and activity of the renal organic cation transporter 2 (OCT2) was markedly increased, suggesting that the enhanced renal clearance of metformin in CIA rats may be due to the up-regulated activity of OCT2. In conclusion, our study suggested that the reduced bioavailability of metformin in CIA rats is possibly related to the up-regulated function of the renal protein OCT2.
Collapse
Affiliation(s)
- Minghao Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Pharmacy, The First Affiliated Hospital of Jinan University (Guangzhou Overseas Chinese Hospital), Guangzhou 510630, China
| | - Guoquan You
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Cong Xie
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ruopeng Yang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wanyu Hu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhijie Zheng
- Clinical Pharmacology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Shuwen Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou 510515, China.
| | - Ling Ye
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
12
|
Zhou T, Lin W, Lin S, Zhong Z, Luo Y, Lin Z, Xie W, Shen W, Hong K. Association of Nuclear Receptor Coactivators with Hypoxia-Inducible Factor-1 α in the Serum of Patients with Chronic Kidney Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1587915. [PMID: 32884936 PMCID: PMC7455818 DOI: 10.1155/2020/1587915] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/29/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023]
Abstract
Nuclear receptor coactivators (NCOAs), consisting of coactivators and corepressors, dramatically enhance the transcriptional activity of nuclear receptors. Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that plays a major role under hypoxic conditions. This study was performed with the focus on the association of NCOAs with HIF-1α in the serum of chronic kidney disease (CKD) patients. Sixty patients with stage 5 CKD and 30 healthy controls from The Second Affiliated Hospital of Shantou University Medical College, between March 21, 2019, and October 30, 2019, were recruited in this prospective cohort study. We analyzed the serum levels of NCOAs (NCOA1, NCOA2, and NCOA3), HIF-1α, vascular endothelial growth factor (VEGF), etc. and assessed whether there was any relationship between these parameters and CKD disease. We found that circulating NCOA1 was positively associated with circulating NCOA2, NCOA3, and HIF-1α. A positive correlation was also observed between NCOA2 and NCOA1, NCOA3, HIF-1α, and VEGF. Furthermore, statistically significant correlations between NCOA3 and NCOA1, NCOA2, and HIF-1α were observed. The serum levels of VEGF in the CKD group were higher than those of the healthy control group. Circulating NCOA1 and circulating NCOA2 were negatively associated with procalcitonin. In conclusion, there was an association between circulating NCOA1, NCOA2, NCOA3, and circulating HIF-1α, and circulating VEGF was a risk factor for CKD disease. However, more studies should be performed to confirm this hypothesis.
Collapse
Affiliation(s)
- Tianbiao Zhou
- Department of Nephrology, The Second Affiliated Hospital, Shantou University Medical College, 515041 Shantou, China
| | - Wenshan Lin
- Department of Nephrology, The Second Affiliated Hospital, Shantou University Medical College, 515041 Shantou, China
| | - Shujun Lin
- Department of Nephrology, The Second Affiliated Hospital, Shantou University Medical College, 515041 Shantou, China
| | - Zhiqing Zhong
- Department of Nephrology, The Second Affiliated Hospital, Shantou University Medical College, 515041 Shantou, China
| | - Yuanyuan Luo
- Department of Nephrology, The Second Affiliated Hospital, Shantou University Medical College, 515041 Shantou, China
| | - Zhijun Lin
- Department of Nephrology, The Second Affiliated Hospital, Shantou University Medical College, 515041 Shantou, China
| | - Weiji Xie
- Department of Nephrology, The Second Affiliated Hospital, Shantou University Medical College, 515041 Shantou, China
| | - Weitao Shen
- Department of Clinical Laboratory, The Second Affiliated Hospital, Shantou University Medical College, 515041 Shantou, China
| | - Kai Hong
- Department of Clinical Laboratory, The Second Affiliated Hospital, Shantou University Medical College, 515041 Shantou, China
| |
Collapse
|
13
|
Gao C, Zou X, Chen H, Shang R, Wang B. Long Non-Coding RNA Nuclear Paraspeckle Assembly Transcript 1 (NEAT1)Relieves Sepsis-Induced Kidney Injury and Lipopolysaccharide (LPS)-Induced Inflammation in HK-2 Cells. Med Sci Monit 2020; 26:e921906. [PMID: 32724027 PMCID: PMC7414528 DOI: 10.12659/msm.921906] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Long non-coding RNAs (lncRNAs) play key roles in the development and progression of diseases, including sepsis. Therefore, this study aimed to clarify the role and underlying molecular mechanisms of lncRNA NEAT1 in sepsis. Material/Methods We used real-time quantitative polymerase chain reaction (RT-qPCR) to analyze the expression of lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1), let-7b-5p, and tumor necrosis factor receptor-associated factor 6 (TRAF6). Western blot assay was used to measure the protein expression levels. After treatment with lipopolysaccharide (LPS), the biological behaviors of human renal tubular epithelial cells (HK-2), such as proliferation and apoptosis, were determined using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazol-3-ium bromide (MTT) and flow cytometry assays, respectively. The interaction relationship among NEAT1, TRAF6, and let-7b-5p was analyzed by the bioinformatics starBase database and dual-luciferase reporter assay. Results lncRNA NEAT1 was expressed at higher levels in kidney tissues from sepsis patients than in healthy kidney tissues. Interestingly, LPS induced high expression of lncRNA NEAT1 in HK-2 cells in a time- and dose-dependent manner. Furthermore, silencing of NEAT1 weakened LPS-induced apoptosis, inflammation, and inhibition of proliferation, which was overturned by silencing of let-7b-5p. In addition, overexpression of TRAF6 abolished the overexpression of let-7b-5p-induced effects on apoptosis, inflammation, and growth of HK-2 cells exposed to LPS. In summary, NEAT1 regulated TRAF6 expression by sponging let-7b-5p in HK-2 cells, which promotes LPS-induced injury and inflammation in HK-2 cells. Conclusions Our data show that the lower expression of NEAT1 impeded sepsis development and LPS-induced injury inflammation by targeting let-7b-5p/TRAF6 axis, and NEAT1 may be a target for treatment of sepsis patients.
Collapse
Affiliation(s)
- Cheng Gao
- Department of Critical Care Medicine, Jingzhou Central Hospital (The Second Clinical Medical College, Yangtze University), Jingzhou, Hubei, China (mainland)
| | - Xiequn Zou
- Department of Critical Care Medicine, Jingzhou Central Hospital (The Second Clinical Medical College, Yangtze University), Jingzhou, Hubei, China (mainland)
| | - Huan Chen
- Department of Obstetrics, Jingzhou Central Hospital (The Second Clinical Medical College, Yangtze University), Jingzhou, Hubei, China (mainland)
| | - Rong Shang
- Department of Orthopaedics, Jingzhou Central Hospital (The Second Clinical Medical College, Yangtze University), Jingzhou, Hubei, China (mainland)
| | - Bing Wang
- Department of Critical Care Medicine, Jingzhou Central Hospital (The Second Clinical Medical College, Yangtze University), Jingzhou, Hubei, China (mainland)
| |
Collapse
|
14
|
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: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
15
|
Key role of organic cation transporter 2 for the nephrotoxicity effect of triptolide in rheumatoid arthritis. Int Immunopharmacol 2019; 77:105959. [PMID: 31644961 DOI: 10.1016/j.intimp.2019.105959] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/23/2019] [Accepted: 10/01/2019] [Indexed: 12/21/2022]
Abstract
Tripterygium wilfordii Hook. F. (TwHF), a traditional Chinese Medicine, is effective in treating rheumatoid arthritis (RA), but its severe nephrotoxicity limits its extensive application. The nephrotoxic mechanism of Triptolide (TP), the main pharmacological and toxic component of TwHF, has not been fully revealed. This study was designed to explore the nephrotoxicity of TP in the RA state and the potential molecular mechanism. A rat collagen-induced arthritis (CIA) model was constructed and administered with TP for 28 days in vivo. Results showed that the kidney injury induced by TP was aggravated in the CIA state, the concentration of TP in the renal cortex was higher than that of the medulla after TP administration in the CIA rats, and the expression of organic cation transporter 2 (Oct2) in kidney was up-regulated under CIA condition. Besides, rat kidney slice study demonstrated that TP was transported by Oct2 and this was confirmed by transient silencing and overexpression of OCT2 in HEK-293T cells. Furthermore, cytoinflammatory models on HK-2 and HEK-293T cell lines were constructed by exposure of TNF-α or IL-1β to further explore the TP's renal toxicity. Results suggested that TNF-α exposure aggravated TP's toxicity and up-regulated the protein expression of OCT2 in both cell lines. TNF-α treatment also increased the function of OCT2 and finally OCT2 silencing confirmed OCT2 mediated nephrotoxicity of TP in HEK-293T cells. In summary, the exposure of TNF-α in RA state induced the expression of OCT2, which transported more TP into kidney cortex, subsequently exacerbated the kidney injury.
Collapse
|
16
|
Duong N, Curley K, Brown A, Campanelli A, Do MA, Levy D, Tantry A, Marriott G, Lu B. Decoy exosomes as a novel biologic reagent to antagonize inflammation. Int J Nanomedicine 2019; 14:3413-3425. [PMID: 31190800 PMCID: PMC6514129 DOI: 10.2147/ijn.s196975] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/09/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Exosomes are ubiquitous naturally secreted stable nanovesicles that can be engineered to target and deliver novel therapeutics to treat a host of human diseases. Methods: We engineered the surfaces of cell-derived nanovesicles to act as decoys in the treatment of inflammation by antagonizing the major proinflammatory cytokine, tumor necrosis factor alpha (TNFα). Results: Decoy exosomes were generated by displaying the TNFα binding domain of human TNF receptor-1 (hTNFR1) on the outer surface of exosomes using stably transfected HEK293 cells. We developed an efficient method to purify the engineered exosomes from conditioned medium based on sequential centrifugation, ultrafiltration, and precipitation. We characterized decoy exosomes using immune-quantification, nanoparticle tracking analysis, and confocal microscopy to confirm that they retain the correct orientation, size, and shape of naturally produced exosomes. We demonstrated the engineered decoy exosomes specifically antagonize activities of TNFα using an inflammatory reporter cell line. Conclusions: Decoy exosomes produced in human cells serve as a novel biologic reagent for antagonizing inflammatory signaling mediated by TNFα.
Collapse
Affiliation(s)
- Natalie Duong
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA 95053, USA
| | - Kevin Curley
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA 95053, USA
| | - Annie Brown
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA 95053, USA
| | - Alexander Campanelli
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA 95053, USA
| | - Mai Anh Do
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA 95053, USA
| | - Daniel Levy
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA 95053, USA
| | - Adarsh Tantry
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA 95053, USA
| | - Gerard Marriott
- Department of Bioengineering and Tsinghua-Berkeley Shenzhen Institute, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Biao Lu
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA 95053, USA
| |
Collapse
|
17
|
Mitochondrial-Targeted Antioxidant MitoQ Prevents E. coli Lipopolysaccharide-Induced Accumulation of Triacylglycerol and Lipid Droplets Biogenesis in Epithelial Cells. J Lipids 2018; 2018:5745790. [PMID: 30245885 PMCID: PMC6139225 DOI: 10.1155/2018/5745790] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/02/2018] [Accepted: 07/25/2018] [Indexed: 01/26/2023] Open
Abstract
The effect of bacterial lipopolysaccharide (LPS) on eukaryotic cell could be accompanied by a significant metabolic shift that includes accumulation of triacylglycerol (TAG) in lipid droplets (LD), ubiquitous organelles associated with fatty acid storage, energy regulation and demonstrated tight spatial and functional connections with mitochondria. The impairment of mitochondrial activity under pathological stimuli has been shown to provoke TAG storage and LD biogenesis. However the potential mechanisms that link mitochondrial disturbances and TAG accumulation are not completely understood. We hypothesize that mitochondrial ROS (mROS) may play a role of a trigger leading to subsequent accumulation of intracellular TAG and LD in response to a bacterial stimulus. Using isolated epithelial cells from the frog urinary bladder, we showed that LPS decreased fatty acids oxidation, enhanced TAG deposition, and promoted LD formation. LPS treatment did not affect the mitochondrial membrane potential but increased cellular ROS production and led to impairment of mitochondrial function as revealed by decreased ATP production and a reduced maximal oxygen consumption rate (OCR) and OCR directed at ATP turnover. The mitochondrial-targeted antioxidant MitoQ at a dose of 25 nM did not prevent LPS-induced alterations in cellular respiration, but, in contrast to nonmitochondrial antioxidant α-tocopherol, reduced the effect of LPS on the generation of ROS, restored the LPS-induced decline of fatty acids oxidation, and prevented accumulation of TAG and LD biogenesis. The data obtained indicate the key signaling role of mROS in the lipid metabolic shift that occurs under the impact of a bacterial pathogen in epithelial cells.
Collapse
|
18
|
Chan JD, Day TA, Marchant JS. Coalescing beneficial host and deleterious antiparasitic actions as an antischistosomal strategy. eLife 2018; 7:35755. [PMID: 30059006 PMCID: PMC6095690 DOI: 10.7554/elife.35755] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/24/2018] [Indexed: 12/28/2022] Open
Abstract
Conventional approaches for antiparasitic drug discovery center upon discovering selective agents that adversely impact parasites with minimal host side effects. Here, we show that agents with a broad polypharmacology, often considered ‘dirtier’ drugs, can have unique efficacy if they combine deleterious effects on the parasite with beneficial actions in the host. This principle is evidenced through a screen for drugs to treat schistosomiasis, a parasitic flatworm disease that impacts over 230 million people. A target-based screen of a Schistosoma serotoninergic G protein coupled receptor yielded the potent agonist, ergotamine, which disrupted worm movement. In vivo, ergotamine decreased mortality, parasite load and intestinal egg counts but also uniquely reduced organ pathology through engagement of host GPCRs that repressed hepatic stellate cell activation, inflammatory damage and fibrosis. The unique ability of ergotamine to engage both host and parasite GPCRs evidences a future strategy for anthelmintic drug design that coalesces deleterious antiparasitic activity with beneficial host effects. More than 200 million people worldwide are infected with parasitic worms that cause the disease schistosomiasis. Most cases occur in sub-Saharan Africa. Long-term infections can damage organs, and children who are affected may suffer delayed growth and learning difficulties. Despite its significant health and economic impact, schistosomiasis is still considered a ‘neglected’ tropical disease. This means there has not been adequate investment into developing new treatments or cures. A drug called praziquantel is currently the only treatment for schistosomiasis. However, the drug has unpleasant side effects, cannot cure all infected individuals, and there is a concern that worms may develop resistance to its effects. This means there is an urgent need to develop new therapies. One possible approach would be to develop drugs that interfere with the worm’s ability to move. Chan et al. screened thousands of existing chemicals for interactions with a protein that is known to control how the worms move. A drug called ergotamine, which is currently used to treat migraines, strongly interacted with the protein. Treating infected mice with ergotamine eliminated the parasites and reduced the organ damage caused by the infection. Praziquantel also reduced the number of parasites in the mice but it did not prevent organ damage. The results presented by Chan et al. show that a single drug can interact with targets in both the worm and the animals it infects. Searching for drugs that have this dual effect may help to develop more effective treatments for schistosomiasis and other diseases caused by parasites. Ergotamine itself is unlikely to be used to treat people for schistosomiasis because of the side effects produced when using it repeatedly. However, these findings will help researchers identify and develop safer drugs with similar benefits.
Collapse
Affiliation(s)
- John D Chan
- Department of Biomedical Sciences, Iowa State University, Ames, United States
| | - Timothy A Day
- Department of Biomedical Sciences, Iowa State University, Ames, United States
| | - Jonathan S Marchant
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, United States
| |
Collapse
|
19
|
Chen L, Yao Q, Xu S, Wang H, Qu P. Inhibition of the NLRP3 inflammasome attenuates foam cell formation of THP-1 macrophages by suppressing ox-LDL uptake and promoting cholesterol efflux. Biochem Biophys Res Commun 2018; 495:382-387. [DOI: 10.1016/j.bbrc.2017.11.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 12/12/2022]
|
20
|
Selvaraj S, Oh JH, Spanel R, Länger F, Han HY, Lee EH, Yoon S, Borlak J. The pathogenesis of diclofenac induced immunoallergic hepatitis in a canine model of liver injury. Oncotarget 2017; 8:107763-107824. [PMID: 29296203 PMCID: PMC5746105 DOI: 10.18632/oncotarget.21201] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/31/2017] [Indexed: 12/19/2022] Open
Abstract
Hypersensitivity to non-steroidal anti-inflammatory drugs is a common adverse drug reaction and may result in serious inflammatory reactions of the liver. To investigate mechanism of immunoallergic hepatitis beagle dogs were given 1 or 3 mg/kg/day (HD) oral diclofenac for 28 days. HD diclofenac treatment caused liver function test abnormalities, reduced haematocrit and haemoglobin but induced reticulocyte, WBC, platelet, neutrophil and eosinophil counts. Histopathology evidenced hepatic steatosis and glycogen depletion, apoptosis, acute lobular hepatitis, granulomas and mastocytosis. Whole genome scans revealed 663 significantly regulated genes of which 82, 47 and 25 code for stress, immune response and inflammation. Immunopathology confirmed strong induction of IgM, the complement factors C3&B, SAA, SERPING1 and others of the classical and alternate pathway. Alike, marked expression of CD205 and CD74 in Kupffer cells and lymphocytes facilitate antigen presentation and B-cell differentiation. The highly induced HIF1A and KLF6 protein expression in mast cells and macrophages sustain inflammation. Furthermore, immunogenomics discovered 24, 17, 6 and 11 significantly regulated marker genes to hallmark M1/M2 polarized macrophages, lymphocytic and granulocytic infiltrates; note, the latter was confirmed by CAE staining. Other highly regulated genes included alpha-2-macroglobulin, CRP, hepcidin, IL1R1, S100A8 and CCL20. Diclofenac treatment caused unprecedented induction of myeloperoxidase in macrophages and oxidative stress as shown by SOD1/SOD2 immunohistochemistry. Lastly, bioinformatics defined molecular circuits of inflammation and consisted of 161 regulated genes. Altogether, the mechanism of diclofenac induced liver hypersensitivity reactions involved oxidative stress, macrophage polarization, mastocytosis, complement activation and an erroneous programming of the innate and adaptive immune system.
Collapse
Affiliation(s)
- Saravanakumar Selvaraj
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625 Hannover, Germany
| | - Jung-Hwa Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, 34114 Gajeong-ro, Yuseong, Daejeon, Republic of Korea
| | - Reinhard Spanel
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625 Hannover, Germany.,Institute of Pathology, 41747 Viersen, Germany
| | - Florian Länger
- Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany
| | - Hyoung-Yun Han
- Department of Predictive Toxicology, Korea Institute of Toxicology, 34114 Gajeong-ro, Yuseong, Daejeon, Republic of Korea
| | - Eun-Hee Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, 34114 Gajeong-ro, Yuseong, Daejeon, Republic of Korea
| | - Seokjoo Yoon
- Department of Predictive Toxicology, Korea Institute of Toxicology, 34114 Gajeong-ro, Yuseong, Daejeon, Republic of Korea
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
21
|
Abstract
Acute kidney injury (AKI) arising from diverse etiologies is characterized by mitochondrial dysfunction. The peroxisome proliferator-activated receptor γ coactivator-1alpha (PGC1α), a master regulator of mitochondrial biogenesis, has been shown to be protective in AKI. Interestingly, reduction of PGC1α has also been implicated in the development of diabetic kidney disease and renal fibrosis. The beneficial renal effects of PGC1α make it a prime target for therapeutics aimed at ameliorating AKI, forms of chronic kidney disease (CKD), and their intersection. This review summarizes the current literature on the relationship between renal health and PGC1α and proposes areas of future interest.
Collapse
Affiliation(s)
- Matthew R Lynch
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts
| | - Mei T Tran
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts
| | - Samir M Parikh
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
22
|
Matsushita N, Hassanein MT, Martinez-Clemente M, Lazaro R, French SW, Xie W, Lai K, Karin M, Tsukamoto H. Gender difference in NASH susceptibility: Roles of hepatocyte Ikkβ and Sult1e1. PLoS One 2017; 12:e0181052. [PMID: 28797077 PMCID: PMC5552280 DOI: 10.1371/journal.pone.0181052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 06/26/2017] [Indexed: 12/22/2022] Open
Abstract
Myeloid cell and hepatocyte IKKβ may mediate the genesis of obesity and insulin resistance in mice fed high fat diet. However, their gender-specific roles in the pathogenesis of non-alcoholic steatohepatitis (NASH) are not known. Here we demonstrate myeloid IKKβ deficiency prevents Western diet-induced obesity and visceral adiposity in females but not in males, and attenuates hyperglycemia, global IR, and NASH in both genders. In contrast, all metabolic sequela including NASH are aggravated by hepatocyte IKKβ deficiency (IkbkbΔhep) in male but not female mice. Gene profiling identifies sulfotransferase family 1E (Sult1e1), which encodes a sulfotransferase E1 responsible for inactivation of estrogen, as a gene upregulated in NASH in both genders and most conspicuously in male IkbkbΔhep mice having worst NASH and lowest plasma estradiol levels. LXRα is enriched to LXRE on Sult1e1 promoter in male WT and IkbkbΔhep mice with NASH, and a Sult1e1 promoter activity is increased by LXRα and its ligand and augmented by expression of a S32A mutant of IκBα. These results demonstrate striking gender differences in regulation by IKKβ of high cholesterol saturated fat diet-induced metabolic changes including NASH and suggest hepatocyte IKKβ is protective in male due at least in part to its ability to repress LXR-induced Sult1e1. Our findings also raise a caution for systemic IKK inhibition for the treatment of NASH as it may exacerbate the disease in male patients.
Collapse
Affiliation(s)
- Noriko Matsushita
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Mohamed T. Hassanein
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Marcos Martinez-Clemente
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Raul Lazaro
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Samuel W. French
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Wen Xie
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, United States of America
| | - Keane Lai
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Michael Karin
- Departments of Pharmacology and Pathology, University of California San Diego, La Jolla, California, United States of America
| | - Hidekazu Tsukamoto
- Southern California Research Center for ALPD and Cirrhosis of the University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
- Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
23
|
Du F, Gesang Q, Cao J, Qian M, Ma L, Wu D, Yu H. Isoliquiritigenin Attenuates Atherogenesis in Apolipoprotein E-Deficient Mice. Int J Mol Sci 2016; 17:ijms17111932. [PMID: 27869741 PMCID: PMC5133927 DOI: 10.3390/ijms17111932] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/09/2016] [Accepted: 11/11/2016] [Indexed: 12/22/2022] Open
Abstract
Isoliquiritigenin (ISL) exhibits antioxidation and anti-inflammation activity. We sought to investigate the effects and mechanism of ISL on the development of atherosclerotic lesions in apolipoprotein E-deficient (apoE−/−) mice. Firstly, we determined that ISL reduced the mRNA levels of inflammatory factors interleukin 6 (IL-6), tumor necrosis factor α (TNF-α), and monocyte chemotactic protein-1 (MCP-1), while it increased the expression of several lipoprotein-related genes in peritoneal macrophages treated with lipopolysaccharide (LPS). ISL also enhanced peroxisome proliferator-activated receptor gamma (PPARγ) protein levels and reversed the changes of ATP-binding cassette transporter A (ABCA1) and cluster of differentiation 36 (CD36) in macrophages treated with oxidative low-density lipoprotein (ox-LDL). Then, in an in vivo study, female apoE−/− mice were fed a Western diet with ISL (0, 20, 100 mg/kg/day) added for 12 weeks. We found that ISL decreased the plasma cholesterol levels of very low-density lipoprotein (VLDL)/LDL, promoted plasma superoxide dismutase (SOD) and paraoxonase-1 (PON1) activities, and decreased plasma IL-6, TNF-α, and MCP-1 levels. Moreover, ISL significantly reduced the atherosclerotic lesions and hepatic steatosis in apoE−/− mice. In the liver, ISL altered the expression of several key genes (such as SRBI, ABCA1, ABCG8, PPARγ, and FASN) involving cholesterol-selective uptake and excretion into bile, triglyceride (TG) biosynthesis, and inflammation. These results suggest that the atheroprotective effects of ISL are due to the improvement of lipid metabolism, antioxidation, and anti-inflammation, which involve PPARγ-dependent signaling.
Collapse
Affiliation(s)
- Fen Du
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, 185 Donghu Road, Bldg. 2, 2-209, Wuhan 430071, China.
| | - Quzhen Gesang
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, 185 Donghu Road, Bldg. 2, 2-209, Wuhan 430071, China.
- Medical College, Tibet University, Lhasa 850000, China.
| | - Jia Cao
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, 185 Donghu Road, Bldg. 2, 2-209, Wuhan 430071, China.
| | - Mei Qian
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, 185 Donghu Road, Bldg. 2, 2-209, Wuhan 430071, China.
| | - Li Ma
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, 185 Donghu Road, Bldg. 2, 2-209, Wuhan 430071, China.
| | - Dongfang Wu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Hong Yu
- Department of Biochemistry and Molecular Biology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University School of Basic Medical Sciences, 185 Donghu Road, Bldg. 2, 2-209, Wuhan 430071, China.
| |
Collapse
|
24
|
Zhou J, Lam B, Neogi SG, Yeo GSH, Azizan EAB, Brown MJ. Transcriptome Pathway Analysis of Pathological and Physiological Aldosterone-Producing Human Tissues. Hypertension 2016; 68:1424-1431. [PMID: 27777363 PMCID: PMC5100803 DOI: 10.1161/hypertensionaha.116.08033] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 07/08/2016] [Accepted: 09/19/2016] [Indexed: 01/10/2023]
Abstract
Supplemental Digital Content is available in the text. Primary aldosteronism is present in ≈10% of hypertensives. We previously performed a microarray assay on aldosterone-producing adenomas and their paired zona glomerulosa and fasciculata. Confirmation of top genes validated the study design and functional experiments of zona glomerulosa selective genes established the role of the encoded proteins in aldosterone regulation. In this study, we further analyzed our microarray data using AmiGO 2 for gene ontology enrichment and Ingenuity Pathway Analysis to identify potential biological processes and canonical pathways involved in pathological and physiological aldosterone regulation. Genes differentially regulated in aldosterone-producing adenoma and zona glomerulosa were associated with steroid metabolic processes gene ontology terms. Terms related to the Wnt signaling pathway were enriched in zona glomerulosa only. Ingenuity Pathway Analysis showed "NRF2-mediated oxidative stress response pathway" and "LPS (lipopolysaccharide)/IL-1 (interleukin-1)–mediated inhibition of RXR (retinoid X receptor) function" were affected in both aldosterone-producing adenoma and zona glomerulosa with associated genes having up to 21- and 8-fold differences, respectively. Comparing KCNJ5-mutant aldosterone-producing adenoma, zona glomerulosa, and zona fasciculata samples with wild-type samples, 138, 56, and 59 genes were differentially expressed, respectively (fold-change >2; P<0.05). ACSS3, encoding the enzyme that synthesizes acetyl-CoA, was the top gene upregulated in KCNJ5-mutant aldosterone-producing adenoma compared with wild-type. NEFM, a gene highly upregulated in zona glomerulosa, was upregulated in KCNJ5 wild-type aldosterone-producing adenomas. NR4A2, the transcription factor for aldosterone synthase, was highly expressed in zona fasciculata adjacent to a KCNJ5-mutant aldosterone-producing adenoma. Further interrogation of these genes and pathways could potentially provide further insights into the pathology of primary aldosteronism.
Collapse
Affiliation(s)
- Junhua Zhou
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, United Kingdom (J.Z., M.J.B.); Clinical Pharmacology Unit, Department of Medicine, University of Cambridge (J.Z.), University of Cambridge Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science (B.L., G.S.H.Y.), Cambridge University Hospitals NHS Foundation Trust (S.G.N.), Addenbrooke's Hospital, United Kingdom; and Department of Medicine, Faculty of Medicine, The National University of Malaysia (UKM) Medical Centre, Kuala Lumpur (E.A.B.A.)
| | - Brian Lam
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, United Kingdom (J.Z., M.J.B.); Clinical Pharmacology Unit, Department of Medicine, University of Cambridge (J.Z.), University of Cambridge Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science (B.L., G.S.H.Y.), Cambridge University Hospitals NHS Foundation Trust (S.G.N.), Addenbrooke's Hospital, United Kingdom; and Department of Medicine, Faculty of Medicine, The National University of Malaysia (UKM) Medical Centre, Kuala Lumpur (E.A.B.A.)
| | - Sudeshna G Neogi
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, United Kingdom (J.Z., M.J.B.); Clinical Pharmacology Unit, Department of Medicine, University of Cambridge (J.Z.), University of Cambridge Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science (B.L., G.S.H.Y.), Cambridge University Hospitals NHS Foundation Trust (S.G.N.), Addenbrooke's Hospital, United Kingdom; and Department of Medicine, Faculty of Medicine, The National University of Malaysia (UKM) Medical Centre, Kuala Lumpur (E.A.B.A.)
| | - Giles S H Yeo
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, United Kingdom (J.Z., M.J.B.); Clinical Pharmacology Unit, Department of Medicine, University of Cambridge (J.Z.), University of Cambridge Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science (B.L., G.S.H.Y.), Cambridge University Hospitals NHS Foundation Trust (S.G.N.), Addenbrooke's Hospital, United Kingdom; and Department of Medicine, Faculty of Medicine, The National University of Malaysia (UKM) Medical Centre, Kuala Lumpur (E.A.B.A.)
| | - Elena A B Azizan
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, United Kingdom (J.Z., M.J.B.); Clinical Pharmacology Unit, Department of Medicine, University of Cambridge (J.Z.), University of Cambridge Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science (B.L., G.S.H.Y.), Cambridge University Hospitals NHS Foundation Trust (S.G.N.), Addenbrooke's Hospital, United Kingdom; and Department of Medicine, Faculty of Medicine, The National University of Malaysia (UKM) Medical Centre, Kuala Lumpur (E.A.B.A.).
| | - Morris J Brown
- From the Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, United Kingdom (J.Z., M.J.B.); Clinical Pharmacology Unit, Department of Medicine, University of Cambridge (J.Z.), University of Cambridge Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science (B.L., G.S.H.Y.), Cambridge University Hospitals NHS Foundation Trust (S.G.N.), Addenbrooke's Hospital, United Kingdom; and Department of Medicine, Faculty of Medicine, The National University of Malaysia (UKM) Medical Centre, Kuala Lumpur (E.A.B.A.)
| |
Collapse
|
25
|
Obesity-related glomerulopathy: clinical and pathologic characteristics and pathogenesis. Nat Rev Nephrol 2016; 12:453-71. [PMID: 27263398 DOI: 10.1038/nrneph.2016.75] [Citation(s) in RCA: 475] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prevalence of obesity-related glomerulopathy is increasing in parallel with the worldwide obesity epidemic. Glomerular hypertrophy and adaptive focal segmental glomerulosclerosis define the condition pathologically. The glomerulus enlarges in response to obesity-induced increases in glomerular filtration rate, renal plasma flow, filtration fraction and tubular sodium reabsorption. Normal insulin/phosphatidylinositol 3-kinase/Akt and mTOR signalling are critical for podocyte hypertrophy and adaptation. Adipokines and ectopic lipid accumulation in the kidney promote insulin resistance of podocytes and maladaptive responses to cope with the mechanical forces of renal hyperfiltration. Although most patients have stable or slowly progressive proteinuria, up to one-third develop progressive renal failure and end-stage renal disease. Renin-angiotensin-aldosterone blockade is effective in the short-term but weight loss by hypocaloric diet or bariatric surgery has induced more consistent and dramatic antiproteinuric effects and reversal of hyperfiltration. Altered fatty acid and cholesterol metabolism are increasingly recognized as key mediators of renal lipid accumulation, inflammation, oxidative stress and fibrosis. Newer therapies directed to lipid metabolism, including SREBP antagonists, PPARα agonists, FXR and TGR5 agonists, and LXR agonists, hold therapeutic promise.
Collapse
|
26
|
Lin J, Cheng Y, Wang T, Tang L, Sun Y, Lu X, Yu H. Soyasaponin Ab inhibits lipopolysaccharide-induced acute lung injury in mice. Int Immunopharmacol 2016; 30:121-128. [PMID: 26672918 DOI: 10.1016/j.intimp.2015.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 11/04/2015] [Accepted: 12/01/2015] [Indexed: 11/23/2022]
Abstract
Soyasaponin Ab (SA) has been reported to have anti-inflammatory effect. However, the effects of SA on lipopolysaccharide (LPS)-induced acute lung injury (ALI) have not been reported. The aim of this study was to investigate the anti-inflammatory effects of SA on LPS-induced ALI and clarify the possible mechanism. The mice were stimulated with LPS to induce ALI. SA was given 1h after LPS treatment. 12h later, lung tissues were collected to assess pathological changes and edema. Bronchoalveolar lavage fluid (BALF) was collected to assess inflammatory cytokines and nitric oxide (NO) production. In vitro, mice alveolar macrophages were used to investigate the anti-inflammatory mechanism of SA. Our results showed that SA attenuated LPS-induced lung pathological changes, edema, the expression of cycloxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) in lung tissues, as well as TNF-α, IL-6, IL-1β, and NO production in mice. Meanwhile, SA up-regulated the activities of superoxide dismutase (SOD) and catalase decreased by LPS in mice. SA also inhibited LPS-induced TNF-α, IL-6 and IL-1β production as well as NF-κB activation in alveolar macrophages. Furthermore, SA could activate Liver X Receptor Alpha (LXRα) and knockdown of LXRα by RNAi abrogated the anti-inflammatory effects of SA. In conclusion, the current study demonstrated that SA exhibited protective effects against LPS-induced acute lung injury and the possible mechanism was involved in activating LXRα, thereby inhibiting LPS-induced inflammatory response.
Collapse
Affiliation(s)
- Jing Lin
- Chinese Medicine Department of The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Yanwen Cheng
- Pharmaceutical Department of The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Tao Wang
- Basic Medical College of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China
| | - Lihua Tang
- Medical Record Quality Department of The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Yan Sun
- Chinese Medicine Department of The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Xiuyun Lu
- Chinese Medicine Department of The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Huimin Yu
- Chinese Medicine Department of The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China.
| |
Collapse
|
27
|
Abstract
Per milligram of tissue, only the heart exceeds the kidney's abundance of mitochondria. Not surprisingly, renal mitochondria are most densely concentrated in the epithelium of the nephron, at sites where the chemical work of moving solutes against electrochemical gradients places large and constant demands for adenosine triphosphate. Derangements of renal epithelial mitochondria appear to be a hallmark for diverse forms of acute kidney injury (AKI). The pathogenesis of multiple-organ dysfunction syndrome in sepsis is complex, but a substantial body of experimental and observational human data supports the twin concepts that mitochondrial dysfunction contributes to impaired filtration and that recovery of mitochondrial structure and function is essential for recovery from sepsis-associated AKI. These insights have suggested novel methods to diagnose, stratify, prevent, or even treat this common and deadly complication of critical illness. This review will do the following: (1) describe the structure and functions of healthy mitochondria and how renal energy metabolism relates to solute transport; (2) provide an overview of the evidence linking mitochondrial pathology to renal disease; (3) summarize the mitochondrial lesions observed in septic AKI; (4) analyze the role of mitochondrial processes including fission/fusion, mitophagy, and biogenesis in the development of septic AKI and recovery from this disease; and (5) explore the potential for therapeutically targeting mitochondria to prevent or treat septic AKI.
Collapse
|
28
|
Ding J, Reynolds LM, Zeller T, Müller C, Lohman K, Nicklas BJ, Kritchevsky SB, Huang Z, de la Fuente A, Soranzo N, Settlage RE, Chuang CC, Howard T, Xu N, Goodarzi MO, Chen YDI, Rotter JI, Siscovick DS, Parks JS, Murphy S, Jacobs DR, Post W, Tracy RP, Wild PS, Blankenberg S, Hoeschele I, Herrington D, McCall CE, Liu Y. Alterations of a Cellular Cholesterol Metabolism Network Are a Molecular Feature of Obesity-Related Type 2 Diabetes and Cardiovascular Disease. Diabetes 2015; 64:3464-74. [PMID: 26153245 PMCID: PMC4587646 DOI: 10.2337/db14-1314] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 05/28/2015] [Indexed: 12/20/2022]
Abstract
Obesity is linked to type 2 diabetes (T2D) and cardiovascular diseases; however, the underlying molecular mechanisms remain unclear. We aimed to identify obesity-associated molecular features that may contribute to obesity-related diseases. Using circulating monocytes from 1,264 Multi-Ethnic Study of Atherosclerosis (MESA) participants, we quantified the transcriptome and epigenome. We discovered that alterations in a network of coexpressed cholesterol metabolism genes are a signature feature of obesity and inflammatory stress. This network included 11 BMI-associated genes related to sterol uptake (↑LDLR, ↓MYLIP), synthesis (↑SCD, FADS1, HMGCS1, FDFT1, SQLE, CYP51A1, SC4MOL), and efflux (↓ABCA1, ABCG1), producing a molecular profile expected to increase intracellular cholesterol. Importantly, these alterations were associated with T2D and coronary artery calcium (CAC), independent from cardiometabolic factors, including serum lipid profiles. This network mediated the associations between obesity and T2D/CAC. Several genes in the network harbored C-phosphorus-G dinucleotides (e.g., ABCG1/cg06500161), which overlapped Encyclopedia of DNA Elements (ENCODE)-annotated regulatory regions and had methylation profiles that mediated the associations between BMI/inflammation and expression of their cognate genes. Taken together with several lines of previous experimental evidence, these data suggest that alterations of the cholesterol metabolism gene network represent a molecular link between obesity/inflammation and T2D/CAC.
Collapse
Affiliation(s)
- Jingzhong Ding
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Lindsay M Reynolds
- Department of Epidemiology & Prevention, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Tanja Zeller
- University Heart Center Hamburg, Clinic for General and Interventional Cardiology, and DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Christian Müller
- University Heart Center Hamburg, Clinic for General and Interventional Cardiology, and DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Kurt Lohman
- Department of Epidemiology & Prevention, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Barbara J Nicklas
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | | | | | - Alberto de la Fuente
- Department of Biomathematics and Bioinformatics, Institute of Genetics and Biometry, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | | | | | - Chia-Chi Chuang
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Timothy Howard
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Ning Xu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Y-D Ida Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA
| | | | - John S Parks
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | | | - David R Jacobs
- School of Public Health, University of Minnesota, Minneapolis, MN
| | | | | | - Philipp S Wild
- Center for Thrombosis and Hemostasis and Department of Medicine 2, University Medical Center of the Johannes Gutenberg University Mainz, and DZHK (German Centre for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany
| | - Stefan Blankenberg
- University Heart Center Hamburg, Clinic for General and Interventional Cardiology, and DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Ina Hoeschele
- Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA
| | - David Herrington
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Charles E McCall
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Yongmei Liu
- Department of Epidemiology & Prevention, Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|
29
|
Yang M, Wang R, Sun J, Yu K, Chen B, Xu L, Zhao B, Wang H. The liver X receptor agonist TO901317 protects mice against cisplatin-induced kidney injury. Exp Biol Med (Maywood) 2015; 240:1717-27. [PMID: 26062799 DOI: 10.1177/1535370215589906] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 04/21/2015] [Indexed: 12/13/2022] Open
Abstract
Liver X receptors are in the nuclear receptor superfamily and are contained in the regulation of lipid and cholesterol metabolism. Besides, liver X receptors are considered crucial regulators of the inflammatory response and innate immunity. The current study evaluates the in vivo effects that the synthetic liver X receptor agonist TO901317 protects against cisplatin-induced kidney injury in mice. Mice received cisplatin administration through a single intraperitoneal injection (20 mg/kg in saline). And then the mice were treated with the TO901317 by daily gavage (10 mg/kg/day) 12 h postcisplatin administration, and cisplatin nephrotoxicity was evaluated. At 72 h after cisplatin treatment, elevated plasma urea and creatinine levels (P < 0.05) were evidenced which indicates the renal dysfunction of the vehicle-treated mice, consistent with tubular necrosis, protein cast, dilation of renal tubules, and desquamation of epithelial cells in renal tubules. In contrast, the severity of renal dysfunction and histological damage was reduced in TO901317 treated mice (P < 0.05). In accordance, circulating tumor necrosis factor alpha levels, renal tumor necrosis factor alpha, p47(phox), gp91(phox), and protein expression levels and COX-2 mRNA, renal monocyte chemoattractant protein 1, VACAM-1 mRNA and intercellular adhesion molecule-1 contents, and renal prostaglandin E2 amounts, were higher in samples from cisplatin-treated mice in comparison with controls (P < 0.05) but attenuated in the TO901317 treatment group (P < 0.05). Taken together, treatment with the liver X receptor agonist TO901317 ameliorated the inflammatory response and oxidative stress in cisplatin-induced kidney injury in mice.
Collapse
Affiliation(s)
- Meng Yang
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Rong Wang
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Jing Sun
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Kezhou Yu
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Bing Chen
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Liang Xu
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Bing Zhao
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| | - Haiping Wang
- Department of Nephrology, Provincial Hospital Affiliated to Shandong University, Jinan 250013, Shandong, China
| |
Collapse
|
30
|
Choi JY, Seo JY, Yoon YS, Lee YJ, Kim HS, Kang JL. Mer signaling increases the abundance of the transcription factor LXR to promote the resolution of acute sterile inflammation. Sci Signal 2015; 8:ra21. [PMID: 25714463 DOI: 10.1126/scisignal.2005864] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The receptor tyrosine kinase Mer plays a central role in inhibiting the inflammatory response of immune cells to pathogens. We aimed to understand the function of Mer signaling in the resolution of sterile inflammation in experiments with a Mer-neutralizing antibody or with Mer-deficient (Mer-/-) mice in a model of sterile, zymosan-induced acute inflammation. We found that inhibition or deficiency of Mer enhanced local and systemic inflammatory responses. The exacerbated inflammatory responses induced by the lack of Mer signaling were associated with reduced abundance of the transcription factors liver X receptor α (LXRα) and LXRβ and decreased expression of their target genes in peritoneal macrophages, spleens, and lungs. Similarly, treatment of mice with a Mer/Fc fusion protein, which prevents the Mer ligand Gas6 (growth arrest-specific protein 6) from binding to Mer, exacerbated the inflammatory response and decreased the abundance of LXR. Coadministration of the LXR agonist T0901317 with the Mer-neutralizing antibody inhibited the aggravating effects of the antibody on inflammation in mice. In vitro exposure of RAW264.7 cells or primary peritoneal macrophages to Gas6 increased LXR abundance in an Akt-dependent manner. Thus, we have elucidated a previously uncharacterized pathway involved in the resolution of acute sterile inflammation: Enhanced Mer signaling during the recovery phase increases the abundance and activity of LXR to inactivate the inflammatory response in macrophages.
Collapse
Affiliation(s)
- Ji-Yeon Choi
- Department of Physiology, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 158-056, Korea
| | - Jeong Yeon Seo
- Department of Physiology, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 158-056, Korea
| | - Young-So Yoon
- Department of Physiology, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 158-056, Korea
| | - Ye-Ji Lee
- Department of Physiology, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 158-056, Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 158-056, Korea
| | - Jihee Lee Kang
- Department of Physiology, Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul 158-056, Korea.
| |
Collapse
|
31
|
Secor McVoy JR, Oughli HA, Oh U. Liver X receptor-dependent inhibition of microglial nitric oxide synthase 2. J Neuroinflammation 2015; 12:27. [PMID: 25889344 PMCID: PMC4332930 DOI: 10.1186/s12974-015-0247-2] [Citation(s) in RCA: 20] [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: 06/16/2014] [Accepted: 01/14/2015] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The nuclear receptor liver X receptor (LXR) exerts transcriptional control over lipid metabolism and inflammatory response in cells of the myeloid lineage, suggesting that LXR may be a potential target in a number of chronic neuroinflammatory and neurodegenerative diseases where persistent microglial activation has been implicated in the pathogenesis. METHODS The effect of LXR activation on microglia and central nervous system (CNS) inflammation was studied using a synthetic LXR agonist in cultured microglia, a microglial cell line and experimental allergic encephalomyelitis (EAE), an animal model of CNS inflammation. RESULTS LXR activation inhibited nitric oxide synthase 2, inducible (Nos2) expression and nitric oxide production in lipopolysaccharide (LPS)-stimulated microglia. Inhibition of microglial activation in response to interferon-γ was less reliable. In LPS-stimulated cells, LXR activation did not inhibit nuclear translocation of NF-kappaB1 p50. Instead, LXR-dependent Nos2 repression was associated with inhibition of histone 4 acetylation and inhibition of NF-kappaB1 p50 binding at the Nos2 promoter. Histone acetylation and NF-kappaB1 p50 binding were mechanistically linked, and histone deacetylase (HDAC) activity appeared to be important for LXR-dependent transcriptional repression of Nos2. Analysis of CNS gene expression in animals undergoing EAE showed that the expressions of Lxr and LXR-dependent genes were downregulated during CNS inflammation. Nevertheless, administration of LXR agonist GW3965 during the effector phase of EAE delayed the onset of clinical disease and reversed the diminished expression of LXR-dependent reverse cholesterol transport genes. However, the CNS expressions of Nos2 and other inflammatory genes were not significantly inhibited by LXR activation in EAE, and clinical disease severity was comparable to vehicle controls at later time points in LXR agonist treated animals. CONCLUSIONS LXR can be targeted to modulate microglial activation. LXR-dependent repression of inflammatory genes may be stimulus-dependent and impaired by HDAC inhibition. Endogenous LXR activity does not appear to modulate CNS inflammation, but LXR activity can be partially restored in the CNS by administration of exogenous LXR agonist with an impact on clinical disease severity at early, but not late, time points in EAE.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Benzoates/therapeutic use
- Benzylamines/therapeutic use
- Cells, Cultured
- Chromatin Immunoprecipitation
- Cytokines/metabolism
- Deoxyribonucleases/metabolism
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Gene Expression Profiling
- Histones/metabolism
- Lipopolysaccharides/pharmacology
- Liver X Receptors
- Mice
- Mice, Inbred C57BL
- Mice, Inbred ICR
- Microglia/drug effects
- Microglia/metabolism
- Myelin-Oligodendrocyte Glycoprotein/immunology
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Nitric Oxide Synthase Type II/metabolism
- Orphan Nuclear Receptors/antagonists & inhibitors
- Orphan Nuclear Receptors/metabolism
- Peptide Fragments/immunology
- Peptide Fragments/toxicity
- Protein Binding/drug effects
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
Collapse
Affiliation(s)
- Julie R Secor McVoy
- Department of Neurology, Virginia Commonwealth University School of Medicine, PO Box 980599, VA, 23298, Richmond, USA.
| | - Hanadi Ajam Oughli
- Department of Neurology, Virginia Commonwealth University School of Medicine, PO Box 980599, VA, 23298, Richmond, USA.
| | - Unsong Oh
- Department of Neurology, Virginia Commonwealth University School of Medicine, PO Box 980599, VA, 23298, Richmond, USA.
| |
Collapse
|
32
|
Kurakula K, Sommer D, Sokolovic M, Moerland PD, Scheij S, van Loenen PB, Koenis DS, Zelcer N, van Tiel CM, de Vries CJM. LIM-only protein FHL2 is a positive regulator of liver X receptors in smooth muscle cells involved in lipid homeostasis. Mol Cell Biol 2015; 35:52-62. [PMID: 25332231 PMCID: PMC4295390 DOI: 10.1128/mcb.00525-14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 05/06/2014] [Accepted: 10/07/2014] [Indexed: 11/20/2022] Open
Abstract
The LIM-only protein FHL2 is expressed in smooth muscle cells (SMCs) and inhibits SMC-rich-lesion formation. To further elucidate the role of FHL2 in SMCs, we compared the transcriptomes of SMCs derived from wild-type (WT) and FHL2 knockout (KO) mice. This revealed that in addition to the previously recognized involvement of FHL2 in SMC proliferation, the cholesterol synthesis and liver X receptor (LXR) pathways are altered in the absence of FHL2. Using coimmunoprecipitation experiments, we found that FHL2 interacts with the two LXR isoforms, LXRα and LXRβ. Furthermore, FHL2 strongly enhances transcriptional activity of LXR element (LXRE)-containing reporter constructs. Chromatin immunoprecipitation (ChIP) experiments on the ABCG1 promoter revealed that FHL2 enhances the association of LXRβ with DNA. In line with these observations, we observed reduced basal transcriptional LXR activity in FHL2-KO SMCs compared to WT SMCs. This was also reflected in reduced expression of LXR target genes in intact aorta and aortic SMCs of FHL2-KO mice. Functionally, the absence of FHL2 resulted in attenuated cholesterol efflux to both ApoA-1 and high-density lipoprotein (HDL), in agreement with reduced LXR signaling. Collectively, our findings demonstrate that FHL2 is a transcriptional coactivator of LXRs and points toward FHL2 being an important determinant of cholesterol metabolism in SMCs.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Daniela Sommer
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Milka Sokolovic
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands European Food Information Council, Brussels, Belgium
| | - Perry D Moerland
- Bioinformatics Laboratory, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Amsterdam, The Netherlands
| | - Saskia Scheij
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Pieter B van Loenen
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Duco S Koenis
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Noam Zelcer
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Claudia M van Tiel
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Carlie J M de Vries
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Stashi E, York B, O'Malley BW. Steroid receptor coactivators: servants and masters for control of systems metabolism. Trends Endocrinol Metab 2014; 25:337-47. [PMID: 24953190 PMCID: PMC4108168 DOI: 10.1016/j.tem.2014.05.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 05/12/2014] [Accepted: 05/13/2014] [Indexed: 11/30/2022]
Abstract
Coregulator recruitment to nuclear receptors (NRs) and other transcription factors is essential for proper metabolic gene regulation, with coactivators enhancing and corepressors attenuating gene transcription. The steroid receptor coactivator (SRC) family is composed of three homologous members (SRC-1, SRC-2, and SRC-3), which are uniquely important for mediating steroid hormone and mitogenic actions. An accumulating body of work highlights the diverse array of metabolic functions regulated by the SRCs, including systemic metabolite homeostasis, inflammation, and energy regulation. We discuss here the cooperative and unique functions among the SRCs to provide a comprehensive atlas of systemic SRC metabolic regulation. Deciphering the fractional and synergistic contributions of the SRCs to metabolic homeostasis is crucial to understanding fully the networks underlying metabolic transcriptional regulation.
Collapse
Affiliation(s)
- Erin Stashi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Brian York
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
34
|
The importance of the retinoid X receptor alpha in modulating inflammatory signaling in acute murine colitis. Dig Dis Sci 2014; 59:753-9. [PMID: 24146318 PMCID: PMC4009739 DOI: 10.1007/s10620-013-2902-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 09/27/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND In order for vitamin D to signal and regulate inflammatory pathways, it must bind to its receptor (VDR) which must heterodimerize with the retinoid X receptor alpha (RXRα). Although the role that vitamin D signaling plays in the development and progression of colitis, a disease characterized by excessive inflammation, has been suggested, little research has been done on determining the role that RXRα plays in acute colitis development. AIMS This study sought to determine the effects that reduced availability of RXRα would have on the development of acute murine colitis. Expression of inflammatory markers, VDR and RXRα were investigated to determine if the reduction in expression of RXRα in RXRα(+/-) mice would result in increased inflammatory signaling and receptor downregulation as compared to their wild-type littermates. METHODS An acute murine model of colitis, the axozymethane (AOM) and dextran sulfate sodium (DSS) model was utilized in wild-type and RXRα(+/-) mice. Gross manifestations of colitis measured included weight loss and colitis score. Immunblots and real-time PCR were performed for inflammatory markers and receptor expression. RESULTS RXRα(+/-) mice induced with AOM/DSS colitis demonstrated increased gene expression of Snail and Snail2, transcription factors downstream of inflammatory mediators, as compared to their wild-type littermates. CONCLUSIONS This demonstrates the importance of RXRα in regulating inflammation in acute colitis and also identifies RXRα expression as a new consideration when developing successful interventions for acute colitis due to the requirement of numerous receptors for RXRα.
Collapse
|
35
|
Patel M, Wang XX, Magomedova L, John R, Rasheed A, Santamaria H, Wang W, Tsai R, Qiu L, Orellana A, Advani A, Levi M, Cummins CL. Liver X receptors preserve renal glomerular integrity under normoglycaemia and in diabetes in mice. Diabetologia 2014; 57:435-46. [PMID: 24201575 DOI: 10.1007/s00125-013-3095-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/01/2013] [Indexed: 10/26/2022]
Abstract
AIMS/HYPOTHESIS Liver X receptors (LXRs) α and β are nuclear hormone receptors that are widely expressed in the kidney. They promote cholesterol efflux from cells and inhibit inflammatory responses by regulating gene transcription. Here, we hypothesised (1) that LXR deficiency would promote renal decline in a mouse model of diabetes by accelerating intraglomerular cholesterol accumulation and, conversely, (2) that LXR agonism would attenuate renal decline in diabetes. METHODS Diabetes was induced with streptozotocin (STZ) and maintained for 14 weeks in Lxrα/β (+/+) (Lxrα, also known as Nr1h3; Lxrβ, also known as Nr1h2) and Lxrα/β (-/-) mice. In addition, STZ-injected DBA/2J mice were treated with vehicle or the LXR agonist N,N-dimethyl-hydroxycholenamide (DMHCA) (80 mg/kg daily) for 10 weeks. To determine the role of cholesterol in diabetic nephropathy (DN), mice were placed on a Western diet after hyperglycaemia developed. RESULTS Even in the absence of diabetes, Lxrα/β (-/-) mice exhibited a tenfold increase in the albumin:creatinine ratio and a 40-fold increase in glomerular lipid accumulation compared with Lxrα/β (+/+) mice. When challenged with diabetes, Lxrα/β (-/-) mice showed accelerated mesangial matrix expansion and glomerular lipid accumulation, with upregulation of inflammatory and oxidative stress markers. In the DN-sensitive STZ DBA/2J mouse model, DMHCA treatment significantly decreased albumin and nephrin excretion (by 50% each), glomerular lipids and plasma triacylglycerol (by 70%) and cholesterol (by 48%); it also decreased kidney inflammatory and oxidative stress markers compared with vehicle-treated mice. CONCLUSIONS/INTERPRETATION These data support the idea that LXR plays an important role in the normal and diabetic kidney, while showing that LXR, through its inhibitory effect on inflammation and cholesterol accumulation in glomeruli, could also be a novel therapeutic target for DN.
Collapse
Affiliation(s)
- Monika Patel
- Faculty of Pharmacy, University of Toronto, 144 College St, Toronto, ON, M5S 3M2, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Infliximab reverses suppression of cholesterol efflux proteins by TNF-α: a possible mechanism for modulation of atherogenesis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:312647. [PMID: 24587984 PMCID: PMC3920897 DOI: 10.1155/2014/312647] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 10/29/2013] [Accepted: 10/30/2013] [Indexed: 02/08/2023]
Abstract
Tumor necrosis factor- (TNF-) α is a proinflammatory proatherogenic cytokine. Infliximab, an anti-TNF-α monoclonal antibody, is effective in treating rheumatoid arthritis. However, its impact on cardiovascular burden and lipid transport is unclear. The present study investigates the effect of TNF-α and infliximab on reverse cholesterol transport (RCT) proteins. Uptake of modified lipoproteins by macrophages in the vasculature leads to atherogenic foam cell formation. RCT is mediated by proteins including ATP binding cassette transporters A1 (ABCA1), G1 (ABCG1), liver X receptor- (LXR-) α, and 27-hydroxylase. RCT counteracts lipid overload by ridding cells of excess cholesterol. THP-1 human monocytes were incubated with either TNF-α alone or TNF-α with infliximab. Expression of proteins involved in cholesterol efflux was analyzed. TNF-α significantly reduced both ABCA1 and LXR-α mRNA (to 68.5 ± 1.59%, P < 0.05, and 41.2 ± 0.25%, P < 0.01, versus control set as 100%, resp.). Infliximab nullified the TNF-α effect. Results were confirmed by Western blot. Infliximab abolished the increase in foam cells induced by TNF-α. TNF-α treatment significantly reduces ABCA1 and LXR-α expression in monocytes, thus bringing about a proatherogenic state. The anti-TNF drug infliximab, commonly used in rheumatology, restored RCT proteins. This is the first report of an atheroprotective effect of infliximab on RCT in monocytes.
Collapse
|
37
|
Herman-Edelstein M, Scherzer P, Tobar A, Levi M, Gafter U. Altered renal lipid metabolism and renal lipid accumulation in human diabetic nephropathy. J Lipid Res 2013; 55:561-72. [PMID: 24371263 DOI: 10.1194/jlr.p040501] [Citation(s) in RCA: 445] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Animal models link ectopic lipid accumulation to renal dysfunction, but whether this process occurs in the human kidney is uncertain. To this end, we investigated whether altered renal TG and cholesterol metabolism results in lipid accumulation in human diabetic nephropathy (DN). Lipid staining and the expression of lipid metabolism genes were studied in kidney biopsies of patients with diagnosed DN (n = 34), and compared with normal kidneys (n = 12). We observed heavy lipid deposition and increased intracellular lipid droplets. Lipid deposition was associated with dysregulation of lipid metabolism genes. Fatty acid β-oxidation pathways including PPAR-α, carnitine palmitoyltransferase 1, acyl-CoA oxidase, and L-FABP were downregulated. Downregulation of renal lipoprotein lipase, which hydrolyzes circulating TGs, was associated with increased expression of angiopoietin-like protein 4. Cholesterol uptake receptor expression, including LDL receptors, oxidized LDL receptors, and acetylated LDL receptors, was significantly increased, while there was downregulation of genes effecting cholesterol efflux, including ABCA1, ABCG1, and apoE. There was a highly significant correlation between glomerular filtration rate, inflammation, and lipid metabolism genes, supporting a possible role of abnormal lipid metabolism in the pathogenesis of DN. These data suggest that renal lipid metabolism may serve as a target for specific therapies aimed at slowing the progression of glomerulosclerosis.
Collapse
Affiliation(s)
- Michal Herman-Edelstein
- Felsenstein Medical Research Center, and Departments of Nephrology Rabin Medical Center, Sackler School of Medicine Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | |
Collapse
|
38
|
Freilich RW, Woodbury ME, Ikezu T. Integrated expression profiles of mRNA and miRNA in polarized primary murine microglia. PLoS One 2013; 8:e79416. [PMID: 24244499 PMCID: PMC3823621 DOI: 10.1371/journal.pone.0079416] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 09/23/2013] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation contributes to many neurologic disorders including Alzheimer’s disease, multiple sclerosis, and stroke. Microglia is brain resident myeloid cells and have emerged as a key driver of the neuroinflammatory responses. MicroRNAs (miRNAs) provide a novel layer of gene regulation and play a critical role in regulating the inflammatory response of peripheral macrophages. However, little is known about the miRNA in inflammatory activation of microglia. To elucidate the role that miRNAs have on microglial phenotypes under classical (M1) or alternative (M2) activation under lipopolysaccharide (‘M1’-skewing) and interleukin-4 (‘M2a’-skewing) stimulation conditions, we performed microarray expression profiling and bioinformatics analysis of both mRNA and miRNA using primary cultured murine microglia. miR-689, miR-124, and miR-155 were the most strongly associated miRNAs predicted to mediate pro-inflammatory pathways and M1-like activation phenotype. miR-155, the most strongly up-regulated miRNA, regulates the signal transducer and activator of transcription 3 signaling pathway enabling the late phase response to M1-skewing stimulation. Reduced expression in miR-689 and miR-124 are associated with dis-inhibition of many canonical inflammatory pathways. miR-124, miR-711, miR-145 are the strongly associated miRNAs predicted to mediate anti-inflammatory pathways and M2-like activation phenotype. Reductions in miR-711 and miR-124 may regulate inflammatory signaling pathways and peroxisome proliferator-activated receptor-gamma pathway. miR-145 potentially regulate peripheral monocyte/macrophage differentiation and faciliate the M2-skewing phenotype. Overall, through combined miRNA and mRNA expression profiling and bioinformatics analysis we have identified six miRNAs and their putative roles in M1 and M2-skewing of microglial activation through different signaling pathways.
Collapse
Affiliation(s)
- Robert W. Freilich
- Laboratory of Molecular NeuroTherapeutics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Maya E. Woodbury
- Laboratory of Molecular NeuroTherapeutics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Graduate Program in Neuroscience, Boston University, Boston, Massachusetts, United States of America
| | - Tsuneya Ikezu
- Laboratory of Molecular NeuroTherapeutics, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Neurology and Alzheimer’s Disease Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
39
|
Smith AJP, Howard P, Shah S, Eriksson P, Stender S, Giambartolomei C, Folkersen L, Tybjærg-Hansen A, Kumari M, Palmen J, Hingorani AD, Talmud PJ, Humphries SE. Use of allele-specific FAIRE to determine functional regulatory polymorphism using large-scale genotyping arrays. PLoS Genet 2012; 8:e1002908. [PMID: 22916038 PMCID: PMC3420950 DOI: 10.1371/journal.pgen.1002908] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 07/02/2012] [Indexed: 11/18/2022] Open
Abstract
Following the widespread use of genome-wide association studies (GWAS), focus is turning towards identification of causal variants rather than simply genetic markers of diseases and traits. As a step towards a high-throughput method to identify genome-wide, non-coding, functional regulatory variants, we describe the technique of allele-specific FAIRE, utilising large-scale genotyping technology (FAIRE-gen) to determine allelic effects on chromatin accessibility and regulatory potential. FAIRE-gen was explored using lymphoblastoid cells and the 50,000 SNP Illumina CVD BeadChip. The technique identified an allele-specific regulatory polymorphism within NR1H3 (coding for LXR-α), rs7120118, coinciding with a previously GWAS-identified SNP for HDL-C levels. This finding was confirmed using FAIRE-gen with the 200,000 SNP Illumina Metabochip and verified with the established method of TaqMan allelic discrimination. Examination of this SNP in two prospective Caucasian cohorts comprising 15,000 individuals confirmed the association with HDL-C levels (combined beta = 0.016; p = 0.0006), and analysis of gene expression identified an allelic association with LXR-α expression in heart tissue. Using increasingly comprehensive genotyping chips and distinct tissues for examination, FAIRE-gen has the potential to aid the identification of many causal SNPs associated with disease from GWAS.
Collapse
Affiliation(s)
- Andrew J P Smith
- Centre for Cardiovascular Genetics, British Heart Foundation Laboratories, Institute of Cardiovascular Sciences, University College London, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Involvement of the Retinoid X Receptor Ligand in the Anti-Inflammatory Effect Induced by Peroxisome Proliferator-Activated Receptor γ Agonist In Vivo. PPAR Res 2011; 2011:840194. [PMID: 22190910 PMCID: PMC3236425 DOI: 10.1155/2011/840194] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 09/13/2011] [Indexed: 12/18/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) forms a heterodimeric DNA-binding complex with retinoid X receptors (RXRs). It has been reported that the effect of the PPAR agonist is reduced in hepatocyte RXR-deficient mice. Therefore, it is suggested that the endogenous RXR ligand is involved in the PPARγ agonist-induced anti-inflammatory effect. However, the participation of the RXR ligand in the PPARγ-induced anti-inflammatory effect is unknown. Here, we investigated the influence of RXR antagonist on the anti-inflammatory effect of PPARγ agonist pioglitazone in carrageenan test. In addition, we also examined the influence of PPAR antagonist on the anti-inflammatory effect induced by RXR agonist NEt-3IP. The RXR antagonist suppressed the antiedema effect of PPARγ agonist. In addition, the anti-inflammatory effect of RXR agonist was suppressed by PPARγ antagonist. PPARγ agonist-induced anti-inflammatory effects were reversed by the RXR antagonist. Thus, we showed that the endogenous RXR ligand might contribute to the PPARγ agonist-induced anti-inflammatory effect.
Collapse
|
41
|
Tran M, Tam D, Bardia A, Bhasin M, Rowe GC, Kher A, Zsengeller ZK, Akhavan-Sharif MR, Khankin EV, Saintgeniez M, David S, Burstein D, Karumanchi SA, Stillman IE, Arany Z, Parikh SM. PGC-1α promotes recovery after acute kidney injury during systemic inflammation in mice. J Clin Invest 2011; 121:4003-14. [PMID: 21881206 DOI: 10.1172/jci58662] [Citation(s) in RCA: 396] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 07/13/2011] [Indexed: 01/16/2023] Open
Abstract
Sepsis-associated acute kidney injury (AKI) is a common and morbid condition that is distinguishable from typical ischemic renal injury by its paucity of tubular cell death. The mechanisms underlying renal dysfunction in individuals with sepsis-associated AKI are therefore less clear. Here we have shown that endotoxemia reduces oxygen delivery to the kidney, without changing tissue oxygen levels, suggesting reduced oxygen consumption by the kidney cells. Tubular mitochondria were swollen, and their function was impaired. Expression profiling showed that oxidative phosphorylation genes were selectively suppressed during sepsis-associated AKI and reactivated when global function was normalized. PPARγ coactivator-1α (PGC-1α), a major regulator of mitochondrial biogenesis and metabolism, not only followed this pattern but was proportionally suppressed with the degree of renal impairment. Furthermore, tubular cells had reduced PGC-1α expression and oxygen consumption in response to TNF-α; however, excess PGC-1α reversed the latter effect. Both global and tubule-specific PGC-1α-knockout mice had normal basal renal function but suffered persistent injury following endotoxemia. Our results demonstrate what we believe to be a novel mechanism for sepsis-associated AKI and suggest that PGC-1α induction may be necessary for recovery from this disorder, identifying a potential new target for future therapeutic studies.
Collapse
Affiliation(s)
- Mei Tran
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kuiper JW, Vaschetto R, Della Corte F, Plötz FB, Groeneveld ABJ. Bench-to-bedside review: Ventilation-induced renal injury through systemic mediator release--just theory or a causal relationship? CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:228. [PMID: 21884646 PMCID: PMC3387589 DOI: 10.1186/cc10282] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We review the current literature on the molecular mechanisms involved in the pathogenesis of acute kidney injury induced by plasma mediators released by mechanical ventilation. A comprehensive literature search in the PubMed database was performed and articles were identified that showed increased plasma levels of mediators where the increase was solely attributable to mechanical ventilation. A subsequent search revealed articles delineating the potential effects of each mediator on the kidney or kidney cells. Limited research has focused specifically on the relationship between mechanical ventilation and acute kidney injury. Only a limited number of plasma mediators has been implicated in mechanical ventilation-associated acute kidney injury. The number of mediators released during mechanical ventilation is far greater and includes pro- and anti-inflammatory mediators, but also mediators involved in coagulation, fibrinolysis, cell adhesion, apoptosis and cell growth. The potential effects of these mediators is pleiotropic and include effects on inflammation, cell recruitment, adhesion and infiltration, apoptosis and necrosis, vasoactivity, cell proliferation, coagulation and fibrinolysis, transporter regulation, lipid metabolism and cell signaling. Most research has focused on inflammatory and chemotactic mediators. There is a great disparity of knowledge of potential effects on the kidney between different mediators. From a theoretical point of view, the systemic release of several mediators induced by mechanical ventilation may play an important role in the pathophysiology of acute kidney injury. However, evidence supporting a causal relationship is lacking for the studied mediators.
Collapse
Affiliation(s)
- Jan Willem Kuiper
- Department of Pediatric Intensive Care, VUmc Medical Center, 1007 MB Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
43
|
Van Bogaert T, Vandevyver S, Dejager L, Van Hauwermeiren F, Pinheiro I, Petta I, Engblom D, Kleyman A, Schütz G, Tuckermann J, Libert C. Tumor necrosis factor inhibits glucocorticoid receptor function in mice: a strong signal toward lethal shock. J Biol Chem 2011; 286:26555-67. [PMID: 21646349 DOI: 10.1074/jbc.m110.212365] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
As glucocorticoid resistance (GCR) and the concomitant burden pose a worldwide problem, there is an urgent need for a more effective glucocorticoid therapy, for which insights into the molecular mechanisms of GCR are essential. In this study, we addressed the hypothesis that TNFα, a strong pro-inflammatory mediator in numerous inflammatory diseases, compromises the protective function of the glucocorticoid receptor (GR) against TNFα-induced lethal inflammation. Indeed, protection of mice by dexamethasone against TNFα lethality was completely abolished when it was administered after TNFα stimulation, indicating compromised GR function upon TNFα challenge. TNFα-induced GCR was further demonstrated by impaired GR-dependent gene expression in the liver. Furthermore, TNFα down-regulates the levels of both GR mRNA and protein. However, this down-regulation seems to occur independently of GC production, as TNFα also resulted in down-regulation of GR levels in adrenalectomized mice. These findings suggest that the decreased amount of GR determines the GR response and outcome of TNFα-induced shock, as supported by our studies with GR heterozygous mice. We propose that by inducing GCR, TNFα inhibits a major brake on inflammation and thereby amplifies the pro-inflammatory response. Our findings might prove helpful in understanding GCR in inflammatory diseases in which TNFα is intimately involved.
Collapse
Affiliation(s)
- Tom Van Bogaert
- Department for Molecular Biomedical Research, VIB, 9052 Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Normally macrophages localized in the arterial vessel wall perform the "reverse transfer" of cholesterol, which includes endocytosis of low density lipoproteins (LDL), cholesterol transfer to newly formed high density lipoprotein particles, and their following elimination by the liver. The homeostatic function of macrophages for cholesterol involves a system of lipid sensors. Oxysterol sensors LXRs, oxysterol and cholesterol sensors INSIG and SCAP acting through controlled transcription factors SREBP, as well as sensors for oxidized fatty acids and their derivatives, PPAR, are the best studied. Activation of LXR and PPAR is also accompanied by inhibition of macrophage functions related to inflammation. Accumulation of oxidized and otherwise modified LDL in the subendothelial space induced by endothelium injury, infection, or other pathogenic factors instead of stimulation of the homeostatic functions of macrophages leads to their weakening with a concurrent increase in the inflammatory potential of these cells. These shifts seem to drive the transformation of macrophages into foam cells, which form the core of sclerotic plaques. The intervention of another lipid sensor, TLR4, can trigger such a radical change in the functional activity of macrophages. The interaction of modified LDL with this signaling receptor results in inhibition of the homeostatic oxysterol signaling, induction of additional LDL transporters, and activation of the phagocytic function of macrophages. The re-establishment of cholesterol homeostasis under these circumstances can be achieved by administration of LXR and PPARgamma agonists. Therefore, it is urgent to design ligands with reduced side effects.
Collapse
|
45
|
Daneman R, Zhou L, Agalliu D, Cahoy JD, Kaushal A, Barres BA. The mouse blood-brain barrier transcriptome: a new resource for understanding the development and function of brain endothelial cells. PLoS One 2010; 5:e13741. [PMID: 21060791 PMCID: PMC2966423 DOI: 10.1371/journal.pone.0013741] [Citation(s) in RCA: 439] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 08/24/2010] [Indexed: 01/21/2023] Open
Abstract
The blood-brain barrier (BBB) maintains brain homeostasis and limits the entry of toxins and pathogens into the brain. Despite its importance, little is known about the molecular mechanisms regulating the development and function of this crucial barrier. In this study we have developed methods to highly purify and gene profile endothelial cells from different tissues, and by comparing the transcriptional profile of brain endothelial cells with those purified from the liver and lung, we have generated a comprehensive resource of transcripts that are enriched in the BBB forming endothelial cells of the brain. Through this comparison we have identified novel tight junction proteins, transporters, metabolic enzymes, signaling components, and unknown transcripts whose expression is enriched in central nervous system (CNS) endothelial cells. This analysis has identified that RXRalpha signaling cascade is specifically enriched at the BBB, implicating this pathway in regulating this vital barrier. This dataset provides a resource for understanding CNS endothelial cells and their interaction with neural and hematogenous cells.
Collapse
Affiliation(s)
- Richard Daneman
- Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America.
| | | | | | | | | | | |
Collapse
|
46
|
Fon Tacer K, Pompon D, Rozman D. Adaptation of cholesterol synthesis to fasting and TNF-alpha: profiling cholesterol intermediates in the liver, brain, and testis. J Steroid Biochem Mol Biol 2010; 121:619-25. [PMID: 20206258 DOI: 10.1016/j.jsbmb.2010.02.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 02/16/2010] [Accepted: 02/24/2010] [Indexed: 12/18/2022]
Abstract
Key players in pathogenesis of metabolic disorders are disturbed cholesterol balance and inflammation. In addition to cholesterol also sterol intermediates are biologically active, however, surprisingly little is known about their synthesis and roles. The aim of our study was to assess the interplay between the inflammatory cytokine TNF-alpha and cholesterol synthesis by measuring cholesterol and its intermediates in the liver, brain, and testis. Liquid chromatography-mass spectrometry has been applied to profile sterols of normally fed mice, during fasting and after TNF-alpha administration. In mice on normal chow diet, sterols other than cholesterol represent 0.5% in the liver, 1% in brain and 5% in testis. In the liver only 7-dehydrocholesterol, lanosterol and desmosterol were detected. Major sterol intermediates of the brain are desmosterol, testis meiosis activating sterol (T-MAS), and 7-dehydrocholesterol while in testis T-MAS predominates (4%), followed by desmosterol, lanosterol, 7-dehydrocholesterol and others. In 20h fasting there is no significant change in cholesterol of the three tissues, and no significant change in intermediates of the liver. In the brain sterol intermediates are lowered (significant for zymosterol) while in the testis the trend is opposite. TNF-alpha provokes a significant raise of some intermediates whereas the level of cholesterol is again unchanged. The proportion of sterols in the liver rises from 0.5% in controls to 1.2% in TNF-alpha-treated mice, which is in accordance with published expression profiling data. In conclusion, our data provide novel insights into the interaction between the inflammatory cytokine TNF-alpha and the tissue-specific cholesterol biosynthesis of the liver, brain and testis.
Collapse
Affiliation(s)
- Klementina Fon Tacer
- Center for Functional Genomic and Biochips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| | | | | |
Collapse
|
47
|
Vitamin A deficiency results in dysregulation of lipid efflux pathway in rat kidney. Pediatr Nephrol 2010; 25:1435-44. [PMID: 20480185 DOI: 10.1007/s00467-010-1532-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2009] [Revised: 03/03/2010] [Accepted: 03/25/2010] [Indexed: 10/19/2022]
Abstract
The mechanisms of action of vitamin A deficiency (VAD) on lipid metabolism in the rat kidney were investigated in adult female rats and their offspring. The rats were randomized into three groups: (1) control, in which the mother and offspring received a normal diet (4000 retinol IU/kg diet) for 8 weeks; (2) VAD group, in which the mothers and offspring received a VAD diet (400 retinol IU/kg diet) for 8 weeks; (3) vitamin A-refed group, in which a group of pups on a VAD diet for 8 weeks received a complete diet (6500 retinol IU/kg diet) for 15 days. The lipid metabolism of the offsprings' kidneys and its relation to the expression of apolipoprotein B100 (Apo-B100), liver X receptor alpha (LXRalpha), and retinoid X receptor-alpha/beta (RXRalpha/beta) mRNA was analyzed. VAD was found to alter renal lipid metabolism and its immune environment due to the expression of Apo-B100. Compared with the control, VAD rats had significantly higher levels of transforming growth factor-beta 1 and lower levels of ABCA1, a key gene involved in cholesterol efflux and tissue lipid homeostasis. The expression of LXRalpha and RXRalpha/beta mRNA also decreased in the VAD rat kidney. Vitamin A refeeding reversed all of the changes. Lipid metabolism involved in renal reverse cholesterol transport may be mediated by decreasing the signaling through the ABCA1 cholesterol efflux pathway, which is significantly modified in kidneys of vitamin A-deficient rats.
Collapse
|
48
|
Yin K, Liao DF, Tang CK. ATP-binding membrane cassette transporter A1 (ABCA1): a possible link between inflammation and reverse cholesterol transport. Mol Med 2010; 16:438-49. [PMID: 20485864 DOI: 10.2119/molmed.2010.00004] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 05/11/2010] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is characterized by a chronic inflammatory condition that involves numerous cellular and molecular inflammatory components. A wide array of inflammatory mediators, such as cytokines and proteins produced by macrophages and other cells, play a critical role in the development and progression of the disease. ATP-binding membrane cassette transporter A1 (ABCA1) is crucial for cellular cholesterol efflux and reverse cholesterol transport (RCT) and is also identified as an important target in antiatherosclerosis treatment. Evidence from several recent studies indicates that inflammation, along with other atherogenic-related mediators, plays distinct regulating roles in ABCA1 expression. Proatherogenic cytokines such as interferon (IFN)-γ and interleukin (IL)-1β have been shown to inhibit the expression of ABCA1, while antiatherogenic cytokines, including IL-10 and transforming growth factor (TGF)-β1, have been shown to promote the expression of ABCA1. Moreover, some cytokines such as tumor necrosis factor (TNF)-α seem to regulate ABCA1 expression in species-specific and dose-dependent manners. Inflammatory proteins such as C-reactive protein (CRP) and cyclooxygenase (COX)-2 are likely to inhibit ABCA1 expression during inflammation, and inflammation induced by lipopolysaccharide (LPS) was also found to block the expression of ABCA1. Interestingly, recent experiments revealed ABCA1 can function as an antiinflammatory receptor to suppress the expression of inflammatory factors, suggesting that ABCA1 may be the molecular basis for the interaction between inflammation and RCT. This review aims to summarize recent findings on the role of inflammatory cytokines, inflammatory proteins, inflammatory lipids, and the endotoxin-mediated inflammatory process in expression of ABCA1. Also covered is the current understanding of the function of ABCA1 in modulating the immune response and inflammation through its direct and indirect antiinflammatory mechanisms including lipid transport, high-density lipoprotein (HDL) formation and apoptosis.
Collapse
Affiliation(s)
- Kai Yin
- Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Life Science Research Center, University of South China, Hengyang, China
| | | | | |
Collapse
|
49
|
Van Bogaert T, De Bosscher K, Libert C. Crosstalk between TNF and glucocorticoid receptor signaling pathways. Cytokine Growth Factor Rev 2010; 21:275-86. [PMID: 20456998 DOI: 10.1016/j.cytogfr.2010.04.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
TNF is a Janus-faced protein. It possesses impressive anti-tumor activities, but it is also one of the strongest known pro-inflammatory cytokines, which hampers its use as a systemic anti-cancer agent. TNF has been shown to play a detrimental role in inflammatory diseases such as rheumatoid arthritis and inflammatory bowel disease. Glucocorticoids are strongly anti-inflammatory and exert their therapeutic effects through binding to their receptor, the glucocorticoid receptor. Therefore, glucocorticoids have been used for over half a century for the treatment of inflammatory diseases. However, many patients are or become resistant to the therapeutic effects of glucocorticoids. Inflammatory cytokines have been suggested to play an important role in this steroid insensitivity or glucocorticoid resistance. This review aims to highlight the mechanisms of mutual inhibition between TNF and GR signaling pathways.
Collapse
Affiliation(s)
- Tom Van Bogaert
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | | | | |
Collapse
|
50
|
Effects of Liver x receptor agonist treatment on signal transduction pathways in acute lung inflammation. Respir Res 2010; 11:19. [PMID: 20175894 PMCID: PMC2836283 DOI: 10.1186/1465-9921-11-19] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Accepted: 02/22/2010] [Indexed: 12/16/2022] Open
Abstract
Background Liver × receptor α (LXRα) and β (LXRβ) are members of the nuclear receptor super family of ligand-activated transcription factors, a super family which includes the perhaps better known glucocorticoid receptor, estrogen receptor, thyroid receptor, and peroxisome proliferator-activated receptors. There is limited evidence that LXL activation may reduces acute lung inflammation. The aim of this study was to investigate the effects of T0901317, a potent LXR receptor ligand, in a mouse model of carrageenan-induced pleurisy. Methods Injection of carrageenan into the pleural cavity of mice elicited an acute inflammatory response characterized by: accumulation of fluid containing a large number of neutrophils (PMNs) in the pleural cavity, infiltration of PMNs in lung tissues and subsequent lipid peroxidation, and increased production of nitrite/nitrate (NOx), tumor necrosis factor-α, (TNF-α) and interleukin-1β (IL-1β). Furthermore, carrageenan induced the expression of iNOS, nitrotyrosine and PARP, as well as induced apoptosis (TUNEL staining and Bax and Bcl-2 expression) in the lung tissues. Results Administration of T0901317, 30 min after the challenge with carrageenan, caused a significant reduction in a dose dependent manner of all the parameters of inflammation measured. Conclusions Thus, based on these findings we propose that LXR ligand such as T0901317, may be useful in the treatment of various inflammatory diseases.
Collapse
|