51
|
Jiang Y, Feng YP, Tang LX, Yan YL, Bai JW. The protective role of NR4A3 in acute myocardial infarction by suppressing inflammatory responses via JAK2-STAT3/NF-κB pathway. Biochem Biophys Res Commun 2019; 517:697-702. [DOI: 10.1016/j.bbrc.2019.07.116] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 07/29/2019] [Indexed: 02/01/2023]
|
52
|
NURR1 Impairment in Multiple Sclerosis. Int J Mol Sci 2019; 20:ijms20194858. [PMID: 31574937 PMCID: PMC6801584 DOI: 10.3390/ijms20194858] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/27/2019] [Accepted: 09/28/2019] [Indexed: 02/07/2023] Open
Abstract
The transcription factor NURR1 is a constitutively active orphan receptor belonging to the steroid hormone receptor class NR4A. Although a genetic association between NURR1 and autoimmune inflammatory diseases has never emerged from genome-wide association studies (GWAS), alterations in the expression of NURR1 have been observed in various autoimmune diseases. Specifically, its role in autoimmune inflammatory diseases is mainly related to its capability to counteract inflammation. In fact, NURR1 exerts anti-inflammatory functions inhibiting the transcription of the molecules involved in proinflammatory pathways, not only in the peripheral blood compartment, but also in the cerebral parenchyma acting in microglial cells and astrocytes. In parallel, NURR1 has been also linked to dopamine-associated brain disorders, such as Parkinson’s disease (PD) and schizophrenia, since it is involved in the development and in the maintenance of midbrain dopaminergic neurons (mDA). Considering its role in neuro- and systemic inflammatory processes, here we review the evidences supporting its contribution to multiple sclerosis (MS), a chronic inflammatory autoimmune disease affecting the central nervous system (CNS). To date, the specific role of NURR1 in MS is still debated and few authors have studied this topic. Here, we plan to clarify this issue analyzing the reported association between NURR1 and MS in human and murine model studies.
Collapse
|
53
|
NR4A1 regulates cerebral ischemia-induced brain injury by regulating neuroinflammation through interaction with NF-κB/p65. Biochem Biophys Res Commun 2019; 518:59-65. [PMID: 31445702 DOI: 10.1016/j.bbrc.2019.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 08/03/2019] [Indexed: 12/12/2022]
Abstract
Stroke is reported as a leading cause of mortality and disability in the world. Neuroinflammation is significantly induced responding to ischemic stroke, and this process is accompanied with microglial activation. However, the pathogenesis contributing to ischemic stroke remains unclear. NR4A1 (Nur77) is a nuclear receptor that is expressed in macrophages, playing a significant role in regulating inflammatory response. In the present study, we attempted to explore the effects of NR4A1 on ischemic stroke using in vivo and in vitro studies. Results suggested that NR4A1 expression in microglia was markedly increased after cerebral ischemic damage. Then, we found that NR4A1 knockout attenuated ischemia-triggered infarction volume and neuron injury. Also, cognitive impairments were improved in ischemic mice with NR4A1 deficiency, resulting in functional improvements. Moreover, M1 polarization in microglia and neutrophil recruitment was significantly alleviated by NR4A1 deletion, as evidenced by the reduced expression of M1 markers, chemokines, as well as intracellular adhesion molecule-1 (ICAM-1) and myeloperoxidase (MPO) levels. Importantly, we found that NR4A1 could interact with nuclear factor-κB (NF-κB)/p65 based on in vivo and in vitro results. Suppressing p65 activation by the use of its inhibitor clearly reduced the NR4A1 expression, M1 polarization and neutrophil recruitments, while rescued the expression of anti-inflammatory factors in microglia treated with oxygen-glucose deprivation (OGD). Therefore, NR4A1 suppression in microglia restrained neuroinflammation through interacting with NF-κB/p65 to attenuate ischemic stroke.
Collapse
|
54
|
Leopold Wager CM, Arnett E, Schlesinger LS. Macrophage nuclear receptors: Emerging key players in infectious diseases. PLoS Pathog 2019; 15:e1007585. [PMID: 30897154 PMCID: PMC6428245 DOI: 10.1371/journal.ppat.1007585] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nuclear receptors (NRs) are ligand-activated transcription factors that are expressed in a variety of cells, including macrophages. For decades, NRs have been therapeutic targets because their activity can be pharmacologically modulated by specific ligands and small molecule inhibitors. NRs regulate a variety of processes, including those intersecting metabolic and immune functions, and have been studied in regard to various autoimmune diseases. However, the complex roles of NRs in host response to infection are only recently being investigated. The NRs peroxisome proliferator-activated receptor γ (PPARγ) and liver X receptors (LXRs) have been most studied in the context of infectious diseases; however, recent work has also linked xenobiotic pregnane X receptors (PXRs), vitamin D receptor (VDR), REV-ERBα, the nuclear receptor 4A (NR4A) family, farnesoid X receptors (FXRs), and estrogen-related receptors (ERRs) to macrophage responses to pathogens. Pharmacological inhibition or antagonism of certain NRs can greatly influence overall disease outcome, and NRs that are protective against some diseases can lead to susceptibility to others. Targeting NRs as a novel host-directed treatment approach to infectious diseases appears to be a viable option, considering that these transcription factors play a pivotal role in macrophage lipid metabolism, cholesterol efflux, inflammatory responses, apoptosis, and production of antimicrobial byproducts. In the current review, we discuss recent findings concerning the role of NRs in infectious diseases with an emphasis on PPARγ and LXR, the two most studied. We also highlight newer work on the activity of emerging NRs during infection.
Collapse
Affiliation(s)
| | - Eusondia Arnett
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Larry S. Schlesinger
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| |
Collapse
|
55
|
A partial agonist for retinoid X receptor mitigates experimental colitis. Int Immunol 2018; 31:251-262. [DOI: 10.1093/intimm/dxy089] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 12/24/2018] [Indexed: 12/14/2022] Open
|
56
|
Dihydromyricetin from ampelopsis grossedentata protects against vascular neointimal formation via induction of TR3. Eur J Pharmacol 2018; 838:23-31. [PMID: 30194942 DOI: 10.1016/j.ejphar.2018.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 11/21/2022]
Abstract
Vine tea has been used as a medicinal herb in traditional Chinese medicine for hundreds of years. As the most abundant ingredient in vine tea, Dihydromyricetin (DHM) has been reported to exert anti-inflammatory, antioxidant, and anti-cardiovascular disease. However, the role of DHM in injury-induced neointimal formation remains poorly characterized. We determined the effects of DHM on ligation-induced carotid artery neointimal formation. We found that ligation-induced carotid artery neointimal formation could be significantly attenuated by DHM treatment. We provide evidence that DHM increases orphan nuclear receptor TR3 expression in smooth muscle cell (SMC) and carotid artery. Moreover, overexpression and loss-of-function strategies of TR3 were done to overexpression and knockdown of TR3, and demonstrate that DHM promotes SMC differentiation, however, inhibits SMC proliferation and migration, via regulating expression of TR3. Collectively, we reveal that DHM may be a therapeutic agent for the treatment of injury-induced vascular diseases.
Collapse
|
57
|
Huang B, Pei HZ, Chang HW, Baek SH. The E3 ubiquitin ligase Trim13 regulates Nur77 stability via casein kinase 2α. Sci Rep 2018; 8:13895. [PMID: 30224829 PMCID: PMC6141542 DOI: 10.1038/s41598-018-32391-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/24/2018] [Indexed: 01/23/2023] Open
Abstract
Nur77 is a member of the NR4A subfamily of nuclear receptors and has been shown to regulate various biological processes such as apoptosis and inflammation. Here, we show that Nur77 ubiquitination is mediated by the tripartite motif 13 (Trim13), a RING-type E3 ubiquitin ligase. The interaction between Nur77 and Trim13 was confirmed by co-immunoprecipitation. Moreover, we found that Lys539 in Nur77 ubiquitination is targeted for Trim13, which leads to Nur77 degradation. The Trim13-mediated ubiquitination of Nur77 was optimal in the presence of the E2 enzyme UbcH5. Importantly, in addition to Trim13-mediated ubiquitination, the stability of Nur77 was also regulated by casein kinase 2α (CK2α). Pharmacological inhibition of CK2 markedly increased Nur77 levels, whereas overexpression of CK2α, but not its inactive mutant, dramatically decreased Nur77 levels by promoting Nur77 ubiquitination. CK2α phosphorylated Ser154 in Nur77 and thereby regulated Nur77 protein levels by promoting its ubiquitin-mediated degradation. Importantly, we also show that degradation of Nur77 is involved in TNFα-mediated IL-6 production via CK2α and Trim13. Taken together, these results suggest that the sequential phosphorylation and ubiquitination of Nur77 controls its degradation, and provide a therapeutic approach for regulating Nur77 activity through the CK2α-Trim13 axis as a mechanism to control the inflammatory response.
Collapse
Affiliation(s)
- Bin Huang
- Department of Biochemistry & Molecular Biology, College of Medicine, Yeungnam University, Daegu, South Korea
| | - Han Zhong Pei
- Department of Biochemistry & Molecular Biology, College of Medicine, Yeungnam University, Daegu, South Korea
| | - Hyeun-Wook Chang
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea.
| | - Suk-Hwan Baek
- Department of Biochemistry & Molecular Biology, College of Medicine, Yeungnam University, Daegu, South Korea.
| |
Collapse
|
58
|
Afzali MF, Popichak KA, Burton LH, Klochak AL, Wilson WJ, Safe S, Tjalkens RB, Legare ME. A novel diindolylmethane analog, 1,1-bis(3'-indolyl)-1-(p-chlorophenyl) methane, inhibits the tumor necrosis factor-induced inflammatory response in primary murine synovial fibroblasts through a Nurr1-dependent mechanism. Mol Immunol 2018; 101:46-54. [PMID: 29870816 PMCID: PMC6138555 DOI: 10.1016/j.molimm.2018.05.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/10/2018] [Accepted: 05/25/2018] [Indexed: 12/15/2022]
Abstract
The progression of rheumatoid arthritis involves the thickening of the synovial lining due to the proliferation of fibroblast-like synoviocytes (FLS) and infiltration by inflammatory cells. Tumor necrosis factor alpha (TNFα) is a pro-inflammatory cytokine involved in progression of the disease. Under rheumatoid conditions, FLS express the tumor necrosis factor (TNF)-recognition complex (TNFR1, TNFR2, VCAM-1 and ICAM-1), which induces local macrophage activation and leads to downstream nuclear factor κB (NF-κB) signaling. The NF-κB-regulated inflammatory gene, cyclooxygenase (COX), increases synthesis of prostaglandins that contribute to the propagation of inflammatory damage within the joint. Because the nuclear orphan receptor, NR4A2 (Nurr1), can negatively regulate NF-κB-dependent inflammatory gene expression in macrophages, we postulated that activation of this receptor by the Nurr1 ligand 1,1-bis(3'-indolyl)-1-(p-chlorophenyl) methane (C-DIM12) would modulate inflammatory gene expression in synovial fibroblasts by inhibiting NF-κB. Treatment with C-DIM12 suppressed TNFα-induced expression of adhesion molecules and NF-κB regulated genes in primary synovial fibroblasts including vascular adhesion molecule 1 (VCAM-1), PGE2 and COX-2. Immunofluorescence studies indicated that C-DIM12 did not prevent translocation of p65 and stabilized nuclear localization of Nurr1 in synovial fibroblasts. Knockdown of Nurr1 expression by RNA interference prevented the inhibitory effects of C-DIM12 on inflammatory gene expression, indicating that the anti-inflammatory effects of this compound are Nurr1-dependent. Collectively, these data suggest that this receptor may be a viable therapeutic target in RA.
Collapse
Affiliation(s)
- Maryam F Afzali
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Center for Environmental Medicine, Colorado State University, Fort Collins, CO, USA
| | - Katriana A Popichak
- Cell & Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
| | - Lindsey H Burton
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Center for Environmental Medicine, Colorado State University, Fort Collins, CO, USA
| | - Anna L Klochak
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Center for Environmental Medicine, Colorado State University, Fort Collins, CO, USA
| | - William J Wilson
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Center for Environmental Medicine, Colorado State University, Fort Collins, CO, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Ronald B Tjalkens
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Center for Environmental Medicine, Colorado State University, Fort Collins, CO, USA
| | - Marie E Legare
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Center for Environmental Medicine, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
59
|
Monson MS, Van Goor AG, Ashwell CM, Persia ME, Rothschild MF, Schmidt CJ, Lamont SJ. Immunomodulatory effects of heat stress and lipopolysaccharide on the bursal transcriptome in two distinct chicken lines. BMC Genomics 2018; 19:643. [PMID: 30165812 PMCID: PMC6117931 DOI: 10.1186/s12864-018-5033-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 08/22/2018] [Indexed: 02/07/2023] Open
Abstract
Background Exposure to heat stress suppresses poultry immune responses, which can increase susceptibility to infectious diseases and, thereby, intensify the negative effects of heat on poultry welfare and performance. Identifying genes and pathways that are affected by high temperatures, especially heat-induced changes in immune responses, could provide targets to improve disease resistance in chickens. This study utilized RNA-sequencing (RNA-seq) to investigate transcriptome responses in the bursa of Fabricius, a primary immune tissue, after exposure to acute heat stress and/or subcutaneous immune stimulation with lipopolysaccharide (LPS) in a 2 × 2 factorial design: Thermoneutral + Saline, Heat + Saline, Thermoneutral + LPS and Heat + LPS. All treatments were investigated in two chicken lines: a relatively heat- and disease-resistant Fayoumi line and a more susceptible broiler line. Results Differential expression analysis determined that Heat + Saline had limited impact on gene expression (N = 1 or 63 genes) in broiler or Fayoumi bursa. However, Thermoneutral + LPS and Heat + LPS generated many expression changes in Fayoumi bursa (N = 368 and 804 genes). Thermoneutral + LPS was predicted to increase immune-related cell signaling and cell migration, while Heat + LPS would activate mortality-related functions and decrease expression in WNT signaling pathways. Further inter-treatment comparisons in the Fayoumi line revealed that heat stress prevented many of the expression changes caused by LPS. Although fewer significant expression changes were observed in the broiler bursa after exposure to Thermoneutral + LPS (N = 59 genes) or to Heat + LPS (N = 146 genes), both treatments were predicted to increase cell migration. Direct comparison between lines (broiler to Fayoumi) confirmed that each line had distinct responses to treatment. Conclusions Transcriptome analysis identified genes and pathways involved in bursal responses to heat stress and LPS and elucidated that these effects were greatest in the combined treatment. The interaction between heat and LPS was line dependent, with suppressive expression changes primarily in the Fayoumi line. Potential target genes, especially those involved in cell migration and immune signaling, can inform future research on heat stress in poultry and could prove useful for improving disease resistance. Electronic supplementary material The online version of this article (10.1186/s12864-018-5033-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melissa S Monson
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | | | | | - Michael E Persia
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Max F Rothschild
- Department of Animal Science, Iowa State University, Ames, IA, USA
| | - Carl J Schmidt
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, USA
| | - Susan J Lamont
- Department of Animal Science, Iowa State University, Ames, IA, USA.
| |
Collapse
|
60
|
The nuclear receptor NOR-1 modulates redox homeostasis in human vascular smooth muscle cells. J Mol Cell Cardiol 2018; 122:23-33. [PMID: 30096407 DOI: 10.1016/j.yjmcc.2018.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 08/01/2018] [Accepted: 08/05/2018] [Indexed: 12/13/2022]
Abstract
The nuclear receptor NOR-1 (Neuron-derived Orphan Receptor-1) has recently been involved in vascular remodeling and coronary artery disease, however, to date, only a few NOR-1 target genes have been described. We aimed to identify genes regulated by NOR-1 in human vascular smooth muscle cells (VSMC). Lentiviral overexpression of NOR-1 increases reactive oxygen species (ROS) in human VSMC. In accordance, NOR-1 strongly increased NADPH oxidase NOX1 mRNA and protein levels, while NOR-1 silencing significantly reduced NOX1 expression. Luciferase reporter, site-directed mutagenesis and EMSA studies identified two nerve growth factor-induced clone B (NGFI-B)-response elements (NBREs) in NOX1 promoter as essential elements for NOR-1 responsiveness. NOR-1 and NOX1 were co-expressed by VSMC in human atherosclerotic lesions, and NOX1 knockdown counteracted the increased ROS production and cell migration induced by NOR-1 overexpression. NOR-1 also modulated the expression of other enzymes involved in cellular redox status, in particular, upregulated superoxide dismutase-1 (SOD1) and SOD3 while downregulated SOD2 and NOX4. NOR-1 induced SOD1 and SOD3 transcriptional activity and participated in the modulation of SOD3 by inflammatory stimuli. By contrast, NOR-1 impaired SOD2 transcription antagonizing NFκB signaling. These results indicate that NOR-1 induces NOX1 in human VSMC and participates in the complex gene networks regulating oxidative stress and redox homeostasis in the vasculature.
Collapse
|
61
|
Tel-Karthaus N, Kers-Rebel ED, Looman MW, Ichinose H, de Vries CJ, Ansems M. Nuclear Receptor Nur77 Deficiency Alters Dendritic Cell Function. Front Immunol 2018; 9:1797. [PMID: 30123220 PMCID: PMC6085422 DOI: 10.3389/fimmu.2018.01797] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/20/2018] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) are the professional antigen-presenting cells of the immune system. Proper function of DCs is crucial to elicit an effective immune response against pathogens and to induce antitumor immunity. Different members of the nuclear receptor (NR) family of transcription factors have been reported to affect proper function of immune cells. Nur77 is a member of the NR4A subfamily of orphan NRs that is expressed and has a function within the immune system. We now show that Nur77 is expressed in different murine DCs subsets in vitro and ex vivo, in human monocyte-derived DCs (moDCs) and in freshly isolated human BDCA1+ DCs, but its expression is dispensable for DC development in the spleen and lymph nodes. We show, by siRNA-mediated knockdown of Nur77 in human moDCs and by using Nur77-/- murine DCs, that Nur77-deficient DCs have enhanced inflammatory responses leading to increased T cell proliferation. Treatment of human moDCs with 6-mercaptopurine, an activator of Nur77, leads to diminished DC activation resulting in an impaired capacity to induce IFNγ production by allogeneic T cells. Altogether, our data show a yet unexplored role for Nur77 in modifying the activation status of murine and human DCs. Ultimately, targeting Nur77 may prove to be efficacious in boosting or diminishing the activation status of DCs and may lead to the development of improved DC-based immunotherapies in, respectively, cancer treatment or treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Nina Tel-Karthaus
- Department of Radiation Oncology, Radiotherapy & OncoImmunology Laboratory, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Esther D Kers-Rebel
- Department of Radiation Oncology, Radiotherapy & OncoImmunology Laboratory, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Maaike W Looman
- Department of Radiation Oncology, Radiotherapy & OncoImmunology Laboratory, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hiroshi Ichinose
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Carlie J de Vries
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Marleen Ansems
- Department of Radiation Oncology, Radiotherapy & OncoImmunology Laboratory, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
62
|
Wang D, Uhrin P, Mocan A, Waltenberger B, Breuss JM, Tewari D, Mihaly-Bison J, Huminiecki Ł, Starzyński RR, Tzvetkov NT, Horbańczuk J, Atanasov AG. Vascular smooth muscle cell proliferation as a therapeutic target. Part 1: molecular targets and pathways. Biotechnol Adv 2018; 36:1586-1607. [PMID: 29684502 DOI: 10.1016/j.biotechadv.2018.04.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/15/2018] [Accepted: 04/18/2018] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases are a major cause of human death worldwide. Excessive proliferation of vascular smooth muscle cells contributes to the etiology of such diseases, including atherosclerosis, restenosis, and pulmonary hypertension. The control of vascular cell proliferation is complex and encompasses interactions of many regulatory molecules and signaling pathways. Herein, we recapitulated the importance of signaling cascades relevant for the regulation of vascular cell proliferation. Detailed understanding of the mechanism underlying this process is essential for the identification of new lead compounds (e.g., natural products) for vascular therapies.
Collapse
Affiliation(s)
- Dongdong Wang
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria; Institute of Clinical Chemistry, University Hospital Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Andrei Mocan
- Department of Pharmaceutical Botany, "Iuliu Hațieganu" University of Medicine and Pharmacy, Strada Gheorghe Marinescu 23, 400337 Cluj-Napoca, Romania; Institute for Life Sciences, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, Calea Mănăştur 3-5, 400372 Cluj-Napoca, Romania
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Johannes M Breuss
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria
| | - Devesh Tewari
- Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Bhimtal, 263136 Nainital, Uttarakhand, India
| | - Judit Mihaly-Bison
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria
| | - Łukasz Huminiecki
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Rafał R Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Nikolay T Tzvetkov
- Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; NTZ Lab Ltd., Krasno Selo 198, 1618 Sofia, Bulgaria
| | - Jarosław Horbańczuk
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Atanas G Atanasov
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria.
| |
Collapse
|
63
|
Lappas M. Effect of spontaneous term labour on the expression of the NR4A receptors nuclear receptor related 1 protein (Nurr1), neuron-derived clone 77 (Nur77) and neuron-derived orphan receptor 1 (NOR1) in human fetal membranes and myometrium. Reprod Fertil Dev 2018; 28:893-906. [PMID: 25408954 DOI: 10.1071/rd14315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 10/19/2014] [Indexed: 12/27/2022] Open
Abstract
Inflammation has been implicated in the mechanisms responsible for human labour. Emerging evidence indicates that nuclear receptor subfamily 4A (NR4A) receptors regulate the transcription of genes involved in inflammation. The aim of the present study was to determine the effect of spontaneous term labour, Toll-like receptor (TLR) ligands and nucleotide-binding oligomerisation domain-containing (NOD) ligands on the expression of nuclear receptor related 1 protein (Nurr1), neuron-derived clone 77 (Nur77) and neuron-derived orphan receptor 1 (NOR1) in human fetal membranes and myometrium. Human fetal membranes and myometrium were collected from term non-labouring women and women after spontaneous labour onset. Tissue explants were used to determine the effect of the bacterial products lipopolysaccharide (LPS; TLR4 ligand), flagellin (TLR5 ligand), fibroblast-stimulating lipopeptide (FSL-1) (TLR2 ligand), γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP) (NOD1 ligand) or minimal peptidoglycan muramyl dipeptide (MDP; NOD2 ligand) on Nurr1, Nur77 and NOR1 expression. Term labour was associated with significantly higher Nurr1 and Nur77, but not NOR1, expression in fetal membranes and myometrium. LPS and MDP increased Nurr1, Nur77 and NOR in fetal membranes; flagellin increased Nurr1 in fetal membranes and the myometrium, as well as NOR1 in the myometrium; and FSL-1 increased Nurr1 expression in fetal membranes. In summary, human labour and bacterial products increase Nurr1, Nur77 and/or NOR1 expression in human fetal membranes and myometrium. This increase in NR4A receptors may contribute to the expression of proinflammatory and pro-labour genes associated with fetal membrane rupture and myometrial contractions.
Collapse
Affiliation(s)
- Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Vic. 3010, Australia
| |
Collapse
|
64
|
Ruiter MS, Pesce M. Mechanotransduction in Coronary Vein Graft Disease. Front Cardiovasc Med 2018; 5:20. [PMID: 29594150 PMCID: PMC5861212 DOI: 10.3389/fcvm.2018.00020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/22/2018] [Indexed: 12/19/2022] Open
Abstract
Autologous saphenous veins are the most commonly used conduits in revascularization of the ischemic heart by coronary artery bypass graft surgery, but are subject to vein graft failure. The current mini review aims to provide an overview of the role of mechanotransduction signalling underlying vein graft failure to further our understanding of the disease progression and to improve future clinical treatment. Firstly, limitation of damage during vein harvest and engraftment can improve outcome. In addition, cell cycle inhibition, stimulation of Nur77 and external grafting could form interesting therapeutic options. Moreover, the Hippo pathway, with the YAP/TAZ complex as the main effector, is emerging as an important node controlling conversion of mechanical signals into cellular responses. This includes endothelial cell inflammation, smooth muscle cell proliferation/migration, and monocyte attachment/infiltration. The combined effects of expression levels and nuclear/cytoplasmic translocation make YAP/TAZ interesting novel targets in the prevention and treatment of vein graft disease. Pharmacological, molecular and/or mechanical conditioning of saphenous vein segments between harvest and grafting may potentiate targeted and specific treatment to improve long-term outcome.
Collapse
Affiliation(s)
- Matthijs Steven Ruiter
- Cardiovascular Tissue Engineering Unit, Centro Cardiologico Monzino (IRCCS), Milan, Italy
| | - Maurizio Pesce
- Cardiovascular Tissue Engineering Unit, Centro Cardiologico Monzino (IRCCS), Milan, Italy
| |
Collapse
|
65
|
Wang LM, Zhang Y, Li X, Zhang ML, Zhu L, Zhang GX, Xu YM. Nr4a1 plays a crucial modulatory role in Th1/Th17 cell responses and CNS autoimmunity. Brain Behav Immun 2018; 68:44-55. [PMID: 28962999 DOI: 10.1016/j.bbi.2017.09.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/25/2017] [Accepted: 09/25/2017] [Indexed: 12/12/2022] Open
Abstract
Nuclear receptor4 group A1 (Nr4a1), an orphan nuclear receptor, is down-regulated in peripheral blood mononuclear cells (MNCs) of individuals with multiple sclerosis (MS), and Nr4a1 deficiency results in severe experimental autoimmune encephalomyelitis (EAE), an animal model of MS, caused by increased macrophage infiltration into the central nervous system (CNS). However, the role of Nr4a1 in macrophage phenotype and T cell responses remains poorly understood. In the present study we show that macrophages/microglia of Nr4a1-/- mice, which exhibited earlier onset and more severe clinical EAE, were polarized to an enhanced type 1 (M1) phenotype and produced higher levels of IL-12 and TNF-α than wild type mice. Significantly increased numbers of CD4+ T cells and frequency of CD4+IFN-γ+ and CD4+IL-17+ T cells were observed in the CNS and spleen of Nr4a1-/- mice, with decreased percentages of apoptosis in CD4+ T cells. The percentages of CD4+Foxp3+ Treg cells in the CNS of Nr4a1-/- mice were also reduced. Furthermore, purified CD4+ T cells from naïve Nr4a1-/- mice exhibited enhanced Th1 and Th17 differentiation capacity, and MOG-reactive Th17 cells from Nr4a1-/- mice adoptively transferred more severe EAE in recipient mice. Our results, for the first time, demonstrate that Nr4a1 not only induces Type 2 macrophages/microglia phenotype, but is also a critical inhibitory molecule for Th1/Th17 cell differentiation. This finding indicates that Nr4a1-related molecule(s) may have therapeutic potential in MS and likely other autoimmune disorders.
Collapse
Affiliation(s)
- Li-Mei Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Yuan Zhang
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shanxi Normal University, Xi'an, China
| | - Xing Li
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, College of Life Sciences, Shanxi Normal University, Xi'an, China
| | - Ming-Liang Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
66
|
Tsai SY, Catts VS, Fullerton JM, Corley SM, Fillman SG, Weickert CS. Nuclear Receptors and Neuroinflammation in Schizophrenia. MOLECULAR NEUROPSYCHIATRY 2018; 3:181-191. [PMID: 29888229 DOI: 10.1159/000485565] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/21/2017] [Indexed: 01/23/2023]
Abstract
Introduction Several nuclear receptor family members have been associated with schizophrenia and inflammation. Vitamins A and D exert anti-inflammatory actions, but their receptors (mainly nuclear receptors) have not been extensively studied in either schizophrenia brains or in association with neuroinflammation. We examined the expression of vitamin A (RARs and RXRs) and vitamin D and protein disulphide-isomerase A3 (PDIA3) receptors, as well as nuclear orphan receptors (NR4As), in the context of elevated cytokine expression in the dorsolateral prefrontal cortex (DLPFC). Methods mRNA levels of nuclear receptors were measured in DLPFC tissues via RT-qPCR. ANCOVAs comparing high inflammation schizophrenia, low inflammation schizophrenia and low inflammation control groups were performed. Results RARG, RXRB, NR4A1 and NR4A3 transcripts showed significant differential expression across the three groups (ANCOVA p = 0.02-0.001). Post hoc testing revealed significant reductions in RARG expression in schizophrenia with low inflammation compared to schizophrenia with high inflammation and to controls, and RXRB mRNA was significantly reduced in schizophrenia with low inflammation compared to controls. NR4A1 and NR4A3 mRNAs were decreased in schizophrenia with high inflammation compared to schizophrenia with low inflammation, with NR4A1 also significantly different to controls. Conclusion In schizophrenia, changes in nuclear receptor mRNA levels involved with mediating actions of vitamin A derivatives vary according to the inflammatory state of brains.
Collapse
Affiliation(s)
- Shan-Yuan Tsai
- Schizophrenia Research Institute, Sydney, New South Wales, Australia.,Neuroscience Research Australia, Sydney, New South Wales, Australia.,School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Vibeke S Catts
- Schizophrenia Research Institute, Sydney, New South Wales, Australia.,Neuroscience Research Australia, Sydney, New South Wales, Australia.,School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Janice M Fullerton
- Schizophrenia Research Institute, Sydney, New South Wales, Australia.,Neuroscience Research Australia, Sydney, New South Wales, Australia.,School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Susan M Corley
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Stuart G Fillman
- Schizophrenia Research Institute, Sydney, New South Wales, Australia.,Neuroscience Research Australia, Sydney, New South Wales, Australia.,School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney, New South Wales, Australia.,Neuroscience Research Australia, Sydney, New South Wales, Australia.,School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
67
|
Aging: a portrait from gene expression profile in blood cells. Aging (Albany NY) 2017; 8:1802-21. [PMID: 27545843 PMCID: PMC5032697 DOI: 10.18632/aging.101016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 08/07/2016] [Indexed: 01/27/2023]
Abstract
The availability of reliable biomarkers of aging is important not only to monitor the effect of interventions and predict the timing of pathologies associated with aging but also to understand the mechanisms and devise appropriate countermeasures. Blood cells provide an easily available tissue and gene expression profiles from whole blood samples appear to mirror disease states and some aspects of the aging process itself. We report here a microarray analysis of whole blood samples from two cohorts of healthy adult and elderly subjects, aged 43±3 and 68±4 years, respectively, to monitor gene expression changes in the initial phase of the senescence process. A number of significant changes were found in the elderly compared to the adult group, including decreased levels of transcripts coding for components of the mitochondrial respiratory chain, which correlate with a parallel decline in the maximum rate of oxygen consumption (VO2max), as monitored in the same subjects. In addition, blood cells show age-related changes in the expression of several markers of immunosenescence, inflammation and oxidative stress. These findings support the notion that the immune system has a major role in tissue homeostasis and repair, which appears to be impaired since early stages of the aging process.
Collapse
|
68
|
Trahtemberg U, Mevorach D. Apoptotic Cells Induced Signaling for Immune Homeostasis in Macrophages and Dendritic Cells. Front Immunol 2017; 8:1356. [PMID: 29118755 PMCID: PMC5661053 DOI: 10.3389/fimmu.2017.01356] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 10/03/2017] [Indexed: 12/24/2022] Open
Abstract
Inefficient and abnormal clearance of apoptotic cells (efferocytosis) contributes to systemic autoimmune disease in humans and mice, and inefficient chromosomal DNA degradation by DNAse II leads to systemic polyarthritis and a cytokine storm. By contrast, efficient clearance allows immune homeostasis, generally leads to a non-inflammatory state for both macrophages and dendritic cells (DCs), and contributes to maintenance of peripheral tolerance. As many as 3 × 108 cells undergo apoptosis every hour in our bodies, and one of the primary “eat me” signals expressed by apoptotic cells is phosphatidylserine (PtdSer). Apoptotic cells themselves are major contributors to the “anti-inflammatory” nature of the engulfment process, some by secreting thrombospondin-1 (TSP-1) or adenosine monophosphate and possibly other immune modulating “calm-down” signals that interact with macrophages and DCs. Apoptotic cells also produce “find me” and “tolerate me” signals to attract and immune modulate macrophages and DCs that express specific receptors for some of these signals. Neither macrophages nor DCs are uniform, and each cell type may variably express membrane proteins that function as receptors for PtdSer or for opsonins like complement or opsonins that bind to PtdSer, such as protein S and growth arrest-specific 6. Macrophages and DCs also express scavenger receptors, CD36, and integrins that function via bridging molecules such as TSP-1 or milk fat globule-EGF factor 8 protein and that differentially engage in various multi-ligand interactions between apoptotic cells and phagocytes. In this review, we describe the anti-inflammatory and pro-homeostatic nature of apoptotic cell interaction with the immune system. We do not review some forms of immunogenic cell death. We summarize the known apoptotic cell signaling events in macrophages and DCs that are related to toll-like receptors, nuclear factor kappa B, inflammasome, the lipid-activated nuclear receptors, Tyro3, Axl, and Mertk receptors, as well as induction of signal transducer and activator of transcription 1 and suppressor of cytokine signaling that lead to immune system silencing and DC tolerance. These properties of apoptotic cells are the mechanisms that enable their successful use as therapeutic modalities in mice and humans in various autoimmune diseases, organ transplantation, graft-versus-host disease, and sepsis.
Collapse
Affiliation(s)
- Uriel Trahtemberg
- General Intensive Care Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Dror Mevorach
- Rheumatology Research Center, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
69
|
Qing H, Jones KL, Heywood EB, Lu H, Daugherty A, Bruemmer D. Deletion of the NR4A nuclear receptor NOR1 in hematopoietic stem cells reduces inflammation but not abdominal aortic aneurysm formation. BMC Cardiovasc Disord 2017; 17:271. [PMID: 29047330 PMCID: PMC5648424 DOI: 10.1186/s12872-017-0701-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/10/2017] [Indexed: 01/16/2023] Open
Abstract
Background The NR4A3 orphan nuclear hormone receptor, NOR1, functions as a constitutively active transcription factor to regulate inflammation, proliferation, and cell survival during pathological vascular remodeling. Inflammatory processes represent key mechanisms leading to abdominal aortic aneurysm (AAA) formation. However, a role of NOR1 in AAA formation has not been investigated previously. Methods Inflammatory gene expression was analyzed in bone marrow-derived macrophages isolated from NOR1-deficient mice. Low-density lipoprotein receptor-deficient (LDLr−/−) mice were irradiated and reconstituted with hematopoietic stem cells obtained from NOR1−/− or wild-type littermate mice. Animals were infused with angiotensin II and fed a diet enriched in saturated fat to induce AAA formation. Quantification of AAA formation was performed by ultrasound and ex vivo measurements. Results Among 184 inflammatory genes that were analyzed, 36 genes were differentially regulated in LPS-treated NOR1-deficient macrophages. Albeit this difference in gene regulation, NOR1-deficiency in hematopoietic stem cells did not affect development of AAA formation in bone marrow-derived stem cell transplanted LDLr-deficient mice. Conclusion NOR1 deletion induced differential inflammatory gene transcription in macrophages but did not influence AAA formation in mice. Electronic supplementary material The online version of this article (10.1186/s12872-017-0701-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hua Qing
- Department of Medicine, Division of Cardiology, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.,Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA.,Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Karrie L Jones
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Elizabeth B Heywood
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Hong Lu
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, 40536, USA.,Department of Physiology, University of Kentucky, Lexington, KY, 40536, USA
| | - Alan Daugherty
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, 40536, USA.,Department of Physiology, University of Kentucky, Lexington, KY, 40536, USA
| | - Dennis Bruemmer
- Department of Medicine, Division of Cardiology, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA. .,Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA. .,Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
70
|
Dehn S, Thorp EB. Myeloid receptor CD36 is required for early phagocytosis of myocardial infarcts and induction of Nr4a1-dependent mechanisms of cardiac repair. FASEB J 2017; 32:254-264. [PMID: 28860151 DOI: 10.1096/fj.201700450r] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/21/2017] [Indexed: 12/20/2022]
Abstract
Phagocytosis after myocardial infarction (MI) is a prerequisite to cardiac repair. Recruited monocytes clear necrotic cardiomyocytes and differentiate into cardiac macrophages. Some studies have linked apoptotic cell receptors on cardiac macrophages to tissue repair; however, the contribution of precursor monocyte phagocytic receptors, which are the first to interact with the cardiac parenchyma, is unclear. The scavenger receptor cluster of differentiation (CD)36 protein was detected on cardiac Ly6cHI monocytes, and bone marrow-derived Cd36 was essential for both early phagocytosis of dying cardiomyocytes and for smaller infarct sizes in female and male mice after permanent coronary ligation. Cd36 deficiency led to reduced expression of phagocytosis receptor Mertk and nuclear receptor Nr4a1 in cardiac macrophages, the latter previously shown to be required for phagocyte survival. Nr4a1 was required for phagocytosis-induced Mertk expression, and Nr4a1 protein directly bound to Mertk gene regulatory elements. To test the overall contribution of the Cd36-Mertk axis, MI was induced in Cd36-/- Mertk-/- double-knockout mice and led to increases in myocardial rupture. These data implicate monocyte CD36 in the mitigation of early infarct size and transition to Mertk-dependent macrophage function. Increased myocardial rupture in the absence of both Cd36 and Mertk underscore the physiologic significance of phagocytosis during tissue injury.-Dehn, S., Thorp, E. B. Myeloid receptor CD36 is required for early phagocytosis of myocardial infarcts and induction of Nr4a1-dependent mechanisms of cardiac repair.
Collapse
Affiliation(s)
- Shirley Dehn
- Department of Pathology and.,Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Edward B Thorp
- Department of Pathology and .,Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
71
|
Martí-Pàmies I, Cañes L, Alonso J, Rodríguez C, Martínez-González J. The nuclear receptor NOR-1/NR4A3 regulates the multifunctional glycoprotein vitronectin in human vascular smooth muscle cells. FASEB J 2017; 31:4588-4599. [PMID: 28666984 DOI: 10.1096/fj.201700136rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 06/19/2017] [Indexed: 01/04/2023]
Abstract
The nuclear receptor NOR-1 (NR4A3) has recently been involved in the regulation of extracellular matrix (ECM) proteins associated with neointimal thickening and the vascular control of hemostasis. We sought to find as-yet unidentified NOR-1 target genes in human vascular smooth muscle cells (VSMCs). An in silico analysis identified putative NOR-1 response elements in the proximal promoter region of several genes encoding for ECM proteins, including vitronectin (VTN). Lentiviral overexpression of NOR-1 strongly increased VTN mRNA and protein levels, whereas NOR-1 silencing significantly reduced VTN expression. Deletion and site-directed mutagenesis studies, as well as EMSA and chromatin immunoprecipitation, identified the NBRE(-202/-195) site in the VTN promoter as an essential element for NOR-1 responsiveness. Furthermore, NOR-1 and VTN colocalized in VSMCs in human atherosclerotic lesions. VTN levels were increased in cell supernatants from VSMCs that overexpress NOR-1. Cell supernatants from VSMCs overexpressing NOR-1 induced cell migration to a greater extent than supernatants from control cells, and this effect was attenuated when cell supernatants were preincubated with anti-VTN blocking antibodies or VTN was silenced in supernatant-generating cells. These results indicate that VTN is a target of NOR-1 and suggest that this multifunctional glycoprotein may participate in vascular responses mediated by this nuclear receptor.-Martí-Pàmies, I., Cañes, L., Alonso, J., Rodríguez, C., Martínez-González, J. The nuclear receptor NOR-1/NR4A3 regulates the multifunctional glycoprotein vitronectin in human vascular smooth muscle cells.
Collapse
Affiliation(s)
- Ingrid Martí-Pàmies
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Sant Pau Biomedical Research Institute, Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Sant Pau Biomedical Research Institute, Barcelona, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Sant Pau Biomedical Research Institute, Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain
| | - Cristina Rodríguez
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain.,Institut Català de Ciències Cardiovasculars (ICCC), Sant Pau Biomedical Research Institute, Barcelona, Spain
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Sant Pau Biomedical Research Institute, Barcelona, Spain; .,Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Cardiovasculares, Madrid, Spain
| |
Collapse
|
72
|
Li XM, Wang JR, Shen T, Gao SS, He XS, Li JN, Yang TY, Zhang S, Gan WJ, Li JM, Wu H. Nur77 deficiency in mice accelerates tumor invasion and metastasis by facilitating TNFα secretion and lowering CSF-1R expression. PLoS One 2017; 12:e0171347. [PMID: 28170411 PMCID: PMC5295676 DOI: 10.1371/journal.pone.0171347] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/19/2017] [Indexed: 12/26/2022] Open
Abstract
Nur77, an orphan member of the nuclear receptor superfamily, plays critical roles in inflammation and immunity. However, the role of Nur77 in tumor microenvironment remains elusive. Results showed that deletion of Nur77 strikingly enhanced tumor metastasis compared to WT mice. Additionally, compared to the conditioned media derived from Nur77+/+ peritoneal macrophages (CM1), the conditioned media derived from Nur77-/- peritoneal macrophages (CM2) significantly promoted the EMT of cancer cells, and greatly enhanced the migratory and invasive abilities of cancer cells. Moreover, studies using TNF-α blocking antibody demonstrated that pro-inflammatory cytokine TNF-α was indispensable in supporting CM2-induced EMT to drive cancer cells migration and invasion. Furthermore, we found that Nur77 promoted the expression of CSF-1R, a novel downstream target gene of Nur77, and subsequently enhanced the migration of inflammatory cells. Notably, infiltration of inflammatory cells in the tumors of Nur77-/- mice was markedly abrogated compared to Nur77+/+ mice. Collectively, these results revealed that host Nur77 expression was pivotal in antitumor immune response, and in inhibiting tumor metastasis.
Collapse
Affiliation(s)
- Xiu-Ming Li
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Jing-Ru Wang
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Tong Shen
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Shang-Shang Gao
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Xiao-Shun He
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiang-Nan Li
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Tian-Yu Yang
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Shen Zhang
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Wen-Juan Gan
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian-Ming Li
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- * E-mail: (HW); (JML)
| | - Hua Wu
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
- * E-mail: (HW); (JML)
| |
Collapse
|
73
|
McEvoy C, de Gaetano M, Giffney HE, Bahar B, Cummins EP, Brennan EP, Barry M, Belton O, Godson CG, Murphy EP, Crean D. NR4A Receptors Differentially Regulate NF-κB Signaling in Myeloid Cells. Front Immunol 2017; 8:7. [PMID: 28167941 PMCID: PMC5256039 DOI: 10.3389/fimmu.2017.00007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/03/2017] [Indexed: 12/19/2022] Open
Abstract
Dysregulation of inflammatory responses is a hallmark of multiple diseases such as atherosclerosis and rheumatoid arthritis. As constitutively active transcription factors, NR4A nuclear receptors function to control the magnitude of inflammatory responses and in chronic inflammatory disease can be protective or pathogenic. Within this study, we demonstrate that TLR4 stimulation using the endotoxin lipopolysaccharide (LPS) rapidly enhances NR4A1–3 expression in human and murine, primary and immortalized myeloid cells with concomitant gene transcription and protein secretion of MIP-3α, a central chemokine implicated in numerous pathologies. Deficiency of NR4A2 and NR4A3 in human and murine myeloid cells reveals that both receptors function as positive regulators of enhanced MIP-3α expression. In contrast, within the same cell types and conditions, altered NR4A activity leads to suppression of LPS-induced MCP-1 gene and protein expression. An equivalent pattern of inflammatory gene regulation is replicated in TNFα-treated myeloid cells. We show that NF-κB is the critical regulator of NR4A1–3, MIP-3α, and MCP-1 during TLR4 stimulation in myeloid cells and highlight a parallel mechanism whereby NR4A activity can repress or enhance NF-κB target gene expression simultaneously. Mechanistic insight reveals that NR4A2 does not require DNA-binding capacity in order to enhance or repress NF-κB target gene expression simultaneously and establishes a role for NF-κB family member Relb as a novel NR4A target gene involved in the positive regulation of MIP-3α. Thus, our data reveal a dynamic role for NR4A receptors concurrently enhancing and repressing NF-κB activity in myeloid cells leading to altered transcription of key inflammatory mediators.
Collapse
Affiliation(s)
- Caitriona McEvoy
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Monica de Gaetano
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Hugh E Giffney
- Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Bojlul Bahar
- International Institute of Nutritional Sciences and Applied Food Safety Studies, University of Central Lancashire , Preston , UK
| | - Eoin P Cummins
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Eoin P Brennan
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Mary Barry
- St. Vincent's University Hospital , Dublin , Ireland
| | - Orina Belton
- Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin , Dublin , Ireland
| | - Catherine G Godson
- School of Medicine, University College Dublin, Dublin, Ireland; Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Evelyn P Murphy
- Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Daniel Crean
- Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; Diabetes and Complications Research Centre, Conway Institute for Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland; School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
74
|
Bianchi E, Ruberti S, Rontauroli S, Guglielmelli P, Salati S, Rossi C, Zini R, Tagliafico E, Vannucchi AM, Manfredini R. Role of miR-34a-5p in Hematopoietic Progenitor Cells Proliferation and Fate Decision: Novel Insights into the Pathogenesis of Primary Myelofibrosis. Int J Mol Sci 2017; 18:ijms18010145. [PMID: 28098757 PMCID: PMC5297778 DOI: 10.3390/ijms18010145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/05/2017] [Accepted: 01/06/2017] [Indexed: 12/24/2022] Open
Abstract
Primary Myelofibrosis (PMF) is a chronic Philadelphia-negative myeloproliferative neoplasm characterized by a skewed megakaryopoiesis and an overproduction of proinflammatory and profibrotic mediators that lead to the development of bone marrow (BM) fibrosis. Since we recently uncovered the upregulation of miR-34a-5p in PMF CD34+ hematopoietic progenitor cells (HPCs), in order to elucidate its role in PMF pathogenesis here we unravelled the effects of miR-34a-5p overexpression in HPCs. We showed that enforced expression of miR-34a-5p partially constrains proliferation and favours the megakaryocyte and monocyte/macrophage commitment of HPCs. Interestingly, we identified lymphoid enhancer-binding factor 1 (LEF1) and nuclear receptor subfamily 4, group A, member 2 (NR4A2) transcripts as miR-34a-5p-targets downregulated after miR-34a-5p overexpression in HPCs as well as in PMF CD34+ cells. Remarkably, the knockdown of NR4A2 in HPCs mimicked the antiproliferative effects of miR-34a-5p overexpression, while the silencing of LEF1 phenocopied the effects of miR-34a-5p overexpression on HPCs lineage choice, by favouring the megakaryocyte and monocyte/macrophage commitment. Collectively our data unravel the role of miR-34a-5p in HPCs fate decision and suggest that the increased expression of miR-34a-5p in PMF HPCs could be important for the skewing of megakaryopoiesis and the production of monocytes, that are key players in BM fibrosis in PMF patients.
Collapse
Affiliation(s)
- Elisa Bianchi
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy.
| | - Samantha Ruberti
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy.
| | - Sebastiano Rontauroli
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy.
| | - Paola Guglielmelli
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, AOU Careggi and Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| | - Simona Salati
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy.
| | - Chiara Rossi
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy.
| | - Roberta Zini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy.
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, 41125 Modena, Italy.
| | - Alessandro Maria Vannucchi
- CRIMM, Center for Research and Innovation for Myeloproliferative Neoplasms, AOU Careggi and Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| | - Rossella Manfredini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy.
| |
Collapse
|
75
|
Rodríguez-Calvo R, Tajes M, Vázquez-Carrera M. The NR4A subfamily of nuclear receptors: potential new therapeutic targets for the treatment of inflammatory diseases. Expert Opin Ther Targets 2017; 21:291-304. [PMID: 28055275 DOI: 10.1080/14728222.2017.1279146] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Prolonged inflammatory response contributes to the pathogenesis of chronic disease-related disturbances. Among nuclear receptors (NRs), the orphan NR4A subfamily, which includes Nur77 (NR4A1), Nurr1 (NR4A2) and NOR1 (NR4A3), has recently emerged as a therapeutic target for the treatment of inflammation. Areas covered: This review focuses on the capacity of NR4A receptors to counter-regulate the development of the inflammatory response, with a special focus on the molecular transrepression mechanisms. Expert opinion: Recent studies have highlighted the role of NR4A receptors as significant regulators of the inflammatory response. NR4A receptors are rapidly induced by inflammatory stimuli, thus suggesting that they are required for the initiation of inflammation. Nevertheless, NR4A anti-inflammatory properties indicate that this acute regulation could be a protective reaction aimed at resolving inflammation in the later stages. Therefore, NR4A receptors are involved in a negative feedback mechanism to maintain the inflammatory balance. However, the underlying mechanisms are not entirely clear. Only a small number of NR4A-target genes have been identified, and the transcriptional repression mechanisms are only beginning to emerge. Despite further research is needed to fully understand the role of NR4A receptors in inflammation, these NRs should be considered as targets for new therapeutic approaches to inflammatory diseases.
Collapse
Affiliation(s)
- Ricardo Rodríguez-Calvo
- a Vascular Medicine and Metabolism Unit, Research Unit on Lipids and Atherosclerosis, Sant Joan University Hospital, Pere Virgili Health Research Institute (IISPV) and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)-Instituto de Salud Carlos III, Faculty of Medicine and Health Sciences , Rovira i Virgili University , Reus , Spain
| | - Marta Tajes
- b Heart Diseases Biomedical Research Group, Inflammatory and Cardiovascular Disorders Program , Hospital del Mar Medical Research Institute (IMIM), Parc de Salut Mar , Barcelona , Spain
| | - Manuel Vázquez-Carrera
- c Department of Pharmacology, Toxicology and Therapeutic Chemistry, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Institut de Recerca Pediàtrica-Hospital Sant Joan de Déu, and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM)-Instituto de Salud Carlos III, Faculty of Pharmacy, Diagonal 643 , University of Barcelona , Barcelona , Spain
| |
Collapse
|
76
|
Pérez-Matute P, Iñiguez M, Recio-Fernández E, Oteo JA. Deciphering the molecular mechanisms involved in HIV-associated lipoatrophy by transcriptomics: a pilot study. J Physiol Biochem 2017; 73:431-443. [PMID: 28074419 DOI: 10.1007/s13105-016-0547-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 12/26/2016] [Indexed: 01/11/2023]
Abstract
HIV-associated lipoatrophy (LA) has considerable implications for risk of metabolic diseases, quality of life, and adherence to treatments. Although it has decreased in high-income countries, it is still very common in resource-limited countries. Understanding the pathophysiological mechanisms of LA can open the possibility to explore new ways to treat or prevent this condition. To identify new markers for an accurate and quick diagnosis will be also of interest. Thus, we aimed to examine functional classes of genes implicated in LA and to identify potential new markers for an accurate/quick diagnosis of LA and future complications. Eighteen participants were recruited: seven healthy volunteers, five non-LA-HIV patients, and six LA-HIV subjects. Clinical lipoatrophy was considered when changes in fat volume in the cheeks next to the nose, lateral aspect of the face, legs, arms, and buttocks were observed by the physicians. mRNA was isolated from peripheral blood mononuclear cells (PBMCs) to perform a transcriptomic and Gene Ontology analysis. To confirm RNA sequencing results, qPCRs were developed. A total of 55 genes were differentially expressed between LA and non-LA patients. Thirty-seven genes were overexpressed, whereas 18 genes were repressed. Functional analysis showed that overexpressed genes were involved in lymphocyte/neutrophil activation, inflammation, and atherogenesis. Several lymphoma markers and members of the lipocalin and aquaporin families were also found more expressed in LA patients. In contrast, most of the genes found less expressed in LA subjects were involved in angiogenesis and protection against myocardial infarction. Our results demonstrated a distinct transcriptomic signature in PBMCs of LA patients in comparison with non-LA-HIV subjects and, therefore, provided novel insights to the pathogenesis of HIV-associated lipoatrophy. Our study also highlights the potential usage of some of these genes as early markers of future complications.
Collapse
Affiliation(s)
- Patricia Pérez-Matute
- HIV and Associated Metabolic Alterations Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR)-Hospital San Pedro, Piqueras 98, 26006, Logroño, Spain.
| | - María Iñiguez
- Genomics Core Facility, Center for Biomedical Research of La Rioja (CIBIR), Piqueras 98, 26006, Logroño, Spain
| | - Emma Recio-Fernández
- HIV and Associated Metabolic Alterations Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR)-Hospital San Pedro, Piqueras 98, 26006, Logroño, Spain
| | - José-Antonio Oteo
- HIV and Associated Metabolic Alterations Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR)-Hospital San Pedro, Piqueras 98, 26006, Logroño, Spain
| |
Collapse
|
77
|
Ruiter MS, Doornbos A, de Waard V, de Winter RJ, Attevelt NJM, Steendam R, de Vries CJM. Long-term effect of stents eluting 6-mercaptopurine in porcine coronary arteries. J Negat Results Biomed 2016; 15:20. [PMID: 27916002 PMCID: PMC5137209 DOI: 10.1186/s12952-016-0063-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 10/15/2016] [Indexed: 11/10/2022] Open
Abstract
Background Drug-eluting stents (DES) have dramatically reduced restenosis rates compared to bare metal stents and are widely used in coronary artery angioplasty. The anti-proliferative nature of the drugs reduces smooth muscle cell (SMC) proliferation effectively, but unfortunately also negatively affects endothelialization of stent struts, necessitating prolonged dual anti-platelet therapy. Cell-type specific therapy may prevent this complication, giving rise to safer stents that do not require additional medication. 6-Mercaptopurine (6-MP) is a drug with demonstrated cell-type specific effects on vascular cells both in vitro and in vivo, inhibiting proliferation of SMCs while promoting survival of endothelial cells. In rabbits, we demonstrated that DES locally releasing 6-MP during 4 weeks reduced in-stent stenosis by inhibiting SMC proliferation and reducing inflammation, without negatively affecting endothelialization of the stent surface. The aim of the present study was to investigate whether 6-MP-eluting stents are similarly effective in preventing stenosis in porcine coronary arteries after 3 months, in order to assess the eligibility for human application. Methods 6-MP-eluting and polymer-only control stents (both n = 7) were implanted in porcine coronary arteries after local balloon injury to assess the effect of 6-MP on vascular lesion formation. Three months after implantation, stented coronary arteries were harvested and analyzed. Results Morphometric analyses revealed that stents were implanted reproducibly and with limited injury to the vessel wall. Unexpectedly, both in-stent stenosis (6-MP: 41.1 ± 10.3 %; control: 29.6 ± 5.9 %) and inflammation (6-MP: 2.14 ± 0.51; control: 1.43 ± 0.45) were similar between the groups after 3 months. Conclusion In conclusion, although 6-MP was previously found to potently inhibit SMC proliferation, reduce inflammation and promote endothelial cell survival, thereby effectively reducing in-stent restenosis in rabbits, stents containing 300 μg 6-MP did not reduce stenosis and inflammation in porcine coronary arteries.
Collapse
Affiliation(s)
- Matthijs S Ruiter
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.,Present address: Unit of Tissue Engineering, Monzino Cardiologic Center, Milan, Italy
| | | | - Vivian de Waard
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands
| | - Robbert J de Winter
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Nico J M Attevelt
- Central Laboratory Animal Research Facility, Faculty of Veterinary Medicine, University of Utrecht, Utrecht, The Netherlands
| | - Rob Steendam
- InnoCore Pharmaceuticals, Groningen, The Netherlands
| | - Carlie J M de Vries
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
78
|
Jiang Y, Zeng Y, Huang X, Qin Y, Luo W, Xiang S, Sooranna SR, Pinhu L. Nur77 attenuates endothelin-1 expression via downregulation of NF-κB and p38 MAPK in A549 cells and in an ARDS rat model. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1023-L1035. [PMID: 27765761 PMCID: PMC5206403 DOI: 10.1152/ajplung.00043.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 10/03/2016] [Indexed: 02/07/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by inflammatory injury to the alveolar and capillary barriers that results in impaired gas exchange and severe acute respiratory failure. Nuclear orphan receptor Nur77 has emerged as a regulator of gene expression in inflammation, and its role in the pathogenesis of ARDS is not clear. The objective of this study is to investigate the potential role of Nur77 and its underlying mechanism in the regulation of endothelin-1 (ET-1) expression in lipopolysaccharide (LPS)-induced A549 cells and an ARDS rat model. We demonstrate that LPS induced Nur77 expression and nuclear export in A549 cells. Overexpression of Nur77 markedly decreased basal and LPS-induced ET-1 expression in A549 cells, whereas knockdown of Nur77 increased the ET-1 expression. LPS-induced phosphorylation and nuclear translocation of NF-κB and p38 MAPK were blocked by Nur77 overexpression and augmented by Nur77 knockdown in A549 cells. In vivo, LPS induced Nur77 expression in lung in ARDS rats. Pharmacological activation of Nur77 by cytosporone B (CsnB) inhibited ET-1 expression in ARDS rats, decreased LPS-induced phosphorylation of NF-κB and p38 MAPK, and relieved lung, liver, and kidney injury. Pharmacological deactivation of Nur77 by 1,1-bis-(3'-indolyl)-1-(p-hydroxyphenyl)methane (DIM-C-pPhOH, C-DIM8) had no effect on ET-1 expression and lung injury. These results indicated that Nur77 decreases ET-1 expression by suppressing NF-κB and p38 MAPK in LPS-stimulated A549 cells in vitro, and, in an LPS-induced ARDS rat model, CsnB reduced ET-1 expression and lung injury in ARDS rats.
Collapse
MESH Headings
- A549 Cells
- Active Transport, Cell Nucleus/drug effects
- Animals
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Disease Models, Animal
- Down-Regulation/drug effects
- Endothelin-1/metabolism
- Kidney/drug effects
- Kidney/pathology
- Lipopolysaccharides/pharmacology
- Liver/drug effects
- Liver/pathology
- Lung/drug effects
- Lung/metabolism
- Male
- NF-kappa B/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/agonists
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Phenylacetates/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Respiratory Distress Syndrome/enzymology
- Respiratory Distress Syndrome/genetics
- Respiratory Distress Syndrome/pathology
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Yujie Jiang
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong Province, China
- Department of Respiratory Medicine
| | - Yi Zeng
- Department of Central Laboratory, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xia Huang
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong Province, China
- Department of Respiratory Medicine
| | - Yueqiu Qin
- Department of Digestive, Youjiang Medical University for Nationalities, Baise, Guangxi, China; Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | | | - Shulin Xiang
- Department of Intensive Care Unit, the People's Hospital of Guangxi, Nanning, Guangxi, China
| | - Suren R Sooranna
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdon; and
| | - Liao Pinhu
- Department of Intensive Care Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| |
Collapse
|
79
|
Brunet A, LeBel M, Egarnes B, Paquet-Bouchard C, Lessard AJ, Brown JP, Gosselin J. NR4A1-dependent Ly6C low monocytes contribute to reducing joint inflammation in arthritic mice through Treg cells. Eur J Immunol 2016; 46:2789-2800. [PMID: 27600773 DOI: 10.1002/eji.201646406] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 08/23/2016] [Accepted: 08/31/2016] [Indexed: 12/24/2022]
Abstract
Monocytes are central to the physiopathology of arthritis, but their roles in progression and resolution of the disease remain to be clarified. Using NR4A1-/- mice, which lack patrolling lymphocyte antigen 6C (Ly6Clow ) monocytes, we found that inflammatory Ly6Chigh monocytes contribute to rapid development of arthritis in a serum transfer-induced arthritis (STIA) model. Our experiments suggest that patrolling monocytes do not promote the initiation and progression of arthritis in mice, as severity of symptoms was amplified in NR4A1-/- mice. Moreover, we show that treatment of arthritic wild type (WT) mice with cytosporone B (Csn-B), a NR4A1-specific agonist, significantly reduces severity of disease. Effects of Csn-B were absent in monocyte-depleted mice treated with clodronate until Ly6Clow monocytes were restored. Adoptive transfer of Ly6Clow monocytes in arthritic NR4A1-/- mice treated with Csn-B reduces joint inflammation, supporting the regulatory role of Ly6Clow subset on disease development. Our results also reveal that administration of Csn-B to arthritic mice enhances levels of circulating CD4+ CD25+ FoxP3+ Treg cells, a process requiring the presence of Ly6Clow monocytes. Together, these data indicate that Ly6Chigh monocytes are involved in the initiation and progression of arthritis and Ly6Clow monocytes contribute to reduce joint inflammation through the mobilization of Treg cells.
Collapse
Affiliation(s)
- Alexandre Brunet
- Laboratory of Innate Immunology, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Manon LeBel
- Laboratory of Innate Immunology, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Benoit Egarnes
- Laboratory of Innate Immunology, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Carine Paquet-Bouchard
- Laboratory of Innate Immunology, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Anne-Julie Lessard
- Laboratory of Innate Immunology, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Jacques P Brown
- Division of Rheumatology, CHU de Québec-Université Laval (CHUL), Infectious and Immune Diseases, Centre de recherche du CHU de Québec-Université Laval (CHUL), Québec, QC, Canada
| | - Jean Gosselin
- Laboratory of Innate Immunology, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Department of Molecular Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
80
|
Jin HS, Kim TS, Jo EK. Emerging roles of orphan nuclear receptors in regulation of innate immunity. Arch Pharm Res 2016; 39:1491-1502. [PMID: 27699647 DOI: 10.1007/s12272-016-0841-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/23/2016] [Indexed: 01/25/2023]
Abstract
Innate immunity constitutes the first line of defense against pathogenic and dangerous insults. However, it is a double-edged sword, as it functions in both clearance of infection and inflammatory damage. It is therefore important that innate immune responses are tightly controlled to prevent harmful excessive inflammation. Nuclear receptors (NRs) are a family of transcription factors that play critical roles in various physiological responses. Orphan NRs are a subset of NRs for which the ligands and functions are unclear. Accumulating evidence has revealed that orphan NRs play essential roles in innate immune responses to prevent pathogenic inflammatory responses and to enhance antimicrobial host defenses. In this review, we describe current knowledge on the roles and mechanisms of orphan NRs in the regulation of innate immune responses. Discovery of new functions of orphan NRs would facilitate development of novel preventive and therapeutic strategies against human inflammatory diseases.
Collapse
Affiliation(s)
- Hyo Sun Jin
- Department of Microbiology, Department of Medical Science, Chungnam National University School of Medicine, 6 Munhwa-dong, Jungku, Daejeon, 301-747, South Korea
| | - Tae Sung Kim
- Department of Microbiology, Department of Medical Science, Chungnam National University School of Medicine, 6 Munhwa-dong, Jungku, Daejeon, 301-747, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Department of Medical Science, Chungnam National University School of Medicine, 6 Munhwa-dong, Jungku, Daejeon, 301-747, South Korea.
| |
Collapse
|
81
|
Alonso J, Galán M, Martí-Pàmies I, Romero JM, Camacho M, Rodríguez C, Martínez-González J. NOR-1/NR4A3 regulates the cellular inhibitor of apoptosis 2 (cIAP2) in vascular cells: role in the survival response to hypoxic stress. Sci Rep 2016; 6:34056. [PMID: 27654514 PMCID: PMC5032021 DOI: 10.1038/srep34056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/07/2016] [Indexed: 12/16/2022] Open
Abstract
Vascular cell survival is compromised under pathological conditions such as abdominal aortic aneurysm (AAA). We have previously shown that the nuclear receptor NOR-1 is involved in the survival response of vascular cells to hypoxia. Here, we identify the anti-apoptotic protein cIAP2 as a downstream effector of NOR-1. NOR-1 and cIAP2 were up-regulated in human AAA samples, colocalizing in vascular smooth muscle cells (VSMC). While NOR-1 silencing reduced cIAP2 expression in vascular cells, lentiviral over-expression of this receptor increased cIAP2 mRNA and protein levels. The transcriptional regulation of the human cIAP2 promoter was analyzed in cells over-expressing NOR-1 by luciferase reporter assays, electrophoretic mobility shift analysis and chromatin immunoprecipitation, identifying a NGFI-B site (NBRE-358/-351) essential for NOR-1 responsiveness. NOR-1 and cIAP2 were up-regulated by hypoxia and by a hypoxia mimetic showing a similar time-dependent pattern. Deletion and site-directed mutagenesis studies show that NOR-1 mediates the hypoxia-induced cIAP2 expression. While NOR-1 over-expression up-regulated cIAP2 and limited VSMC apoptosis induced by hypoxic stress, cIAP2 silencing partially prevented this NOR-1 pro-survival effect. These results indicate that cIAP2 is a target of NOR-1, and suggest that this anti-apoptotic protein is involved in the survival response to hypoxic stress mediated by NOR-1 in vascular cells.
Collapse
Affiliation(s)
- Judith Alonso
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - María Galán
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain.,Laboratorio de Angiología, Biología Vascular e Inflamación y Servicio de Cirugía Vascular, IIB-Sant Pau, c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Ingrid Martí-Pàmies
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - José María Romero
- Laboratorio de Angiología, Biología Vascular e Inflamación y Servicio de Cirugía Vascular, IIB-Sant Pau, c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Mercedes Camacho
- Laboratorio de Angiología, Biología Vascular e Inflamación y Servicio de Cirugía Vascular, IIB-Sant Pau, c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Cristina Rodríguez
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - José Martínez-González
- Centro de Investigación Cardiovascular (CSIC-ICCC), Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), c/Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| |
Collapse
|
82
|
Huang HY, Chang HF, Tsai MJ, Chen JS, Wang MJ. 6-Mercaptopurine attenuates tumor necrosis factor-α production in microglia through Nur77-mediated transrepression and PI3K/Akt/mTOR signaling-mediated translational regulation. J Neuroinflammation 2016; 13:78. [PMID: 27075886 PMCID: PMC4831152 DOI: 10.1186/s12974-016-0543-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 04/07/2016] [Indexed: 02/07/2023] Open
Abstract
Background The pathogenesis of several neurodegenerative diseases often involves the microglial activation and associated inflammatory processes. Activated microglia release pro-inflammatory factors that may be neurotoxic. 6-Mercaptopurine (6-MP) is a well-established immunosuppressive drug. Common understanding of their immunosuppressive properties is largely limited to peripheral immune cells. However, the effect of 6-MP in the central nervous system, especially in microglia in the context of neuroinflammation is, as yet, unclear. Tumor necrosis factor-α (TNF-α) is a key cytokine of the immune system that initiates and promotes neuroinflammation. The present study aimed to investigate the effect of 6-MP on TNF-α production by microglia to discern the molecular mechanisms of this modulation. Methods Lipopolysaccharide (LPS) was used to induce an inflammatory response in cultured primary microglia or murine BV-2 microglial cells. Released TNF-α was measured by enzyme-linked immunosorbent assay (ELISA). Gene expression was determined by real-time reverse transcription polymerase chain reaction (RT-PCR). Signaling molecules were analyzed by western blotting, and activation of NF-κB was measured by ELISA-based DNA binding analysis and luciferase reporter assay. Chromatin immunoprecipitation (ChIP) analysis was performed to examine NF-κB p65 and coactivator p300 enrichments and histone modifications at the endogenous TNF-α promoter. Results Treatment of LPS-activated microglia with 6-MP significantly attenuated TNF-α production. In 6-MP pretreated microglia, LPS-induced MAPK signaling, IκB-α degradation, NF-κB p65 nuclear translocation, and in vitro p65 DNA binding activity were not impaired. However, 6-MP suppressed transactivation activity of NF-κB and TNF-α promoter by inhibiting phosphorylation and acetylation of p65 on Ser276 and Lys310, respectively. ChIP analyses revealed that 6-MP dampened LPS-induced histone H3 acetylation of chromatin surrounding the TNF-α promoter, ultimately leading to a decrease in p65/coactivator-mediated transcription of TNF-α gene. Furthermore, 6-MP enhanced orphan nuclear receptor Nur77 expression. Using RNA interference approach, we further demonstrated that Nur77 upregulation contribute to 6-MP-mediated inhibitory effect on TNF-α production. Additionally, 6-MP also impeded TNF-α mRNA translation through prevention of LPS-activated PI3K/Akt/mTOR signaling cascades. Conclusions These results suggest that 6-MP might have a therapeutic potential in neuroinflammation-related neurodegenerative disorders through downregulation of microglia-mediated inflammatory processes. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0543-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hsin-Yi Huang
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Hui-Fen Chang
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Ming-Jen Tsai
- Department of Emergency Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi, Taiwan
| | - Jhih-Si Chen
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Mei-Jen Wang
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan.
| |
Collapse
|
83
|
Yun B, Lee H, Jayaraja S, Suram S, Murphy RC, Leslie CC. Prostaglandins from Cytosolic Phospholipase A2α/Cyclooxygenase-1 Pathway and Mitogen-activated Protein Kinases Regulate Gene Expression in Candida albicans-infected Macrophages. J Biol Chem 2016; 291:7070-86. [PMID: 26841868 PMCID: PMC4807289 DOI: 10.1074/jbc.m116.714873] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/02/2016] [Indexed: 12/31/2022] Open
Abstract
In Candida albicans-infected resident peritoneal macrophages, activation of group IVA cytosolic phospholipase A2(cPLA2α) by calcium- and mitogen-activated protein kinases triggers the rapid production of prostaglandins I2 and E2 through cyclooxygenase (COX)-1 and regulates gene expression by increasing cAMP. InC. albicans-infected cPLA2α(-/-)or COX-1(-/-)macrophages, expression ofI l10,Nr4a2, and Ptgs2 was lower, and expression ofTnfα was higher, than in wild type macrophages. Expression was reconstituted with 8-bromo-cAMP, the PKA activator 6-benzoyl-cAMP, and agonists for prostaglandin receptors IP, EP2, and EP4 in infected but not uninfected cPLA2α(-/-)or COX-1(-/-)macrophages. InC. albicans-infected cPLA2α(+/+)macrophages, COX-2 expression was blocked by IP, EP2, and EP4 receptor antagonists, indicating a role for both prostaglandin I2 and E2 Activation of ERKs and p38, but not JNKs, by C. albicansacted synergistically with prostaglandins to induce expression of Il10,Nr4a2, and Ptgs2. Tnfα expression required activation of ERKs and p38 but was suppressed by cAMP. Results using cAMP analogues that activate PKA or Epacs suggested that cAMP regulates gene expression through PKA. However, phosphorylation of cAMP-response element-binding protein (CREB), the cAMP-regulated transcription factor involved inIl10,Nr4a2,Ptgs2, andTnfα expression, was not mediated by cAMP/PKA because it was similar inC. albicans-infected wild type and cPLA2α(-/-)or COX-1(-/-)macrophages. CREB phosphorylation was blocked by p38 inhibitors and induced by the p38 activator anisomycin but not by the PKA activator 6-benzoyl-cAMP. Therefore, MAPK activation inC. albicans-infected macrophages plays a dual role by promoting the cPLA2α/prostaglandin/cAMP/PKA pathway and CREB phosphorylation that coordinately regulate immediate early gene expression.
Collapse
MESH Headings
- 8-Bromo Cyclic Adenosine Monophosphate/pharmacology
- Animals
- Candida albicans/physiology
- Cyclic AMP/analogs & derivatives
- Cyclic AMP/metabolism
- Cyclic AMP/pharmacology
- Cyclic AMP Response Element-Binding Protein/genetics
- Cyclic AMP Response Element-Binding Protein/immunology
- Cyclooxygenase 1/deficiency
- Cyclooxygenase 1/genetics
- Cyclooxygenase 1/immunology
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/immunology
- Dinoprostone/biosynthesis
- Epoprostenol/biosynthesis
- Gene Expression Regulation
- Group IV Phospholipases A2/deficiency
- Group IV Phospholipases A2/genetics
- Group IV Phospholipases A2/immunology
- Host-Pathogen Interactions
- Interleukin-10/genetics
- Interleukin-10/immunology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/microbiology
- Membrane Proteins/deficiency
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Knockout
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/immunology
- Mitogen-Activated Protein Kinase 3/genetics
- Mitogen-Activated Protein Kinase 3/immunology
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/immunology
- Primary Cell Culture
- Protein Kinase Inhibitors/pharmacology
- Receptors, Prostaglandin/agonists
- Receptors, Prostaglandin/antagonists & inhibitors
- Receptors, Prostaglandin/genetics
- Receptors, Prostaglandin/immunology
- Signal Transduction
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- p38 Mitogen-Activated Protein Kinases/genetics
- p38 Mitogen-Activated Protein Kinases/immunology
Collapse
Affiliation(s)
- Bogeon Yun
- From the Department of Pediatrics, National Jewish Health, Denver, Colorado 80206 and
| | - HeeJung Lee
- From the Department of Pediatrics, National Jewish Health, Denver, Colorado 80206 and
| | - Sabarirajan Jayaraja
- From the Department of Pediatrics, National Jewish Health, Denver, Colorado 80206 and
| | - Saritha Suram
- From the Department of Pediatrics, National Jewish Health, Denver, Colorado 80206 and
| | | | - Christina C Leslie
- From the Department of Pediatrics, National Jewish Health, Denver, Colorado 80206 and the Departments of Pharmacology and Pathology, University of Colorado Denver, Aurora, Colorado 80045
| |
Collapse
|
84
|
Hamers AAJ, Argmann C, Moerland PD, Koenis DS, Marinković G, Sokolović M, de Vos AF, de Vries CJM, van Tiel CM. Nur77-deficiency in bone marrow-derived macrophages modulates inflammatory responses, extracellular matrix homeostasis, phagocytosis and tolerance. BMC Genomics 2016; 17:162. [PMID: 26932821 PMCID: PMC4774191 DOI: 10.1186/s12864-016-2469-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 02/12/2016] [Indexed: 02/08/2023] Open
Abstract
Background The nuclear orphan receptor Nur77 (NR4A1, TR3, or NGFI-B) has been shown to modulate the inflammatory response of macrophages. To further elucidate the role of Nur77 in macrophage physiology, we compared the transcriptome of bone marrow-derived macrophages (BMM) from wild-type (WT) and Nur77-knockout (KO) mice. Results In line with previous observations, SDF-1α (CXCL12) was among the most upregulated genes in Nur77-deficient BMM and we demonstrated that Nur77 binds directly to the SDF-1α promoter, resulting in inhibition of SDF-1α expression. The cytokine receptor CX3CR1 was strongly downregulated in Nur77-KO BMM, implying involvement of Nur77 in macrophage tolerance. Ingenuity pathway analyses (IPA) to identify canonical pathways regulation and gene set enrichment analyses (GSEA) revealed a potential role for Nur77 in extracellular matrix homeostasis. Nur77-deficiency increased the collagen content of macrophage extracellular matrix through enhanced expression of several collagen subtypes and diminished matrix metalloproteinase (MMP)-9 activity. IPA upstream regulator analyses discerned the small GTPase Rac1 as a novel regulator of Nur77-mediated gene expression. We identified an inhibitory feedback loop with increased Rac1 activity in Nur77-KO BMM, which may explain the augmented phagocytic activity of these cells. Finally, we predict multiple chronic inflammatory diseases to be influenced by macrophage Nur77 expression. GSEA and IPA associated Nur77 to osteoarthritis, chronic obstructive pulmonary disease, rheumatoid arthritis, psoriasis, and allergic airway inflammatory diseases. Conclusions Altogether these data identify Nur77 as a modulator of macrophage function and an interesting target to treat chronic inflammatory disease. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2469-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anouk A J Hamers
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands. .,Present address: Department of Inflammation Biology, La Jolla Institute for Allergy and Immunology, San Diego, USA.
| | - Carmen Argmann
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands. .,Present address: Institute for Genomics and Multiscale Biology Mount Sinai Hospital, New York, USA.
| | - Perry D Moerland
- Bioinformatics Laboratory, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Duco S Koenis
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Goran Marinković
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Milka Sokolović
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands. .,Present address: European Food Information Council, Brussels, Belgium.
| | - Alex F de Vos
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Carlie J M de Vries
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Claudia M van Tiel
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
85
|
Safe S, Jin UH, Morpurgo B, Abudayyeh A, Singh M, Tjalkens RB. Nuclear receptor 4A (NR4A) family - orphans no more. J Steroid Biochem Mol Biol 2016; 157:48-60. [PMID: 25917081 PMCID: PMC4618773 DOI: 10.1016/j.jsbmb.2015.04.016] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 03/26/2015] [Accepted: 04/21/2015] [Indexed: 01/17/2023]
Abstract
The orphan nuclear receptors NR4A1, NR4A2 and NR4A3 are immediate early genes induced by multiple stressors, and the NR4A receptors play an important role in maintaining cellular homeostasis and disease. There is increasing evidence for the role of these receptors in metabolic, cardiovascular and neurological functions and also in inflammation and inflammatory diseases and in immune functions and cancer. Despite the similarities of NR4A1, NR4A2 and NR4A3 and their interactions with common cis-genomic elements, they exhibit unique activities and cell-/tissue-specific functions. Although endogenous ligands for NR4A receptors have not been identified, there is increasing evidence that structurally-diverse synthetic molecules can directly interact with the ligand binding domain of NR4A1 and act as agonists or antagonists, and ligands for NR4A2 and NR4A3 have also been identified. Since NR4A receptors are key factors in multiple diseases, there are opportunities for the future development of NR4A ligands for clinical applications in treating multiple health problems including metabolic, neurologic and cardiovascular diseases, other inflammatory conditions, and cancer.
Collapse
MESH Headings
- Arthritis/metabolism
- Cardiovascular Diseases/metabolism
- DNA-Binding Proteins/metabolism
- Homeostasis
- Humans
- Immunity, Cellular
- Inflammation/metabolism
- Ligands
- Metabolic Diseases/genetics
- Metabolic Diseases/metabolism
- Neoplasms/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Receptors, Steroid/metabolism
- Receptors, Thyroid Hormone/metabolism
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA.
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Benjamin Morpurgo
- Texas A&M Institute for Genomic Medicine, Texas A&M University, 670 Raymond Stotzer Pkwy, College Station, TX 77843, USA
| | - Ala Abudayyeh
- Department of General Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mandip Singh
- Department of Pharmaceutics, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Ronald B Tjalkens
- Department of Toxicology and Neuroscience, Colorado State University, 1680Campus Delivery, Fort Collins, CO 80523-1680, USA
| |
Collapse
|
86
|
Shao Q, Han F, Peng S, He B. Nur77 inhibits oxLDL induced apoptosis of macrophages via the p38 MAPK signaling pathway. Biochem Biophys Res Commun 2016; 471:633-8. [DOI: 10.1016/j.bbrc.2016.01.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 01/02/2016] [Indexed: 11/29/2022]
|
87
|
Li XM, Zhang S, He XS, Guo PD, Lu XX, Wang JR, Li JM, Wu H. Nur77-mediated TRAF6 signalling protects against LPS-induced sepsis in mice. JOURNAL OF INFLAMMATION-LONDON 2016; 13:4. [PMID: 26839514 PMCID: PMC4735956 DOI: 10.1186/s12950-016-0112-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/25/2016] [Indexed: 12/20/2022]
Abstract
Background Nur77, a key member of the NR4A receptor subfamily, is involved in the regulation of inflammation and immunity. However, the in vivo regulatory roles of Nur77 in sepsis and the mechanisms involved remains largely elusive. In this study, we used Nur77-deficient (Nur77−/−) mice and investigated the function of Nur77 in sepsis. Findings Compared to wild-type (Nur77+/+) mice, Nur77−/− mice are more susceptible to LPS-induced sepsis and acute liver inflammation. Mechanistically, we observed that Nur77 can interact with TRAF6, a crucial adaptor molecule in the Toll-like receptor-interleukin 1 receptor (TLR-IL-1R) signalling pathway, in in vivo mouse model of sepsis. The interaction may affect TRAF6 auto-ubiquitination, thereby inhibiting NF-κB activation and pro-inflammatory cytokines production. Conclusions These in vivo observations reveals an important protective role for Nur77 in LPS-induced sepsis through its regulation to TRAF6 signalling, and highlights the potential clinical application of Nur77 as a molecular target in prevention and/or treatment of sepsis. Electronic supplementary material The online version of this article (doi:10.1186/s12950-016-0112-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiu-Ming Li
- Pathology Center and Department of Pathology, Soochow University, Suzhou, 215123 China
| | - Shen Zhang
- Pathology Center and Department of Pathology, Soochow University, Suzhou, 215123 China
| | - Xiao-Shun He
- The First Affiliated Hospital of Soochow University, Suzhou, 215006 China
| | - Peng-Da Guo
- Pathology Center and Department of Pathology, Soochow University, Suzhou, 215123 China
| | - Xing-Xing Lu
- Pathology Center and Department of Pathology, Soochow University, Suzhou, 215123 China
| | - Jing-Ru Wang
- Pathology Center and Department of Pathology, Soochow University, Suzhou, 215123 China
| | - Jian-Ming Li
- Pathology Center and Department of Pathology, Soochow University, Suzhou, 215123 China
| | - Hua Wu
- Pathology Center and Department of Pathology, Soochow University, Suzhou, 215123 China
| |
Collapse
|
88
|
Wu H, Li XM, Wang JR, Gan WJ, Jiang FQ, Liu Y, Zhang XD, He XS, Zhao YY, Lu XX, Guo YB, Zhang XK, Li JM. NUR77 exerts a protective effect against inflammatory bowel disease by negatively regulating the TRAF6/TLR-IL-1R signalling axis. J Pathol 2015; 238:457-69. [PMID: 26564988 DOI: 10.1002/path.4670] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 11/02/2015] [Accepted: 11/06/2015] [Indexed: 01/05/2023]
Abstract
Nur77, an immediate-early response gene, participates in a wide range of biological functions. Its human homologue, NUR77, is known by several names and has the HGNC-approved gene symbol NR4A1. However, the role of Nur77 in inflammatory bowel disease (IBD) and its underlying mechanisms remain elusive. Here, using public data from the International Inflammatory Bowel Disease Genetics Consortium (IIBDGC) on the most recent genome-wide association studies (GWAS) for ulcerative colitis (UC) and Crohn's disease (CD), we found that genetic variants of the NUR77 gene are associated with increased risk for both UC and CD. Accordingly, Nur77 expression was significantly reduced in colon tissues from patients with UC or CD and mice treated with DSS. Nur77 deficiency increased the susceptibility of mice to DSS-induced experimental colitis and prevented intestinal recovery, whereas treatment with cytosporone B (Csn-B), an agonist for Nur77, significantly attenuated excessive inflammatory response in the DSS-induced colitis mouse model. Mechanistically, NUR77 acts as a negative regulator of TLR-IL-1R signalling by interacting with TRAF6. This interaction prevented auto-ubiquitination and oligomerization of TRAF6 and subsequently inhibited NF-κB activation and pro-inflammatory cytokine production. Taken together, our GWAS-based analysis and in vitro and in vivo studies have demonstrated that Nur77 is an important regulator of TRAF6/TLR-IL-1R-initiated inflammatory signalling, and loss of Nur77 may contribute to the development of IBD, suggesting Nur77 as a potential target for the prevention and treatment of IBD.
Collapse
Affiliation(s)
- Hua Wu
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Xiu-Ming Li
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Jing-Ru Wang
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Wen-Juan Gan
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China.,First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fu-Quan Jiang
- School of Pharmaceutical Sciences, Xiamen University, People's Republic of China
| | - Yao Liu
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Xin-Dao Zhang
- School of Pharmaceutical Sciences, Xiamen University, People's Republic of China
| | - Xiao-Shun He
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Yuan-Yuan Zhao
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Xing-Xing Lu
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Yan-Bing Guo
- School of Pharmaceutical Sciences, Xiamen University, People's Republic of China.,Sanford-Burnham Medical Research Institute, Cancer Center, La Jolla, CA, USA
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Xiamen University, People's Republic of China.,Sanford-Burnham Medical Research Institute, Cancer Center, La Jolla, CA, USA
| | - Jian-Ming Li
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
89
|
Qing H, Liu Y, Zhao Y, Aono J, Jones KL, Heywood EB, Howatt D, Binkley CM, Daugherty A, Liang Y, Bruemmer D. Deficiency of the NR4A orphan nuclear receptor NOR1 in hematopoietic stem cells accelerates atherosclerosis. Stem Cells 2015; 32:2419-29. [PMID: 24806827 DOI: 10.1002/stem.1747] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 03/17/2014] [Accepted: 04/04/2014] [Indexed: 02/01/2023]
Abstract
The NR4A orphan nuclear receptor NOR1 functions as a constitutively active transcription factor regulating cellular inflammation and proliferation. In this study, we used bone marrow transplantation to determine the selective contribution of NOR1 expression in hematopoietic stem cells to the development of atherosclerosis. Reconstitution of lethally irradiated apoE(-/-) mice with NOR1-deficient hematopoietic stem cells accelerated atherosclerosis formation and macrophage recruitment following feeding a diet enriched in saturated fat. NOR1 deficiency in hematopoietic stem cells induced splenomegaly and monocytosis, specifically the abundance of inflammatory Ly6C(+) monocytes. Bone marrow transplantation studies further confirmed that NOR1 suppresses the proliferation of macrophage and dendritic progenitor (MDP) cells. Expression analysis identified RUNX1, a critical regulator of hematopoietic stem cell expansion, as a target gene suppressed by NOR1 in MDP cells. Finally, in addition to inducing Ly6C(+) monocytosis, NOR1 deletion increased the replicative rate of lesional macrophages and induced local foam cell formation within the atherosclerotic plaque. Collectively, our studies demonstrate that NOR1 deletion in hematopoietic stem cells accelerates atherosclerosis formation by promoting myelopoiesis in the stem cell compartment and by inducing local proatherogenic activities in the macrophage, including lesional macrophage proliferation and foam cell formation.
Collapse
Affiliation(s)
- Hua Qing
- Division of Cardiovascular Medicine, Gill Heart Institute, and Saha Cardiovascular Research Center, Chongqing, People's Republic of China; Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Xie X, Song X, Yuan S, Cai H, Chen Y, Chang X, Liang B, Huang D. Histone acetylation regulates orphan nuclear receptor NR4A1 expression in hypercholesterolaemia. Clin Sci (Lond) 2015; 129:1151-61. [PMID: 26396259 DOI: 10.1042/cs20150346] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/21/2015] [Indexed: 02/04/2023]
Abstract
Hypercholesterolaemia and inflammation are correlated with atherogenesis. Orphan nuclear receptor NR4A1, as a key regulator of inflammation, is closely associated with lipid levels in vivo. However, the mechanism by which lipids regulate NR4A1 expression remains unknown. We aimed to elucidate the underlying mechanism of NR4A1 expression in monocytes during hypercholesterolaemia, and reveal the potential role of NR4A1 in hypercholesterolaemia-induced circulating inflammation. Circulating leucocytes were collected from blood samples of 139 patients with hypercholesterolaemia and 139 sex- and age-matched healthy subjects. We found that there was a low-grade inflammatory state and higher expression of NR4A1 in patients. Both total cholesterol and low-density lipoprotein cholesterol levels in plasma were positively correlated with NR4A1 mRNA level. ChIP revealed that acetylation of histone H3 was enriched in the NR4A1 promoter region in patients. Human mononuclear cell lines THP-1 and U937 were treated with cholesterol. Supporting our clinical observations, cholesterol enhanced p300 acetyltransferase and decreased HDAC7 (histone deacetylase 7) recruitment to the NR4A1 promoter region, resulting in histone H3 hyperacetylation and further contributing to NR4A1 up-regulation in monocytes. Moreover, cytosporone B, an NR4A1 agonist, completely reversed cholesterol-induced IL-6 (interleukin 6) and MCP-1 (monocyte chemoattractant protein 1) expression to below basal levels, and knockdown of NR4A1 expression by siRNA not only mimicked, but also exaggerated the effects of cholesterol on inflammatory biomarker up-regulation. Thus we conclude that histone acetylation contributes to the regulation of NR4A1 expression in hypercholesterolaemia, and that NR4A1 expression reduces hypercholesterolaemia-induced inflammation.
Collapse
MESH Headings
- Acetylation
- Adult
- Aged
- Binding Sites
- Case-Control Studies
- Chemokine CCL2/metabolism
- Cholesterol/metabolism
- Female
- Gene Expression Regulation
- Histone Deacetylases/metabolism
- Histones/metabolism
- Humans
- Hypercholesterolemia/blood
- Hypercholesterolemia/genetics
- Hypercholesterolemia/metabolism
- Inflammation/blood
- Inflammation/genetics
- Inflammation/metabolism
- Inflammation/prevention & control
- Inflammation Mediators/blood
- Inflammation Mediators/metabolism
- Interleukin-6/metabolism
- Male
- Middle Aged
- Monocytes/drug effects
- Monocytes/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/agonists
- Nuclear Receptor Subfamily 4, Group A, Member 1/blood
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Phenylacetates/pharmacology
- Promoter Regions, Genetic
- Protein Processing, Post-Translational
- RNA Interference
- RNA, Messenger/metabolism
- Transfection
- U937 Cells
- p300-CBP Transcription Factors/metabolism
Collapse
Affiliation(s)
- Xina Xie
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Xuhong Song
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Song Yuan
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Haitao Cai
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Yequn Chen
- Department of Community Surveillance, The First Affiliated Hospital of Shantou, University Medical College, Shantou, 515041, China
| | - Xiaolan Chang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Bin Liang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Dongyang Huang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| |
Collapse
|
91
|
Sambri I, Crespo J, Aguiló S, Ingrosso D, Rodríguez C, Martínez González J. miR-17 and -20a Target the Neuron-Derived Orphan Receptor-1 (NOR-1) in Vascular Endothelial Cells. PLoS One 2015; 10:e0141932. [PMID: 26600038 PMCID: PMC4658114 DOI: 10.1371/journal.pone.0141932] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/14/2015] [Indexed: 11/19/2022] Open
Abstract
Neuron-derived orphan receptor-1 (NOR-1) plays a major role in vascular biology by controlling fibroproliferative and inflammatory responses. Because microRNAs (miRNAs) have recently emerged as key players in the regulation of gene expression in the vasculature, here we have investigated the regulation of NOR-1 by miRNAs in endothelial cells. Computational algorithms suggest that NOR-1 could be targeted by members of the miR-17 family. Accordingly, ectopic over-expression of miR-17 or miR-20a in endothelial cells using synthetic premiRNAs attenuated the up-regulation of NOR-1 expression induced by VEGF (as evidenced by real time PCR, Western blot and immunocitochemistry). Conversely, the antagonism of these miRNAs by specific antagomirs prevented the down-regulation of NOR-1 promoted by miR-17 or miR-20a in VEGF-stimulated cells. Disruption of the miRNA-NOR-1 mRNA interaction using a custom designed target protector evidenced the selectivity of these responses. Further, luciferase reporter assays and seed-sequence mutagenesis confirmed that miR-17 and -20a bind to NOR-1 3’-UTR. Finally, miR-17 and -20a ameliorated the up-regulation of VCAM-1 mediated by NOR-1 in VEGF-stimulated cells. Therefore, miR-17 and -20a target NOR-1 thereby regulating NOR-1-dependent gene expression.
Collapse
Affiliation(s)
- Irene Sambri
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
- Department of Biochemistry, Biophysics & General Pathology, School of Medicine & Surgery, Second University of Naples, Naples, Italy
| | - Javier Crespo
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Silvia Aguiló
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Diego Ingrosso
- Department of Biochemistry, Biophysics & General Pathology, School of Medicine & Surgery, Second University of Naples, Naples, Italy
| | - Cristina Rodríguez
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - José Martínez González
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
- * E-mail:
| |
Collapse
|
92
|
Orphan nuclear receptor Nur77 affects cardiomyocyte calcium homeostasis and adverse cardiac remodelling. Sci Rep 2015; 5:15404. [PMID: 26486271 PMCID: PMC4613907 DOI: 10.1038/srep15404] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 09/21/2015] [Indexed: 12/22/2022] Open
Abstract
Distinct stressors may induce heart failure. As compensation, β-adrenergic stimulation enhances myocardial contractility by elevating cardiomyocyte intracellular Ca2+ ([Ca2+]i). However, chronic β-adrenergic stimulation promotes adverse cardiac remodelling. Cardiac expression of nuclear receptor Nur77 is enhanced by β-adrenergic stimulation, but its role in cardiac remodelling is still unclear. We show high and rapid Nur77 upregulation in cardiomyocytes stimulated with β-adrenergic agonist isoproterenol. Nur77 knockdown in culture resulted in hypertrophic cardiomyocytes. Ventricular cardiomyocytes from Nur77-deficient (Nur77-KO) mice exhibited elevated diastolic and systolic [Ca2+]i and prolonged action potentials compared to wild type (WT). In vivo, these differences resulted in larger cardiomyocytes, increased expression of hypertrophic genes, and more cardiac fibrosis in Nur77-KO mice upon chronic isoproterenol stimulation. In line with the observed elevated [Ca2+]i, Ca2+-activated phosphatase calcineurin was more active in Nur77-KO mice compared to WT. In contrast, after cardiac pressure overload by aortic constriction, Nur77-KO mice exhibited attenuated remodelling compared to WT. Concluding, Nur77-deficiency results in significantly altered cardiac Ca2+ homeostasis and distinct remodelling outcome depending on the type of insult. Detailed knowledge on the role of Nur77 in maintaining cardiomyocyte Ca2+ homeostasis and the dual role Nur77 plays in cardiac remodelling will aid in developing personalized therapies against heart failure.
Collapse
|
93
|
Wolf D, Zirlik A, Ley K. Beyond vascular inflammation--recent advances in understanding atherosclerosis. Cell Mol Life Sci 2015; 72:3853-69. [PMID: 26100516 PMCID: PMC4577451 DOI: 10.1007/s00018-015-1971-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 06/10/2015] [Accepted: 06/15/2015] [Indexed: 12/23/2022]
Abstract
Atherosclerosis is the most life-threatening pathology worldwide. Its major clinical complications, stroke, myocardial infarction, and heart failure, are on the rise in many regions of the world--despite considerable progress in understanding cause, progression, and consequences of atherosclerosis. Originally perceived as a lipid-storage disease of the arterial wall (Die cellularpathologie in ihrer begründung auf physiologische und pathologische gewebelehre. August Hirschwald Verlag Berlin, [1871]), atherosclerosis was recognized as a chronic inflammatory disease in 1986 (New Engl J Med 314:488-500, 1986). The presence of lymphocytes in atherosclerotic lesions suggested autoimmune processes in the vessel wall (Clin Exp Immunol 64:261-268, 1986). Since the advent of suitable mouse models of atherosclerosis (Science 258:468-471, 1992; Cell 71:343-353, 1992; J Clin Invest 92:883-893, 1993) and the development of flow cytometry to define the cellular infiltrate in atherosclerotic lesions (J Exp Med 203:1273-1282, 2006), the origin, lineage, phenotype, and function of distinct inflammatory cells that trigger or inhibit the inflammatory response in the atherosclerotic plaque have been studied. Multiphoton microscopy recently enabled direct visualization of antigen-specific interactions between T cells and antigen-presenting cells in the vessel wall (J Clin Invest 122:3114-3126, 2012). Vascular immunology is now emerging as a new field, providing evidence for protective as well as damaging autoimmune responses (Int Immunol 25:615-622, 2013). Manipulating inflammation and autoimmunity both hold promise for new therapeutic strategies in cardiovascular disease. Ongoing work (J Clin Invest 123:27-36, 2013; Front Immunol 2013; Semin Immunol 31:95-101, 2009) suggests that it may be possible to develop antigen-specific immunomodulatory prevention and therapy-a vaccine against atherosclerosis.
Collapse
Affiliation(s)
- Dennis Wolf
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA
| | - Andreas Zirlik
- Atherogenesis Research Group, Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle Drive, La Jolla, CA, 92037, USA.
| |
Collapse
|
94
|
Ruiter MS, van Tiel CM, Doornbos A, Marinković G, Strang AC, Attevelt NJM, de Waard V, de Winter RJ, Steendam R, de Vries CJM. Stents Eluting 6-Mercaptopurine Reduce Neointima Formation and Inflammation while Enhancing Strut Coverage in Rabbits. PLoS One 2015; 10:e0138459. [PMID: 26389595 PMCID: PMC4577071 DOI: 10.1371/journal.pone.0138459] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/31/2015] [Indexed: 11/19/2022] Open
Abstract
Background The introduction of drug-eluting stents (DES) has dramatically reduced restenosis rates compared with bare metal stents, but in-stent thrombosis remains a safety concern, necessitating prolonged dual anti-platelet therapy. The drug 6-Mercaptopurine (6-MP) has been shown to have beneficial effects in a cell-specific fashion on smooth muscle cells (SMC), endothelial cells and macrophages. We generated and analyzed a novel bioresorbable polymer coated DES, releasing 6-MP into the vessel wall, to reduce restenosis by inhibiting SMC proliferation and decreasing inflammation, without negatively affecting endothelialization of the stent surface. Methods Stents spray-coated with a bioresorbable polymer containing 0, 30 or 300 μg 6-MP were implanted in the iliac arteries of 17 male New Zealand White rabbits. Animals were euthanized for stent harvest 1 week after implantation for evaluation of cellular stent coverage and after 4 weeks for morphometric analyses of the lesions. Results Four weeks after implantation, the high dose of 6-MP attenuated restenosis with 16% compared to controls. Reduced neointima formation could at least partly be explained by an almost 2-fold induction of the cell cycle inhibiting kinase p27Kip1. Additionally, inflammation score, the quantification of RAM11-positive cells in the vessel wall, was significantly reduced in the high dose group with 23% compared to the control group. Evaluation with scanning electron microscopy showed 6-MP did not inhibit strut coverage 1 week after implantation. Conclusion We demonstrate that novel stents coated with a bioresorbable polymer coating eluting 6-MP inhibit restenosis and attenuate inflammation, while stimulating endothelial coverage. The 6-MP-eluting stents demonstrate that inhibition of restenosis without leaving uncovered metal is feasible, bringing stents without risk of late thrombosis one step closer to the patient.
Collapse
Affiliation(s)
- Matthijs S. Ruiter
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Claudia M. van Tiel
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Goran Marinković
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Aart C. Strang
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Nico J. M. Attevelt
- Central Laboratory Animal Research Facility, Faculty of Veterinary Medicine, University of Utrecht, Utrecht, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Robbert J. de Winter
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Rob Steendam
- InnoCore Pharmaceuticals, Groningen, The Netherlands
| | - Carlie J. M. de Vries
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
95
|
Liu K, Zhou W, Chen H, Pan H, Sun X, You Q. Autologous vein graft stenosis inhibited by orphan nuclear receptor Nur77-targeted siRNA. Vascul Pharmacol 2015; 73:64-70. [PMID: 26276525 DOI: 10.1016/j.vph.2015.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 07/30/2015] [Accepted: 08/09/2015] [Indexed: 10/23/2022]
Abstract
Neointimal hyperplasia plays an important role in autologous vein graft stenosis, and orphan receptor TR3/nur77 (Nur77) might play an essential role, but the mechanisms are still unclear. Here, we investigated the function of Nur77 in autologous vein graft stenosis. Rat vascular smooth muscle cell A7r5 was used for evaluating the function of Nur77 and screen siRNAs. Meanwhile, rat vein graft models were constructed for investigating the stenosis inhibition effects of Nur77-targeted siRNAs. The mRNA and protein levels of Nur77 were highly expressed in A7r5 cell, and could be significantly inhibited by the pre-designed siRNAs; the proliferation of A7r5 cell was also inhibited by the siRNAs. Furthermore, the intimal thickening in rat vein graft models was inhibited when knocking down the expression of Nur77 by siRNA. The results suggest that Nur77-targeted siRNA can inhibit autologous vein graft stenosis, Nur77 may play an important role in autologous vein graft stenosis, and Nur77 targeted siRNAs may be a therapy method for anti-stenosis of autologous vein graft.
Collapse
Affiliation(s)
- Kun Liu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Wen Zhou
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | | | - Haiyan Pan
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Xiaohui Sun
- Department of Cardiology, Nantong Geriatric Rehabilitation Hosptial, Nantong 226001, China
| | - Qingsheng You
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China.
| |
Collapse
|
96
|
Hamers AAJ, van Dam L, Teixeira Duarte JM, Vos M, Marinković G, van Tiel CM, Meijer SL, van Stalborch AM, Huveneers S, te Velde AA, de Jonge WJ, de Vries CJM. Deficiency of Nuclear Receptor Nur77 Aggravates Mouse Experimental Colitis by Increased NFκB Activity in Macrophages. PLoS One 2015; 10:e0133598. [PMID: 26241646 PMCID: PMC4524678 DOI: 10.1371/journal.pone.0133598] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 06/29/2015] [Indexed: 02/07/2023] Open
Abstract
Nuclear receptor Nur77, also referred to as NR4A1 or TR3, plays an important role in innate and adaptive immunity. Nur77 is crucial in regulating the T helper 1/regulatory T-cell balance, is expressed in macrophages and drives M2 macrophage polarization. In this study we aimed to define the function of Nur77 in inflammatory bowel disease. In wild-type and Nur77-/- mice, colitis development was studied in dextran sodium sulphate (DSS)- and 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced models. To understand the underlying mechanism, Nur77 was overexpressed in macrophages and gut epithelial cells. Nur77 protein is expressed in colon tissues from Crohn's disease and Ulcerative colitis patients and colons from colitic mice in inflammatory cells and epithelium. In both mouse colitis models inflammation was increased in Nur77-/- mice. A higher neutrophil influx and enhanced IL-6, MCP-1 and KC production was observed in Nur77-deficient colons after DSS-treatment. TNBS-induced influx of T-cells and inflammatory monocytes into the colon was higher in Nur77-/- mice, along with increased expression of MCP-1, TNFα and IL-6, and decreased Foxp3 RNA expression, compared to wild-type mice. Overexpression of Nur77 in lipopolysaccharide activated RAW macrophages resulted in up-regulated IL-10 and downregulated TNFα, MIF-1 and MCP-1 mRNA expression through NFκB repression. Nur77 also strongly decreased expression of MCP-1, CXCL1, IL-8, MIP-1α and TNFα in gut epithelial Caco-2 cells. Nur77 overexpression suppresses the inflammatory status of both macrophages and gut epithelial cells and together with the in vivo mouse data this supports that Nur77 has a protective function in experimental colitis. These findings may have implications for development of novel targeted treatment strategies regarding inflammatory bowel disease and other inflammatory diseases.
Collapse
MESH Headings
- Animals
- Cell Line
- Colitis/chemically induced
- Colitis/immunology
- Colitis/metabolism
- Colitis, Ulcerative/metabolism
- Colitis, Ulcerative/pathology
- Colon/metabolism
- Colon/pathology
- Crohn Disease/metabolism
- Crohn Disease/pathology
- Cytokines/biosynthesis
- Cytokines/genetics
- Dextran Sulfate/toxicity
- Disease Models, Animal
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Female
- Forkhead Transcription Factors/biosynthesis
- Forkhead Transcription Factors/genetics
- Gene Expression Regulation
- Humans
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Macrophages/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- NF-kappa B/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/immunology
- RAW 264.7 Cells
- Trinitrobenzenesulfonic Acid/toxicity
Collapse
Affiliation(s)
- Anouk A. J. Hamers
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Laura van Dam
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - José M. Teixeira Duarte
- Tytgat Institute, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Mariska Vos
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Goran Marinković
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Claudia M. van Tiel
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sybren L. Meijer
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Anne-Marieke van Stalborch
- Department of Molecular Cell Biology, Sanquin Research and Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Stephan Huveneers
- Department of Molecular Cell Biology, Sanquin Research and Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Anje A. te Velde
- Tytgat Institute, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Wouter J. de Jonge
- Tytgat Institute, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlie J. M. de Vries
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
97
|
XU YING, HUANG QI, ZHANG WENFANG, WANG YAPING, ZENG QINGLING, HE CHUNYAN, XUE JUNLI, CHEN JIN, HU XUEMEI, XU YANCHENG. Decreased expression levels of Nurr1 are associated with chronic inflammation in patients with type 2 diabetes. Mol Med Rep 2015; 12:5487-93. [DOI: 10.3892/mmr.2015.4105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 04/20/2015] [Indexed: 11/06/2022] Open
|
98
|
Crean D, Cummins EP, Bahar B, Mohan H, McMorrow JP, Murphy EP. Adenosine Modulates NR4A Orphan Nuclear Receptors To Attenuate Hyperinflammatory Responses in Monocytic Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:1436-48. [PMID: 26150530 DOI: 10.4049/jimmunol.1402039] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 06/02/2015] [Indexed: 02/02/2023]
Abstract
Adenosine receptor-mediated regulation of monocyte/macrophage inflammatory responses is critical in the maintenance of tissue homeostasis. In this study, we reveal that adenosine potently modulates the expression of NR4A1, 2, and 3 orphan nuclear receptors in myeloid cells, and this modulation is primarily through the adenosine A2a receptor subtype. We demonstrate that A2a receptor activation of NR4A1-3 receptor synthesis is further enhanced in TLR4-stimulated monocytes. After TLR4 stimulation, NR4A receptor-depleted monocyte/macrophage cells display significantly altered expression of cell-surface markers and produce increased inflammatory cytokine and chemokine secretion rendering the cells an enhanced proinflammatory phenotype. Exposure of TLR4 or TNF-α-stimulated monocytes to adenosine analogs directs changes in the expression of MIP-3α and IL-23p19, with NR4A2 depletion leading to significantly enhanced expression of these factors. Furthermore, we establish that nuclear levels of NF-κB/p65 are increased in TLR/adenosine-stimulated NR4A2-depleted cells. We show that, after TLR/adenosine receptor stimulation, NR4A2 depletion promotes significant binding of NF-κB/p65 to a κB consensus binding motif within the MIP-3α proximal promoter leading to increased protein secretion, confirming a pivotal role for NF-κB activity in controlling cellular responses and gene expression outcomes in response to these mediators. Thus, these data demonstrate that during an inflammatory response, adenosine modulation of NR4A receptor activity acts to limit NF-κB-mediated effects and that loss of NR4A2 expression leads to enhanced NF-κB activity and hyperinflammatory responses in myeloid cells.
Collapse
Affiliation(s)
- Daniel Crean
- UCD Veterinary Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; and Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eoin P Cummins
- Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Bojlul Bahar
- UCD Veterinary Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; and
| | - Helen Mohan
- UCD Veterinary Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; and
| | - Jason P McMorrow
- UCD Veterinary Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; and
| | - Evelyn P Murphy
- UCD Veterinary Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; and Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
99
|
Molecular Interactions between NR4A Orphan Nuclear Receptors and NF-κB Are Required for Appropriate Inflammatory Responses and Immune Cell Homeostasis. Biomolecules 2015; 5:1302-18. [PMID: 26131976 PMCID: PMC4598753 DOI: 10.3390/biom5031302] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/16/2015] [Accepted: 06/16/2015] [Indexed: 12/22/2022] Open
Abstract
Appropriate innate and adaptive immune responses are essential for protection and resolution against chemical, physical or biological insults. Immune cell polarization is fundamental in orchestrating distinct phases of inflammation, specifically acute phase responses followed by resolution and tissue repair. Dysregulation of immune cell and inflammatory responses is a hallmark of multiple diseases encompassing atherosclerosis, rheumatoid arthritis, psoriasis and metabolic syndromes. A master transcriptional mediator of diverse inflammatory signaling and immune cell function is NF-κB, and altered control of this key regulator can lead to an effective switch from acute to chronic inflammatory responses. Members of the nuclear receptor (NR) superfamily of ligand-dependent transcription factors crosstalk with NF-κB to regulate immune cell function(s). Within the NR superfamily the NR4A1-3 orphan receptors have emerged as important regulators of immune cell polarization and NF-κB signaling. NR4A receptors modulate NF-κB activity in a dynamic fashion, either repressing or enhancing target gene expression leading to altered inflammatory outcome. Here we will discuss the pivotal role NR4A’s receptors play in orchestrating immune cell homeostasis through molecular crosstalk with NF-κB. Specifically, we will examine such NR4A/NF-κB interactions within the context of distinct cell phenotypes, including monocyte, macrophage, T cells, endothelial, and mesenchymal cells, which play a role in inflammation-associated disease. Finally, we review the therapeutic potential of altering NR4A/NF-κB interactions to limit hyper-inflammatory responses in vivo.
Collapse
|
100
|
Nur77 deficiency leads to systemic inflammation in elderly mice. JOURNAL OF INFLAMMATION-LONDON 2015; 12:40. [PMID: 26113803 PMCID: PMC4480882 DOI: 10.1186/s12950-015-0085-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 06/18/2015] [Indexed: 01/18/2023]
Abstract
Background Nur77, an orphan member of the nuclear receptor superfamily, has been implicated in the regulation of inflammation. However, the in vivo function of Nur77 remains largely unexplored. In the current study, we investigated the role of Nur77 in inflammation and immunity in mice. Findings We found that elderly 8-month-old Nur77-deficient mice (Nur77−/−) developed systemic inflammation. Compared to wild-type (WT) mice (Nur77+/+), Nur77−/− mice showed splenomegaly, severe infiltration of inflammatory cells in several organs including liver, lung, spleen and kidney, increased hyperplasia of fibrous tissue in the lung and enlargement of kidney glomeruli. Additionally, Nur77−/− mice had increased production of pro-inflammatory cytokines and immunoglobulin, and elicited pro-inflammatory M1-like polarization in macrophages as revealed by increased expression of CXCL11 and INDO, and decreased expression of MRC1. Conclusions These in vivo observations provide evidence for a pivotal role for Nur77 in the regulation of systemic inflammation and emphasize the pathogenic significance of Nur77 in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s12950-015-0085-0) contains supplementary material, which is available to authorized users.
Collapse
|