1
|
Zou X, Liu T, Huang Z, Zhou W, Yuan M, Zhao H, Pan Z, Chen P, Shao Y, Hu X, Zhang S, Zheng S, Zhang Y, Huang P. SOX17 is a Critical Factor in Maintaining Endothelial Function in Pulmonary Hypertension by an Exosome-Mediated Autocrine Manner. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206139. [PMID: 36919784 PMCID: PMC10190640 DOI: 10.1002/advs.202206139] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/20/2023] [Indexed: 05/18/2023]
Abstract
Endothelial dysfunction is considered a predominant driver for pulmonary vascular remodeling in pulmonary hypertension (PH). SOX17, a key regulator of vascular homoeostasis, has been found to harbor mutations in PH patients, which are associated with PH susceptibility. Here, this study explores whether SOX17 mediates the autocrine activity of pulmonary artery ECs to maintain endothelial function and vascular homeostasis in PH and its underlying mechanism. It is found that SOX17 expression is downregulated in the endothelium of remodeled pulmonary arteries in IPH patients and SU5416/hypoxia (Su/hypo)-induced PH mice as well as dysfunctional HPAECs. Endothelial knockdown of SOX17 accelerates the progression of Su/hypo-induced PH in mice. SOX17 overexpression in the pulmonary endothelium of mice attenuates Su/hypo-induced PH. SOX17-associated exosomes block the proliferation, apoptosis, and inflammation of HPAECs, preventing pulmonary arterial remodeling and Su/hypo-induced PH. Mechanistic analyses demonstrates that overexpressing SOX17 promotes the exosome-mediated release of miR-224-5p and miR-361-3p, which are internalized by injured HPAECs in an autocrine manner, ultimately repressing the upregulation of NR4A3 and PCSK9 genes and improving endothelial function. These results suggest that SOX17 is a key gene in maintaining endothelial function and vascular homeostasis in PH through regulating exosomal miRNAs in an autocrine manner.
Collapse
Affiliation(s)
- Xiaozhou Zou
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Ting Liu
- Department of PharmacyAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou310006P. R. China
- Department of Clinical PharmacyKey Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang ProvinceAffiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhou310006P. R. China
| | - Zhongjie Huang
- School of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhou310014P. R. China
| | - Wei Zhou
- Zhongnan Hospital of Wuhan UniversityInstitute of Hepatobiliary Diseases of Wuhan UniversityTransplant Center of Wuhan UniversityHubei Key Laboratory of Medical Technology on TransplantationWuhan430000P. R. China
| | - Mengnan Yuan
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Hongying Zhao
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Zongfu Pan
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Pengcheng Chen
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Yanfei Shao
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Xiaoping Hu
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Su Zhang
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Shuilian Zheng
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Yiwen Zhang
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| | - Ping Huang
- Center for Clinical PharmacyCancer CenterDepartment of PharmacyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhou310014P. R. China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceHangzhou310014P. R. China
| |
Collapse
|
2
|
Ding R, Sun X, Yi B, Liu W, Kazama K, Xu X, Deshpande D, Liang C, Sun J. Nur77 Attenuates Inflammasome Activation by Inhibiting Caspase-1 Expression in Pulmonary Vascular Endothelial Cells. Am J Respir Cell Mol Biol 2021; 65:288-299. [PMID: 33971110 DOI: 10.1165/rcmb.2020-0524oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Inflammasomes are intracellular multiprotein complexes that help trigger and maintain the inflammatory response as part of the innate immune system. Recently, it has been increasingly recognized that aberrant inflammasome activation is critically involved endothelial dysfunction in a variety of human diseases, such as atherosclerosis, acute lung injury (ALI), and type 2 diabetes. The molecular mechanisms underlying endothelial inflammasome activation, however, are not completely elucidated. In the present study, we identified orphan nuclear receptor Nur77 as a novel regulator in controlling inflammasome activation in vascular endothelial cells (ECs). We demonstrated that LPS-induced inflammasome activation was significantly inhibited by ectopic overexpression of Nur77, predominantly through transcriptionally suppression of caspase-1 expression in vascular ECs. Consistent with this observation, we found that LPS-induced inflammasome activation was significantly augmented in lung ECs isolated from Nur77 knockout (KO) mice. Mechanistically, we showed that Nur77-induced inhibition of caspase-1 expression was due to an inhibition of Interferon Regulatory Factor 1 (IRF1) expression and its subsequent binding to the caspase-1 promoter. Importantly, in a mouse model of LPS-induced acute lung injury (ALI), Nur77 KO led to a marked activation of caspase-1 in the lung, increased alveolar and circulating IL-1β levels, and exacerbated ALI, all of which were substantially inhibited by administration of caspase-1 inhibitor. Together, our results support an important role for Nur77 in controlling inflammasome activation in vascular ECs and suggest that Nur77 could be a novel therapeutic target for the treatment of human diseases associated with aberrant inflammasome activation, such as ALI and atherosclerosis.
Collapse
Affiliation(s)
- Ru Ding
- Thomas Jefferson University, 6559, Philadelphia, Pennsylvania, United States
| | - Xiaobo Sun
- Thomas Jefferson University, 6559, Philadelphia, Pennsylvania, United States
| | - Bing Yi
- Thomas Jefferson University, 6559, Philadelphia, Pennsylvania, United States
| | - Wennan Liu
- Thomas Jefferson University, 6559, Philadelphia, Pennsylvania, United States
| | - Kyosuke Kazama
- Thomas Jefferson University, 6559, Philadelphia, Pennsylvania, United States
| | - Xinyun Xu
- Changzheng Hospital, 56652, Shanghai, China
| | - Deepak Deshpande
- Thomas Jefferson University, 6559, Center for Translational Medicine, Philadelphia, Pennsylvania, United States
| | - Chun Liang
- Changzheng Hospital, 56652, Shanghai, China
| | - Jianxin Sun
- Thomas Jefferson University, 6559, Philadelphia, Pennsylvania, United States;
| |
Collapse
|
3
|
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]
|
4
|
Zhu N, Zhang GX, Yi B, Guo ZF, Jang S, Yin Y, Yang F, Summer R, Sun J. Nur77 limits endothelial barrier disruption to LPS in the mouse lung. Am J Physiol Lung Cell Mol Physiol 2019; 317:L615-L624. [PMID: 31461311 DOI: 10.1152/ajplung.00425.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nur77 is an orphan nuclear receptor implicated in the regulation of a wide range of biological processes, including the maintenance of systemic blood vessel homeostasis. Although Nur77 is known to be expressed in the lung, its role in regulating pulmonary vascular functions remains entirely unknown. In this study, we found that Nur77 is expressed at high levels in the lung, and its expression is markedly upregulated in response to LPS administration. While the pulmonary vasculature of mice that lacked Nur77 appeared to function normally under homeostatic conditions, we observed a dramatic decrease in its barrier functions after exposure to LPS, as demonstrated by an increase in serum proteins in the bronchoalveolar lavage fluid and a reduction in the expression of endothelial junctional proteins, such as vascular endothelial cadherin (VE-cadherin) and β-catenin. Similarly, we found that siRNA knockdown of Nur77 in lung microvascular endothelial cells also reduced VE-cadherin and β-catenin expression and increased the quantity of fluorescein isothiocyanate-labeled dextran transporting across LPS-injured endothelial monolayers. Consistent with Nur77 playing a vascular protective role, we found that adenoviral-mediated overexpression of Nur77 both enhanced expression of VE-cadherin and β-catenin and augmented endothelial barrier protection to LPS in cultured cells. Mechanistically, Nur77 appeared to mediate its protective effects, at least in part, by binding to β-catenin and preventing its degradation. Our findings demonstrate a key role for Nur77 in the maintenance of lung endothelial barrier protection to LPS and suggest that therapeutic strategies aimed at augmenting Nur77 levels might be effective in treating a wide variety of inflammatory vascular diseases of the lung.
Collapse
Affiliation(s)
- Ni Zhu
- Center for Translational Medicine and The Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guan-Xin Zhang
- Center for Translational Medicine and The Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Bing Yi
- Center for Translational Medicine and The Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Zhi-Fu Guo
- Center for Translational Medicine and The Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Soohwa Jang
- Center for Translational Medicine and The Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Yongqiang Yin
- Center for Translational Medicine and The Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Fan Yang
- Center for Translational Medicine and The Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ross Summer
- Center for Translational Medicine and The Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jianxin Sun
- Center for Translational Medicine and The Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
5
|
Lee HR, Kong SY, Sung SH, Kim HJ. DA-9801 and its saponins, dioscin and protodioscin, protect primary cortical neurons from hyperglycemia-induced neurotoxicity. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.01.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
6
|
The microRNAs Regulating Vascular Smooth Muscle Cell Proliferation: A Minireview. Int J Mol Sci 2019; 20:ijms20020324. [PMID: 30646627 PMCID: PMC6359109 DOI: 10.3390/ijms20020324] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/01/2019] [Accepted: 01/02/2019] [Indexed: 12/14/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) proliferation plays a critical role in atherosclerosis. At the beginning of the pathologic process of atherosclerosis, irregular VSMC proliferation promotes plaque formation, but in advanced plaques VSMCs are beneficial, promoting the stability and preventing rupture of the fibrous cap. Recent studies have demonstrated that microRNAs (miRNAs) expressed in the vascular system are involved in the control of VSMC proliferation. This review summarizes recent findings on the miRNAs in the regulation of VSMC proliferation, including miRNAs that exhibit the inhibition or promotion of VSMC proliferation, and their targets mediating the regulation of VSMC proliferation. Up to now, most of the studies were performed only in cultured VSMC. While the modulation of miRNAs is emerging as a promising strategy for the regulation of VSMC proliferation, most of the effects of miRNAs and their targets in vivo require further investigation.
Collapse
|
7
|
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: 91] [Impact Index Per Article: 11.4] [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
|
8
|
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
|
9
|
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]
|
10
|
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
|
11
|
Chistiakov DA, Bobryshev YV, Orekhov AN. Changes in transcriptome of macrophages in atherosclerosis. J Cell Mol Med 2015; 19:1163-73. [PMID: 25973901 PMCID: PMC4459832 DOI: 10.1111/jcmm.12591] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/16/2015] [Indexed: 12/20/2022] Open
Abstract
Macrophages display significant phenotypic heterogeneity. Two growth factors, macrophage colony-stimulating factor and chemokine (C-X-C motif) ligand 4, drive terminal differentiation of monocytes to M0 and M4 macrophages respectively. Compared to M0 macrophages, M4 cells have a unique transcriptome, with expression of surface markers such as S100A8, mannose receptor CD206 and matrix metalloproteinase 7. M4 macrophages did not express CD163, a scavenger receptor for haemoglobin/haptoglobin complex. Depending on the stimuli, M0 macrophages could polarize towards the proinflammatory M1 subset by treatment with lipopolysaccharide or interferon-γ. These macrophages produce a range of proinflammatory cytokines, nitric oxide, reactive oxygen species and exhibit high chemotactic and phagocytic activity. The alternative M2 type could be induced from M0 macrophage by stimulation with interleukin (IL)-4. M2 macrophages express high levels of CD206 and produce anti-inflammatory cytokines IL-10 and transforming growth factor-β. M1, M2 and M4 macrophages could be found in atherosclerotic plaques. In the plaque, macrophages are subjected to the intensive influence not only by cytokines and chemokines but also with bioactive lipids such as cholesterol and oxidized phospholipids. Oxidized phospholipids induce a distinct Mox phenotype in murine macrophages that express a unique panel of antioxidant enzymes under control of the redox-regulated transcription factor Klf2, resistant to lipid accumulation. In unstable human lesions, atheroprotective M(Hb) and HA-mac macrophage subsets could be found. These two subsets are induced by the haemoglobin/haptoglobin complex, highly express haeme oxygenase 1 and CD163, and are implicated in clearance of haemoglobin and erythrocyte remnants. In atherogenesis, the macrophage phenotype is plastic and could therefore be switched to proinflammatory (i.e. proatherogenic) and anti-inflammatory (i.e. atheroprotective). The aim of this review was to characterize changes in macrophage transcriptome in atherosclerosis and discuss key markers that characterize different phenotypes of macrophages present in atherosclerotic lesions.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Medical Nanobiotechnology, Pirogov Russian State Medical University, Moscow, Russia
| | - Yuri V Bobryshev
- Faculty of Medicine and St Vincent's Centre for Applied Medical Research, University of New South Wales, Sydney, NSW, Australia.,School of Medicine, University of Western Sydney, Campbelltown, NSW, Australia.,Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Alexander N Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia.,Department of Biophysics, Biological Faculty, Moscow State University, Moscow, Russia
| |
Collapse
|
12
|
Ma Y. Neuron-derived orphan receptor 1: Working towards a common goal. J Mol Cell Cardiol 2015; 80:98-100. [DOI: 10.1016/j.yjmcc.2014.12.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 12/28/2014] [Indexed: 10/24/2022]
|
13
|
Calvayrac O, Rodríguez-Calvo R, Martí-Pamies I, Alonso J, Ferrán B, Aguiló S, Crespo J, Rodríguez-Sinovas A, Rodríguez C, Martínez-González J. NOR-1 modulates the inflammatory response of vascular smooth muscle cells by preventing NFκB activation. J Mol Cell Cardiol 2014; 80:34-44. [PMID: 25536180 DOI: 10.1016/j.yjmcc.2014.12.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 11/19/2014] [Accepted: 12/07/2014] [Indexed: 11/25/2022]
Abstract
Recent work has highlighted the role of NR4A receptors in atherosclerosis and inflammation. In vascular smooth muscle cell (VSMC) proliferation, however, NOR-1 (neuron-derived orphan receptor-1) exerts antagonistic effects to Nur77 and Nurr1. The aim of this study was to analyse the effect of NOR-1 in VSMC inflammatory response. We assessed the consequence of a gain-of-function of this receptor on the response of VSMC to inflammatory stimuli. In human VSMC, lentiviral over-expression of NOR-1 reduced lipopolysaccharide (LPS)-induced up-regulation of cytokines (IL-1β, IL-6 and IL-8) and chemokines (MCP-1 and CCL20). Similar effects were obtained in cells stimulated with TNFα or oxLDL. Conversely, siRNA-mediated NOR-1 inhibition significantly increased the expression of pro-inflammatory mediators. Interestingly, in the aortas from transgenic mice that over-express human NOR-1 in VSMC (TgNOR-1), the up-regulation of cytokine/chemokine by LPS was lower compared to wild-type littermates. Similar results were obtained in VSMC from transgenic animals. NOR-1 reduced the transcriptional activity of NFκB sensitive promoters (in transient transfections), and the binding of NFκB to its responsive element (in electrophoretic mobility shift assays). Furthermore, NOR-1 prevented the activation of NFκB pathway by decreasing IκBα phosphorylation/degradation and inhibiting the phosphorylation and subsequent translocation of p65 to the nucleus (assessed by Western blot and immunocytochemistry). These effects were associated with an attenuated phosphorylation of ERK1/2, p38 MAPK and Jun N-terminal kinase, pathways involved in the activation of NFκB. In mouse challenged with LPS, the activation of the NFκB signalling was also attenuated in the aorta from TgNOR-1. Our data support a role for NOR-1 as a negative modulator of the acute response elicited by pro-inflammatory stimuli in the vasculature.
Collapse
Affiliation(s)
- Olivier Calvayrac
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | | | - Ingrid Martí-Pamies
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Judith Alonso
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Beatriz Ferrán
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Silvia Aguiló
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Javier Crespo
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | | | - Cristina Rodríguez
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain.
| | | |
Collapse
|
14
|
Huo Y, Yi B, Chen M, Wang N, Chen P, Guo C, Sun J. Induction of Nur77 by hyperoside inhibits vascular smooth muscle cell proliferation and neointimal formation. Biochem Pharmacol 2014; 92:590-8. [PMID: 25316569 DOI: 10.1016/j.bcp.2014.09.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/24/2014] [Accepted: 09/24/2014] [Indexed: 02/02/2023]
Abstract
Nur77 is an orphan nuclear receptor that belongs to the nuclear receptor 4A (NR4A) subfamily, which has been implicated in a variety of biological events, such as cell apoptosis, proliferation, inflammation, and metabolism. Activation of Nur77 has recently been shown to be beneficial for the treatment of cardiovascular and metabolic diseases. The purpose of this study is to identify novel natural Nur77 activators and investigate their roles in preventing vascular diseases. By measuring Nur77 expression using quantitative RT-PCR, we screened active ingredients extracted from Chinese herb medicines with beneficial cardiovascular effects. Hyperoside (quercetin 3-D-galactoside) was identified as one of the potent activators for inducing Nur77 expression and activating its transcriptional activity in vascular smooth muscle cells (VSMCs). We demonstrated that hyperoside, in a time and dose dependent manner, markedly increased the expression of Nur77 in rat VSMCs, with an EC50 of ∼0.83 μM. Mechanistically, we found that hyperoside significantly increased the phosphorylation of ERK1/2 MAP kinase and its downstream target cAMP response element-binding protein (CREB), both of which contributed to the hyperoside-induced Nur77 expression in rat VSMCs. Moreover, through activation of Nur77 receptor, hyperoside markedly inhibited both vascular smooth muscle cell proliferation in vitro and the carotid artery ligation-induced neointimal formation in vivo. These findings demonstrate that hyperoside is a potent natural activator of Nur77 receptor, which can be potentially used for prevention and treatment of occlusive vascular diseases.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cell Proliferation/drug effects
- Cells, Cultured
- DNA Primers
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Nuclear Receptor Subfamily 4, Group A, Member 1/biosynthesis
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Polymerase Chain Reaction
- Quercetin/analogs & derivatives
- Quercetin/pharmacology
- RNA, Messenger/genetics
- Rats
- Rats, Sprague-Dawley
- Tunica Intima/drug effects
Collapse
Affiliation(s)
- Yan Huo
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China; Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, 1020 Locust Street, Room 368G, Philadelphia 19107, USA
| | - Bing Yi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, 1020 Locust Street, Room 368G, Philadelphia 19107, USA
| | - Ming Chen
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, 1020 Locust Street, Room 368G, Philadelphia 19107, USA
| | - Nadan Wang
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, 1020 Locust Street, Room 368G, Philadelphia 19107, USA
| | - Pengguo Chen
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Cheng Guo
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.
| | - Jianxin Sun
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, 1020 Locust Street, Room 368G, Philadelphia 19107, USA.
| |
Collapse
|
15
|
YU XIN, LI ZHENG. MicroRNAs regulate vascular smooth muscle cell functions in atherosclerosis (Review). Int J Mol Med 2014; 34:923-33. [DOI: 10.3892/ijmm.2014.1853] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 05/30/2014] [Indexed: 11/06/2022] Open
|
16
|
A Novel Agent with Histone Deacetylase Inhibitory Activity Attenuates Neointimal Hyperplasia. Cardiovasc Drugs Ther 2014; 28:395-406. [DOI: 10.1007/s10557-014-6540-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
17
|
El receptor nuclear NOR-1 regula la activación de las células vasculares y el remodelado vascular en respuesta a estrés hemodinámico. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2014; 26:66-75. [DOI: 10.1016/j.arteri.2013.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 11/04/2013] [Indexed: 01/12/2023]
|
18
|
Hinze AV, Mayer P, Harst A, von Kügelgen I. P2X1 receptor-mediated inhibition of the proliferation of human coronary smooth muscle cells involving the transcription factor NR4A1. Purinergic Signal 2013; 9:677-86. [PMID: 23873636 PMCID: PMC3889386 DOI: 10.1007/s11302-013-9380-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 07/03/2013] [Indexed: 01/28/2023] Open
Abstract
Adenine nucleotides acting at P2X1 receptors are potent vasoconstrictors. Recently, we demonstrated that activation of adenosine A2B receptors on human coronary smooth muscle cells inhibits cell proliferation by the induction of the nuclear receptor subfamily 4, group A, member 1 (NR4A1; alternative notation Nur77). In the present study, we searched for long-term effects mediated by P2X1 receptors by analyzing receptor-mediated changes in cell proliferation and in the expression of NR4A1. Cultured human coronary smooth muscle cells were treated with selective receptor ligands. Effects on proliferation were determined by counting cells and measuring changes in impedance. The induction of transcription factors was assessed by qPCR. The P2X receptor agonist α,β-methylene-ATP and its analog β,γ-methylene-ATP inhibited cell proliferation by about 50 % after 5 days in culture with half-maximal concentrations of 0.3 and 0.08 μM, respectively. The effects were abolished or markedly attenuated by the P2X1 receptor antagonist NF449 (carbonylbis-imino-benzene-triylbis-(carbonylimino)tetrakis-benzene-1,3-disulfonic acid; 100 nM and 1 μM). α,β-methylene-ATP and β,γ-methylene-ATP applied for 30 min to 4 h increased the expression of NR4A1; NF449 blocked or attenuated this effect. Small interfering RNA directed against NR4A1 diminished the antiproliferative effects of α,β-methylene-ATP and β,γ-methylene-ATP. α,β-methylene-ATP (0.1 to 30 μM) decreased migration of cultured human coronary smooth muscle cells in a chamber measuring changes in impedance; NF449 blocked the effect. In conclusion, our results demonstrate for the first time that adenine nucleotides acting at P2X1 receptors inhibit the proliferation of human coronary smooth muscle cells via the induction of the early gene NR4A1.
Collapse
Affiliation(s)
- Annette Viktoria Hinze
- />Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
- />Department of Pharmacology and Toxicology, University of Bonn, 53105 Bonn, Germany
- />Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, 53105 Bonn, Germany
| | - Peter Mayer
- />Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Anja Harst
- />Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Ivar von Kügelgen
- />Department of Pharmacology and Toxicology, University of Bonn, 53105 Bonn, Germany
| |
Collapse
|
19
|
Luan J, Zhang D, Hao L, Qi L, Liu X, Guo H, Li C, Guo Y, Li T, Zhang Q, Zhai G. Preparation, characterization and pharmacokinetics of Amoitone B-loaded long circulating nanostructured lipid carriers. Colloids Surf B Biointerfaces 2013; 114:255-60. [PMID: 24211416 DOI: 10.1016/j.colsurfb.2013.10.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 10/09/2013] [Accepted: 10/11/2013] [Indexed: 11/26/2022]
Abstract
Amoitone B, chemically synthesized as the derivative of Cytosporone B, is a powerful agonist for Nur77 receptor. It has outstanding anticancer activity in vivo. However, the water-insolubility and short biological half-life lead to poor bioavailability, which limits its application. The aim of this study was to develop polyethylene glycol-coated Amoitone B-loaded nanostructured lipid carriers (AmB-PEG-NLC) for parenteral delivery of Amoitone B to prolong drug circulation time in body and enhance the bioavailability. AmB-PEG-NLC were prepared by emulsion-evaporation and low temperature-solidification method, while Amoitone B-loaded NLC (AmB-NLC) were also prepared as control. The characteristics of AmB-PEG-NLC and AmB-NLC such as particle size, zeta potential, entrapment efficiency and drug loading were investigated in detail. The mean particle size was about 200 nm and the zeta potential value was about -15 mV. The X-ray diffraction analysis demonstrated that Amoitone B was not in crystalline state in NLC (AmB-PEG-NLC and AmB-NLC). Drug release pattern with burst release initially and prolonged release afterwards was obtained in vitro for AmB-PEG-NLC. Furthermore, AmB-PEG-NLC exhibited prolonged MRT (mean residence time) and higher AUC (area under drug concentration-time curve) compared with AmB-NLC as well as Amoitone B solution. These results indicated that AmB-PEG-NLC could be a promising delivery system for Amoitone B to prolong the circulation time in body and thus improve its bioavailability.
Collapse
Affiliation(s)
- Jingjing Luan
- College of Pharmacy, Shandong University, Jinan 250012, PR China
| | - Dianrui Zhang
- College of Pharmacy, Shandong University, Jinan 250012, PR China.
| | - Leilei Hao
- College of Pharmacy, Shandong University, Jinan 250012, PR China
| | - Lisi Qi
- College of Pharmacy, Shandong University, Jinan 250012, PR China
| | - Xinquan Liu
- College of Pharmacy, Shandong University, Jinan 250012, PR China
| | - Hejian Guo
- College of Pharmacy, Shandong University, Jinan 250012, PR China
| | - Caiyun Li
- College of Pharmacy, Shandong University, Jinan 250012, PR China
| | - Yuanyuan Guo
- College of Pharmacy, Shandong University, Jinan 250012, PR China
| | - Tingting Li
- College of Pharmacy, Shandong University, Jinan 250012, PR China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100083, PR China
| | - Guangxi Zhai
- College of Pharmacy, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
20
|
Xiao G, Sun T, Songming C, Cao Y. NR4A1 enhances neural survival following oxygen and glucose deprivation: an in vitro study. J Neurol Sci 2013; 330:78-84. [PMID: 23663895 DOI: 10.1016/j.jns.2013.04.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 04/04/2013] [Accepted: 04/10/2013] [Indexed: 12/11/2022]
Abstract
A worldwide epidemic of stroke is exacting a huge level of patient suffering and social cost. The ischemia damage to neural cells and the associated permanent neural function loss are central to the pathophysiology of stroke. In the current study, we were endeavored to identify NR4A1, an orphan nuclear receptor as a novel protector for neural cells in an in vitro neural ischemia model. Our results showed that oxygen and glucose deprivation (OGD) dramatically induced primary culture neural cell apoptosis and NR4A1 expression at both protein and mRNA level. Furthermore, hyperexpression or knock-down of NR4A1 significantly ameliorated or exacerbated OGD induced neural damage as manifested by decreased or increased apoptotic rates and key apoptotic protein expression respectively. As part of effort to identify the underlying mechanism, we also found that survivin is highly inducible following OGD and is required for NR4A1 action in this scenario. Our data seemed to be logical extensions of previous observations showing that NR4As are highly inducible following focal cerebral ischemia. Of note, our results also demonstrated that NR4A1 induction in this scenario may be functionally important as well and targeting NR4A1 protein can be intriguing as part of the effort to develop novel therapeutic strategies for neural protection after stroke.
Collapse
Affiliation(s)
- Guodong Xiao
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215004, China
| | | | | | | |
Collapse
|
21
|
Li P, Liu Y, Yi B, Wang G, You X, Zhao X, Summer R, Qin Y, Sun J. MicroRNA-638 is highly expressed in human vascular smooth muscle cells and inhibits PDGF-BB-induced cell proliferation and migration through targeting orphan nuclear receptor NOR1. Cardiovasc Res 2013; 99:185-93. [PMID: 23554459 DOI: 10.1093/cvr/cvt082] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIMS Aberrant vascular smooth muscle cell (VSMC) proliferation and migration contribute significantly to the development of vascular pathologies, such as atherosclerosis and restenosis. MicroRNAs have recently emerged as critical modulators in cellular processes and the purpose of this study is to identify novel miRNA regulators implicated in human aortic VSMC proliferation and migration. METHODS AND RESULTS To identify miRNAs that are differentially expressed in human VSMCs, we performed miRNA microarray analysis in human aortic smooth muscle cells (SMCs) at different time points after platelet-derived growth factor (PDGF) stimulation. Here, we identified microRNA-638 (miR-638) as a transcript that was one of the most significantly down-regulated in human VSMCs after PDGF stimulation. Furthermore, we confirmed, by Quantitative RT-PCR, that miR-638 is highly expressed in human VSMCs, and its expression is markedly down-regulated in a dose- and time-dependent manner upon PDGF treatment. Consistent with a critical role in SMC proliferation, we found that miR-638 expression was significantly up-regulated in human VSMCs cultured in differentiation medium, a condition that inhibits SMC proliferation. Furthermore, we identified the orphan nuclear receptor NOR1 as a downstream target gene product of miR-638 and down-regulation of NOR1 is critical for miR-638-mediated inhibitory effects on PDGF-induced cyclin D1 expression, cell proliferation, and migration in human aortic SMCs. CONCLUSION These results indicate that miR-638 is a key molecule in regulating human VSMC proliferation and migration by targeting the NOR1/cyclin D pathway and suggest that specific modulation of miR-638 in human VSMCs may represent an attractive approach for the treatment of proliferative vascular diseases.
Collapse
Affiliation(s)
- Pan Li
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Rodríguez-Calvo R, Guadall A, Calvayrac O, Navarro MA, Alonso J, Ferrán B, de Diego A, Muniesa P, Osada J, Rodríguez C, Martínez-González J. Over-expression of neuron-derived orphan receptor-1 (NOR-1) exacerbates neointimal hyperplasia after vascular injury. Hum Mol Genet 2013; 22:1949-59. [PMID: 23390133 DOI: 10.1093/hmg/ddt042] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
We have previously shown that NOR-1 (NR4A3) modulates the proliferation and survival of vascular cells in culture. However, in genetically modified animal models, somewhat conflicting results have been reported concerning the involvement of NOR-1 in neointimal formation after vascular injury. The aim of this study was to generate a transgenic mouse model over-expressing NOR-1 in smooth muscle cells (SMCs) and assess the consequence of a gain of function of this receptor on intimal hyperplasia after vascular injury. The transgene construct (SM22-NOR1) was prepared by ligating the full-length human NOR-1 cDNA (hNOR-1) and a mouse SM22α minimal promoter able to drive NOR-1 expression to SMC. Two founders were generated and two stable transgenic mouse lines (TgNOR-1) were established by backcrossing the transgene-carrying founders with C57BL/6J mice. Real-time PCR and immunohistochemistry confirmed that hNOR-1 was mainly targeted to vascular beds such as aorta and carotid arteries, and was similar in both transgenic lines. Vascular SMC from transgenic animals exhibit increased NOR-1 transcriptional activity (assessed by electrophoretic mobility shift assay and luciferase assays), increased mitogenic activity (determined by [(3)H]-thymidine incorporation; 1.58-fold induction, P < 0.001) and increased expression of embryonic smooth muscle myosin heavy chain (SMemb) than wild-type cells from control littermates. Using the carotid artery ligation model, we show that neointima formation was increased in transgenic versus wild-type mice (2.36-fold induction, P < 0.01). Our in vivo data support a role for NOR-1 in VSMC proliferation and vascular remodelling. This NOR-1 transgenic mouse could be a useful model to study fibroproliferative vascular diseases.
Collapse
|
23
|
Adenosine A3 receptor-induced proliferation of primary human coronary smooth muscle cells involving the induction of early growth response genes. J Mol Cell Cardiol 2012; 53:639-45. [DOI: 10.1016/j.yjmcc.2012.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 08/03/2012] [Accepted: 08/04/2012] [Indexed: 11/22/2022]
|
24
|
Abstract
A number of nuclear receptors are involved in maintenance of normal vessel wall physiology as well as in pathophysiological processes such as atherosclerosis, restenosis and remodelling. Recent studies revealed a previously unrecognized function of the NR4A subfamily of nuclear receptors as key regulatory proteins in vascular disease. The NR4A subfamily comprises the members Nur77, Nurr1 and NOR-1 and in the current review a comprehensive overview is given of the data supporting functional involvement of these nuclear receptors in three major cell types in vascular (patho)physiology; endothelial cells, smooth muscle cells and monocytes-macrophages.
Collapse
Affiliation(s)
- Claudia M van Tiel
- Academic Medical Center, K1-113, University of Amsterdam, Meibergdreef 15, 1105AZ Amsterdam, The Netherlands
| | | |
Collapse
|
25
|
Hao L, Wang X, Zhang D, Xu Q, Song S, Wang F, Li C, Guo H, Liu Y, Zheng D, Zhang Q. Studies on the preparation, characterization and pharmacokinetics of Amoitone B nanocrystals. Int J Pharm 2012; 433:157-64. [PMID: 22579996 DOI: 10.1016/j.ijpharm.2012.05.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 04/27/2012] [Accepted: 05/03/2012] [Indexed: 11/18/2022]
Abstract
Amoitone B, as a new derivative of cytosporone B, has been proved to be a natural agonist for Nur77. It exhibits remarkable anticancer activity in vivo and has the potential to be a therapeutic agent for cancer treatment. However, the poor solubility and dissolution rate result in low therapeutic index for injection and low bioavailability for oral administration, therefore limiting its application. In order to magnify the clinical use of Amoitone B, nanocrystal was selected as an application technology to solve the above problems. In this study, the optimized Amoitone B nanocrystals with small and uniform particle size were successfully prepared by microfluidization method and investigated by morphology, size distribution, and zeta potential. The differential scanning calorimetry (DSC) and X-ray diffraction (XRD) confirmed there was no crystalline state changed in the size reduction process. For Amoitone B nanocrystals, an accelerated dissolution velocity and increased saturation solubility were achieved in vitro and a markedly different pharmacokinetic property in vivo was exhibited with retarded clearance and magnified AUC compared with Amoitone B solution. These results implied that developing Amoitone B as nanocrystals is a promising choice for intravenous delivery and further application for cancer therapy.
Collapse
Affiliation(s)
- Leilei Hao
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Hanna RN, Shaked I, Hubbeling HG, Punt JA, Wu R, Herrley E, Zaugg C, Pei H, Geissmann F, Ley K, Hedrick CC. NR4A1 (Nur77) deletion polarizes macrophages toward an inflammatory phenotype and increases atherosclerosis. Circ Res 2011; 110:416-27. [PMID: 22194622 DOI: 10.1161/circresaha.111.253377] [Citation(s) in RCA: 344] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
RATIONALE NR4A1 (Nur77) is a nuclear receptor that is expressed in macrophages and within atherosclerotic lesions, yet its function in atherosclerosis is unknown. OBJECTIVE Nur77 regulates the development of monocytes, particularly patrolling Ly6C(-) monocytes that may be involved in resolution of inflammation. We sought to determine how absence of nuclear receptor subfamily 4, group A, member 1 (NR4A1) in hematopoietic cells affected atherosclerosis development. METHODS AND RESULTS Nur77(-/-) chimeric mice on a Ldlr(-/-) background showed a 3-fold increase in atherosclerosis development when fed a Western diet for 20 weeks, despite having a drastic reduction in Ly6C(-) patrolling monocytes. In a second model, mice deficient in both Nur77 and ApoE (ApoE(-/-)Nur77(-/-)) also showed increased atherosclerosis after 11 weeks of Western diet. Atherosclerosis was associated with a significant change in macrophage polarization toward a proinflammatory phenotype, with high expression of tumor necrosis factor-α and nitric oxide and low expression of Arginase-I. Moreover, we found increased expression of toll-like receptor 4 mRNA and protein in Nur77(-/-) macrophages as well as increased phosphorylation of the p65 subunit of NFκB. Inhibition of NFκB activity blocked excess activation of Nur77(-/-) macrophages. CONCLUSIONS We conclude that the absence of Nur77 in monocytes and macrophages results in enhanced toll-like receptor signaling and polarization of macrophages toward a proinflammatory M1 phenotype. Despite having fewer monocytes, Nur77(-/-) mice developed significant atherosclerosis when fed a Western diet. These studies indicate that Nur77 is a novel target for modulating the inflammatory phenotype of monocytes and macrophages and may be important for regulation of atherogenesis.
Collapse
Affiliation(s)
- Richard N Hanna
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Johnson MM, Michelhaugh SK, Bouhamdan M, Schmidt CJ, Bannon MJ. The Transcription Factor NURR1 Exerts Concentration-Dependent Effects on Target Genes Mediating Distinct Biological Processes. Front Neurosci 2011; 5:135. [PMID: 22194714 PMCID: PMC3243378 DOI: 10.3389/fnins.2011.00135] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 11/21/2011] [Indexed: 01/24/2023] Open
Abstract
The transcription factor NURR1 plays a pivotal role in the development and maintenance of neurotransmitter phenotype in midbrain dopamine neurons. Conversely, decreased NURR1 expression is associated with a number of dopamine-related CNS disorders, including Parkinson's disease and drug addiction. In order to better understand the nature of NURR1-responsive genes and their potential roles in dopamine neuron differentiation and survival, we used a human neural cellular background (SK-N-AS cells) in which to generate a number of stable clonal lines with graded NURR1 gene expression that approximated that seen in DA cell-rich human substantia nigra. Gene expression profiling data from these NURR1-expressing clonal lines were validated by quantitative RT-PCR and subjected to bioinformatic analyses. The present study identified a large number of NURR1-responsive genes and demonstrated the potential importance of concentration-dependent NURR1 effects in the differential regulation of distinct NURR1 target genes and biological pathways. These data support the promise of NURR1-based CNS therapeutics for the neuroprotection and/or functional restoration of DA neurons.
Collapse
Affiliation(s)
- Magen M Johnson
- Department of Pharmacology, Wayne State University School of Medicine Detroit, MI, USA
| | | | | | | | | |
Collapse
|
28
|
Kurakula K, van der Wal E, Geerts D, van Tiel CM, de Vries CJM. FHL2 protein is a novel co-repressor of nuclear receptor Nur77. J Biol Chem 2011; 286:44336-43. [PMID: 22049082 DOI: 10.1074/jbc.m111.308999] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The three members of the NR4A orphan nuclear receptor subfamily Nur77, Nurr1, and NOR-1, regulate a variety of biological functions including vascular disease and metabolism. In this study, we identified Four and a half LIM domains protein-2 (FHL2) as a novel interacting protein of NR4A nuclear receptors by yeast two-hybrid screen and co-immunoprecipitation studies. Each of the four LIM domains of FHL2 can bind Nur77, and both the amino-terminal domain and the DNA binding domain of Nur77 are involved in the interaction between FHL2 and Nur77. FHL2 represses Nur77 transcriptional activity in a dose-dependent manner, and short hairpin RNA-mediated knockdown of FHL2 results in increased Nur77 transcriptional activity. ChIP experiments on the enolase3 promoter revealed that FHL2 inhibits the association of Nur77 with DNA. FHL2 is highly expressed in human endothelial and smooth muscle cells, but not in monocytes or macrophages. To substantiate functional involvement of FHL2 in smooth muscle cell physiology, we demonstrated that FHL2 overexpression increases the growth of these cells, whereas FHL2 knockdown results in reduced DNA synthesis. Collectively, these studies suggest that association of FHL2 with Nur77 plays a pivotal role in vascular disease.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Medical Biochemistry, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
29
|
Microarray expression analysis in delayed cardioprotection: the effect of exercise, AICAR, or metformin and the possible role of AMP-activated protein kinase (AMPK). Mol Cell Biochem 2011; 360:353-62. [PMID: 21964537 DOI: 10.1007/s11010-011-1075-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 09/16/2011] [Indexed: 12/13/2022]
Abstract
AMP-activated protein kinase (AMPK) is an enzyme which may be involved in cardioprotective mechanisms in the ischemic heart. Exercise, AICAR, and metformin, all known activators of AMPK, induce delayed cardioprotection which protects the heart against ischemia-reperfusion injury. The objective was to determine the effect of exercise, AICAR, and metformin on gene expression profile and to demonstrate possible interactions in different genes and functions. Rats were divided into either an exercise, AICAR, metformin, or control group. 3, 12, and 24 h after either a single bout of exercise training, a single injection of AICAR or a single dose of metformin, hearts were removed and gene expression profiles were analyzed in tissue from the left ventricle using Affymetrix gene chip probe arrays. Ingenuity Pathway Analysis (IPA) tool was used to analyze the regulated genes for relevant functions and diseases. Each gene chip identified up to 30,000 different probesets of which Ingenuity identified approximately up to 12,000 genes. A total of 147, 304, and 114 different genes in the left ventricle whose expressions were altered >2.0-fold were identified in the exercise, AICAR, and metformin group, respectively. Seventy eight different genes were overlapping the exercise and AICAR group at 24 h. Ingenuity identified six overlapping genes between the exercise, AICAR, and metformin groups including NR4A3, TNFRSF12A, HBB, PENK, PAP, and MAP4K4. IPA software revealed an overabundance of focus molecules in all three intervention groups involving functions related to cell death, cellular growth and proliferation, gene expression and cancer. Exercise, AICAR, and metformin regulate several genes in the rat myocardium with the majority of overlapping genes observed in the exercise and AICAR group. Changes in gene programming mainly involved inflammatory and opioid systems recognized as cardioprotective pathways. Some of these genes may represent possible candidate genes involved in the molecular mechanisms of AMPK-induced delayed PC.
Collapse
|
30
|
Gizard F, Zhao Y, Findeisen HM, Qing H, Cohn D, Heywood EB, Jones KL, Nomiyama T, Bruemmer D. Transcriptional regulation of S phase kinase-associated protein 2 by NR4A orphan nuclear receptor NOR1 in vascular smooth muscle cells. J Biol Chem 2011; 286:35485-35493. [PMID: 21868379 DOI: 10.1074/jbc.m111.295840] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Members of the NR4A subgroup of the nuclear hormone receptor superfamily have emerged as key transcriptional regulators of proliferation and inflammation. NOR1 constitutes a ligand-independent transcription factor of this subgroup and induces cell proliferation; however, the transcriptional mechanisms underlying this mitogenic role remain to be defined. Here, we demonstrate that the F-box protein SKP2 (S phase kinase-associated protein 2), the substrate-specific receptor of the ubiquitin ligase responsible for the degradation of p27(KIP1) through the proteasome pathway, constitutes a direct transcriptional target for NOR1. Mitogen-induced Skp2 expression is silenced in vascular smooth muscle cells (VSMC) isolated from Nor1-deficient mice or transfected with Nor1 siRNA. Conversely, adenovirus-mediated overexpression of NOR1 induces Skp2 expression in VSMC and decreases protein abundance of its target p27. Transient transfection experiments establish that NOR1 transactivates the Skp2 promoter through a nerve growth factor-induced clone B response element (NBRE). Electrophoretic mobility shift and chromatin immunoprecipitation assays further revealed that NOR1 is recruited to this NBRE site in the Skp2 promoter in response to mitogenic stimulation. In vivo Skp2 expression is increased during the proliferative response underlying neointima formation, and this transcriptional induction depends on the expression of NOR1. Finally, we demonstrate that overexpression of Skp2 rescues the proliferative arrest of Nor1-deficient VSMC. Collectively, these results characterize Skp2 as a novel NOR1-regulated target gene and detail a previously unrecognized transcriptional cascade regulating mitogen-induced VSMC proliferation.
Collapse
Affiliation(s)
- Florence Gizard
- Saha Cardiovascular Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Yue Zhao
- Saha Cardiovascular Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536; Graduate Center for Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Hannes M Findeisen
- Saha Cardiovascular Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Hua Qing
- Saha Cardiovascular Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Dianne Cohn
- Saha Cardiovascular Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Elizabeth B Heywood
- Saha Cardiovascular Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Karrie L Jones
- Saha Cardiovascular Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Takashi Nomiyama
- Department of Endocrinology and Diabetes Mellitus, School of Medicine, Fukuoka University, Fukuoka City, Fukuoka 814-0180, Japan
| | - Dennis Bruemmer
- Saha Cardiovascular Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536; Graduate Center for Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky 40536.
| |
Collapse
|
31
|
Heard M, Maina CV, Morehead BE, Hoener MC, Nguyen TQ, Williams CC, Rowan BG, Gissendanner CR. A functional NR4A nuclear receptor DNA-binding domain is required for organ development in Caenorhabditis elegans. Genesis 2011; 48:485-91. [PMID: 20506374 DOI: 10.1002/dvg.20646] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
NR4A nuclear receptors are a diverse group of orphan nuclear receptors with critical roles in regulating cell proliferation and cell differentiation. The ortholog of the NR4A nuclear receptor in Caenorhabditis elegans, NHR-6, also has a role in cell proliferation and cell differentiation during organogenesis of the spermatheca. Here we show that NHR-6 is able to bind the canonical NR4A monomer response element and can transactivate from this site in mammalian HEK293 cells. Using a functional GFP-tagged NHR-6 fusion, we also demonstrate that NHR-6 is nuclear localized during development of the spermatheca. Mutation of the DNA-binding domain of NHR-6 abolishes its activity in genetic rescue assays, demonstrating a requirement for the DNA-binding domain. This study represents the first genetic demonstration of an in vivo requirement for an NR4A nuclear receptor DNA-binding domain in a whole organism.
Collapse
|
32
|
Wang SCM, Myers SA, Eriksson NA, Fitzsimmons RL, Muscat GEO. Nr4a1 siRNA expression attenuates α-MSH regulated gene expression in 3T3-L1 adipocytes. Mol Endocrinol 2011; 25:291-306. [PMID: 21239615 DOI: 10.1210/me.2010-0231] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Several recent investigations have underscored the growing role of melanocortin signaling in the peripheral regulation of lipid, glucose, and energy homeostasis. In addition, the melanocortins play a critical role in the central control of satiety. These observations, and the latest reports highlighting the emerging role of the nuclear hormone receptor (NR) 4A subgroup in metabolism, have prompted us to investigate the cross talk between [Nle(4), d-Phe(7)] (NDP)-α-MSH and Nr4a signaling in adipose. We have shown that NDP-MSH strikingly and preferentially induces the expression of the NR4A subgroup (but not any other members of the NR superfamily) in differentiated 3T3-L1 adipocytes. Utilization of quantitative PCR on custom-designed metabolic TaqMan low-density arrays identified the concomitant and marked induction of the mRNAs encoding Il-6, Cox2, Pdk4, and Pck-1 after NDP-MSH treatment. Similar experiments demonstrated that the mRNA expression profile induced by cAMP and NDP-MSH treatment displayed unique but also overlapping properties and suggested that melanocortin-mediated induction of gene expression involves cAMP-dependent and -independent signaling. Nr4a1/Nur77 small interfering RNA (siRNA) expression suppressed NDP-MSH-mediated induction of Nr4a1/Nur77 and Nr4a3/Nor-1 (but not Nr4a2/Nurr1). Moreover, expression of the siRNA-attenuated NDP-MSH mediated induction of the mRNAs encoding Il-6, Cox2/Ptgs2, and Pck-1 expression. In addition, Nur77 siRNA expression attenuated NDP-MSH-mediated glucose uptake. In vivo, ip administration of NDP-MSH to C57 BL/6J (male) mice significantly induced the expression of the mRNA encoding Nur77 and increased IL-6, Cox2, Pck1, and Pdk4 mRNA expression in (inguinal) adipose tissue. We conclude that Nur77 expression is necessary for MSH-mediated induction of gene expression in differentiated adipocytes. Furthermore, this study demonstrates cross talk between MSH and Nr4a signaling in adipocytes.
Collapse
Affiliation(s)
- S-C Mary Wang
- Obesity Research Centre, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | |
Collapse
|
33
|
Mayer P, Hinze AV, Harst A, von Kügelgen I. A₂B receptors mediate the induction of early genes and inhibition of arterial smooth muscle cell proliferation via Epac. Cardiovasc Res 2010; 90:148-56. [PMID: 21109603 DOI: 10.1093/cvr/cvq371] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
AIMS Extracellular adenosine and adenine nucleotides play important roles in the regulation of the blood vessel tonus and platelet aggregation. Less is known about the effects of these extracellular signalling molecules on gene expression in vascular smooth muscle cells involved in long-term vascular effects. In the present study, we therefore searched for adenosine-induced changes in the expression of early genes in cultured human coronary artery smooth muscle cells (HCASMCs). METHODS AND RESULTS Whole-genome DNA array hybridization revealed that adenosine induced a set of early genes including the nuclear receptor subfamily 4, group A, member 1 (NR4A1/Nur77/TR3). The pattern of the effects of adenosine on gene expression resembles the change in expression induced by the direct activator of adenylate cyclase forskolin. Real-time reverse-transcriptase PCR confirmed that adenosine and its analogue N-ethyl-carboxamidoadenosine elicited a strong induction of NR4A1. These effects were markedly attenuated by A(2B) receptor antagonists including 8-[4-(4-benzylpiperazide-1-sulfonyl)phenyl]-1-propylxanthine (PSB-601) and were mimicked by a cyclic AMP (cAMP) analogue [8-(4-chlorophenylthio)-2'-O-methyl-cAMP, 8CPT] acting on the exchange protein activated by cAMP (Epac). Long-term experiments over 5 days showed that 2-chloroadenosine decreased cell proliferation in the presence of platelet-derived growth factor. This effect of 2-chloroadenosine was also attenuated by PSB-601 and mimicked by 8CPT. Treatment with small interfering RNA directed against NR4A1 attenuated the inhibitory effect of 8CPT on proliferation. CONCLUSIONS In summary, our results demonstrate the operation of adenosine A₂(B) receptors mediating an early induction of NR4A1 and a decrease in cell proliferation via the cAMP/Epac pathway in HCASMCs.
Collapse
Affiliation(s)
- Peter Mayer
- Federal Institute for Drugs and Medical Devices, Bonn, Germany.
| | | | | | | |
Collapse
|
34
|
Huang W, Glass CK. Nuclear receptors and inflammation control: molecular mechanisms and pathophysiological relevance. Arterioscler Thromb Vasc Biol 2010; 30:1542-9. [PMID: 20631355 DOI: 10.1161/atvbaha.109.191189] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tissue inflammation is a tightly regulated process that normally serves to recruit the immune system to sites of infection and injury and to facilitate tissue repair processes. When an inflammatory state is excessive or prolonged, local and systemic damage to host tissues can result in loss of normal physiological functions. Here, we briefly review recent studies that advance our understanding of signaling pathways involved in initiation of inflammatory responses at the level of transcription and counterregulation of these pathways by selected members of the nuclear receptor superfamily. Studies of the intersection of nuclear receptors and inflammation have revealed mechanisms of positive and negative transcriptional control that may provide new targets for pharmacological intervention in chronic diseases, such as atherosclerosis.
Collapse
Affiliation(s)
- Wendy Huang
- Department of Cellular and Molecular Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | |
Collapse
|
35
|
Kinter J, Broglio L, Steck AJ, Tolnay M, Fuhr P, Latov N, Kalbermatten D, Sinnreich M, Schaeren-Wiemers N, Renaud S. Gene expression profiling in nerve biopsy of vasculitic neuropathy. J Neuroimmunol 2010; 225:184-9. [DOI: 10.1016/j.jneuroim.2010.05.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 05/04/2010] [Accepted: 05/05/2010] [Indexed: 10/19/2022]
|
36
|
Shao Q, Shen LH, Hu LH, Pu J, Qi MY, Li WQ, Tian FJ, Jing Q, He B. Nuclear receptor Nur77 suppresses inflammatory response dependent on COX-2 in macrophages induced by oxLDL. J Mol Cell Cardiol 2010; 49:304-11. [DOI: 10.1016/j.yjmcc.2010.03.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2010] [Revised: 03/29/2010] [Accepted: 03/30/2010] [Indexed: 12/11/2022]
|
37
|
Park SJ, Kufareva I, Abagyan R. Improved docking, screening and selectivity prediction for small molecule nuclear receptor modulators using conformational ensembles. J Comput Aided Mol Des 2010; 24:459-71. [PMID: 20455005 PMCID: PMC2881208 DOI: 10.1007/s10822-010-9362-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Accepted: 04/26/2010] [Indexed: 12/26/2022]
Abstract
Nuclear receptors (NRs) are ligand dependent transcriptional factors and play a key role in reproduction, development, and homeostasis of organism. NRs are potential targets for treatment of cancer and other diseases such as inflammatory diseases, and diabetes. In this study, we present a comprehensive library of pocket conformational ensembles of thirteen human nuclear receptors (NRs), and test the ability of these ensembles to recognize their ligands in virtual screening, as well as predict their binding geometry, functional type, and relative binding affinity. 157 known NR modulators and 66 structures were used as a benchmark. Our pocket ensemble library correctly predicted the ligand binding poses in 94% of the cases. The models were also highly selective for the active ligands in virtual screening, with the areas under the ROC curves ranging from 82 to a remarkable 99%. Using the computationally determined receptor-specific binding energy offsets, we showed that the ensembles can be used for predicting selectivity profiles of NR ligands. Our results evaluate and demonstrate the advantages of using receptor ensembles for compound docking, screening, and profiling.
Collapse
Affiliation(s)
- So-Jung Park
- Department of Molecular Biology, The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
38
|
Shinone K, Tomita M, Inoue H, Nakagawa Y, Ikemura M, Nata M. Molecular-biological analysis of the effect of methamphetamine on the heart in restrained mice. Leg Med (Tokyo) 2010; 12:79-83. [DOI: 10.1016/j.legalmed.2010.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 11/13/2009] [Accepted: 01/06/2010] [Indexed: 11/16/2022]
|
39
|
Bonta PI, Matlung HL, Vos M, Peters SLM, Pannekoek H, Bakker ENTP, de Vries CJM. Nuclear receptor Nur77 inhibits vascular outward remodelling and reduces macrophage accumulation and matrix metalloproteinase levels. Cardiovasc Res 2010; 87:561-8. [PMID: 20189954 DOI: 10.1093/cvr/cvq064] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS Structural adaptation of the vessel wall in response to sustained alterations in haemodynamic forces is known as vascular remodelling. Detailed knowledge on the mechanism underlying this vascular response is limited, and we aimed to study the function of Nur77 in smooth muscle cells (SMCs) in arterial remodelling. METHODS AND RESULTS Carotid artery ligation in mice results in flow-induced, outward remodelling of the contralateral carotid artery, and we observed enhanced Nur77 expression during this process. Transgenic mice that express Nur77 or its dominant-negative variant, denoted as 'DeltaTA' in arterial SMCs, were exposed to carotid artery ligation, and after 4 weeks pressure-diameter relationships were measured. Structural outward remodelling is inhibited in Nur77-transgenic mice when compared with wild-type and DeltaTA-transgenic mice. The key determinants of remodelling vascular tone and macrophage accumulation were studied. No difference in contractile and relaxant responses was detected in isolated aorta, carotid, and mesenteric artery segments between transgenic and wild-type mice. SMC-specific overexpression of Nur77 in transgenic mice reduced macrophage accumulation and repressed matrix metalloproteinase (MMP)1 and MMP9 expression at early time points. MMP2 protein expression was reduced in Nur77-transgenic mice, whereas in DeltaTA-transgenic mice MMP2 expression was increased. CONCLUSION Nur77 is induced during outward remodelling and inhibits this vascular adaptation in mice. Nur77-mediated inhibition of arterial remodelling involves a reduction in both macrophage accumulation and MMP expression levels.
Collapse
Affiliation(s)
- Peter I Bonta
- Department of Medical Biochemistry, Academic Medical Center K1-113, University of Amsterdam, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
40
|
Serotonin 5-HT2C receptor-independent expression of hypothalamic NOR1, a novel modulator of food intake and energy balance, in mice. Biochem Biophys Res Commun 2009; 386:311-5. [PMID: 19523439 DOI: 10.1016/j.bbrc.2009.06.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Accepted: 06/06/2009] [Indexed: 11/22/2022]
Abstract
NOR1, Nur77 and Nurr1 are orphan nuclear receptors and members of the NR4A subfamily. Here, we report that the expression of hypothalamic NOR1 was remarkably decreased in mildly obese beta-endorphin-deficient mice and obese db/db mice with the leptin receptor mutation, compared with age-matched wild-type mice, whereas there were no genotypic differences in the expression of hypothalamic Nur77 or Nurr1 in these animals. The injection of NOR1 siRNA oligonucleotide into the third cerebral ventricle significantly suppressed food intake and body weight in mice. On the other hand, the decreases in hypothalamic NOR1 expression were not found in non-obese 5-HT2C receptor-deficient mice. Moreover, systemic administration of m-chlorophenylpiperazine (mCPP), a 5-HT2C/1B receptor agonist, had no effect on hypothalamic NOR1 expression, while suppressing food intake in beta-endorphin-deficient mice. These findings suggest that 5-HT2C receptor-independent proopiomelanocortin-derived peptides regulate the expression of hypothalamic NOR1, which is a novel modulator of feeding behavior and energy balance.
Collapse
|
41
|
Kozlova T, Lam G, Thummel CS. Drosophila DHR38 nuclear receptor is required for adult cuticle integrity at eclosion. Dev Dyn 2009; 238:701-7. [PMID: 19235727 DOI: 10.1002/dvdy.21860] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
DHR38 is the only Drosophila member of the NR4A subclass of vertebrate nuclear receptors, which have been implicated in multiple biological pathways, including neuronal function, apoptosis, and metabolism. Although an earlier study identified three point mutations in DHR38, none of these were shown to be a null allele for the locus, leaving it unclear whether a complete loss of DHR38 function might uncover novel roles for the receptor. Here we show that a specific DHR38 null allele, DHR38(Y214), leads to fully penetrant pharate adult lethality, similar to the most severe phenotype associated with the EMS-induced mutations. DHR38(Y214) mutants display minor effects on ecdysone-regulated transcription at the onset of metamorphosis. In contrast, cuticle gene expression is significantly reduced in DHR38(Y214) mutant pupae. These studies define the essential functions of DHR38 and provide a genetic context for further characterization of its roles during development.
Collapse
Affiliation(s)
- Tatiana Kozlova
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah 84112-5330, USA
| | | | | |
Collapse
|
42
|
Kardys I, van Tiel CM, de Vries CJ, Pannekoek H, Uitterlinden AG, Hofman A, Witteman JC, de Maat MP. Haplotypes of theNR4A2/NURR1gene and cardiovascular disease: The Rotterdam Study. Hum Mutat 2009; 30:417-23. [DOI: 10.1002/humu.20902] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
43
|
Abstract
It is well known that the steroid hormone glucocorticoid and its nuclear receptor regulate the inflammatory process, a crucial component in the pathophysiological process related to human diseases that include atherosclerosis, obesity and type II diabetes, inflammatory bowel disease, Alzheimer's disease, multiple sclerosis, and liver tumors. Growing evidence demonstrates that orphan and adopted orphan nuclear receptors, such as peroxisome proliferator-activated receptors, liver x receptors, the farnesoid x receptor, NR4As, retinoid x receptors, and the pregnane x receptor, regulate the inflammatory and metabolic profiles in a ligand-dependent or -independent manner in human and animal models. This review summarizes the regulatory roles of these nuclear receptors in the inflammatory process and the underlying mechanisms.
Collapse
Affiliation(s)
- Kun Wang
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW The nuclear orphan receptors Nur77 (NR4A1), Nurr1 (NR4A2) and NOR-1 (NR4A3) are known to be involved in T-cell apoptosis, brain development, and the hypothalamic-pituitary-adrenal axis. Here, we review our current understanding of the NR4A nuclear receptors in processes that are relevant to vascular disease. RECENT FINDINGS NR4A nuclear receptors have recently been described to play a role in metabolism by regulating gluconeogenesis, lipolysis, energy expenditure, and adipogenesis. The function of NR4A nuclear receptors has also extensively been investigated in cells crucial in vascular lesion formation, such as macrophages, endothelial cells and smooth muscle cells. SUMMARY The involvement of NR4A nuclear receptors in both metabolism and in processes in the vessel wall supports a substantial role for NR4A nuclear receptors in the development of vascular disease.
Collapse
Affiliation(s)
- Thijs W H Pols
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|