1
|
Terrian L, Thompson JM, Bowman DE, Panda V, Contreras GA, Rockwell C, Sather L, Fink GD, Lauver DA, Nault R, Watts SW, Bhattacharya S. Single-nucleus analysis of thoracic perivascular adipose tissue reveals critical changes in cell composition, communication, and gene regulatory networks induced by a high fat hypertensive diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.636878. [PMID: 39990347 PMCID: PMC11844537 DOI: 10.1101/2025.02.13.636878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, with hypertension being its primary causal factor. Most blood vessels are surrounded by perivascular adipose tissue (PVAT), which regulates blood vessel tone through the secretion of vasoactive factors. PVAT is recognized as a key mediator of vascular function and dysfunction in CVD, although the underlying mechanisms remain poorly understood. To investigate PVAT's mechanistic role in hypertension, we performed single nucleus RNA-Sequencing analysis of thoracic aortic PVAT from Dahl SS rats fed a high-fat, hypertensive diet. Computational analysis revealed extensive diet-induced changes in cell-type composition, cell-type specific gene expression, cell-cell communication pathways, and intracellular gene regulatory networks within PVAT. Furthermore, we identified key transcription factors mediating these networks and demonstrated through virtual knock-out experiments that these factors could serve as potential therapeutic targets for preventing or reversing PVAT's hypertensive state.
Collapse
Affiliation(s)
- Leah Terrian
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Denotes individuals contributed equally as first authors to this work
| | - Janice M. Thompson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Denotes individuals contributed equally as first authors to this work
| | - Derek E. Bowman
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Vishal Panda
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - G. Andres Contreras
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - Cheryl Rockwell
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Lisa Sather
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Gregory D. Fink
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - D. Adam Lauver
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Rance Nault
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Stephanie W. Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Denotes lead investigators/funding
| | - Sudin Bhattacharya
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Denotes lead investigators/funding
| |
Collapse
|
2
|
Sheethal G, Verma A, Mall R, Parsa KV, Tokala RK, Bynigeri R, Pondugala PK, Vemula K, Sai Latha S, Sowpati DT, Singh SS, Rao GV, Talukdar R, Kanneganti TD, Reddy DN, Sasikala M. Modulation of Nuclear Receptor 4A1 Expression Improves Insulin Secretion in a Mouse Model of Chronic Pancreatitis. Pancreas 2024; 53:e760-e773. [PMID: 38710022 DOI: 10.1097/mpa.0000000000002370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
OBJECTIVES Diabetes secondary to chronic pancreatitis (CP) presents clinical challenges due to lack of understanding on factor(s) triggering insulin secretory defects. Therefore, we aimed to delineate the molecular mechanism of β-cell dysfunction in CP. MATERIALS AND METHODS Transcriptomic analysis was conducted to identify endocrine-specific receptor expression in mice and human CP on microarray. The identified receptor (NR4A1) was overexpressed in MIN6 cells using PEI linear transfection. RNA-Seq analysis of NR4A1-overexpressed (OE) MIN6 cells on NovaSeq6000 identified aberrant metabolic pathways. Upstream trigger for NR4A1OE was studied by InBio Discover and cytokine exposure, whereas downstream effect was examined by Fura2 AM-based fluorimetric and imaging studies. Mice with CP were treated with IFN-γ-neutralizing monoclonal antibodies to assess NR4A1 expression and insulin secretion. RESULTS Increased expression of NR4A1 associated with decreased insulin secretion in islets (humans: controls 9 ± 0.2, CP 3.7 ± 0.2, mice: controls 8.5 ± 0.2, CP 2.1 ± 0.1 μg/L). NR4A1OE in MIN6 cells (13.2 ± 0.1) showed reduction in insulin secretion (13 ± 5 to 0.2 ± 0.1 μg/mg protein per minute, P = 0.001) and downregulation of calcium and cAMP signaling pathways. IFN-γ was identified as upstream signal for NR4A1OE in MIN6. Mice treated with IFN-γ-neutralizing antibodies showed decreased NR4A1 expression 3.4 ± 0.11-fold ( P = 0.03), showed improved insulin secretion (4.4 ± 0.2-fold, P = 0.01), and associated with increased Ca 2+ levels (2.39 ± 0.06-fold, P = 0.009). CONCLUSIONS Modulating NR4A1 expression can be a promising therapeutic strategy to improve insulin secretion in CP.
Collapse
Affiliation(s)
| | - Archana Verma
- CSIR-CCMB, Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Raghvendra Mall
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN
| | - Kishore Vl Parsa
- Centre for Innovation in Molecular and Pharmaceutical Sciences, Dr Reddy's Institute of Life Sciences
| | | | - Ratnakar Bynigeri
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN
| | | | | | - S Sai Latha
- Centre for Innovation in Molecular and Pharmaceutical Sciences, Dr Reddy's Institute of Life Sciences
| | - Divya Tej Sowpati
- CSIR-CCMB, Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | - G V Rao
- Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | - Rupjyoti Talukdar
- Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | | | - D Nageshwar Reddy
- Asian Institute of Gastroenterology (AIG Hospitals), Hyderabad, India
| | | |
Collapse
|
3
|
Setia Santoso LFA, Nasr K, Roumani AM, Abou Zeid HK, Dabbous MS. Unraveling tea and coffee consumption effects on cardiovascular diseases risk factors: A narrative review. Health Sci Rep 2024; 7:e70105. [PMID: 39323459 PMCID: PMC11422666 DOI: 10.1002/hsr2.70105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024] Open
Abstract
Backgrounds and Aims Daily lifestyle plays a vital role in modifying the risk for cardiovascular diseases (CVDs). Our daily life isn't inseparable from nutrition intake. As such, tea and coffee are often regarded as the most consumed beverages worldwide. There have been a lot of debates on the adverse effects and benefits of consuming these popular beverages. This comprehensive review explores the different types of tea and coffee and their mechanism of action. It delves deeper into their roles in reducing CVD risk, aiding CVD recovery, lowering CVD mortality, and their varying effects across populations and regions. Methods An extensive literature search was conducted on PubMed. Relevant articles were identified through cross-referencing and manual searches. Excluded from the study were commentaries, case reports, clinical vignettes, and non-English articles. Results Tea and coffee contain varying levels of caffeine and other bioactive compounds with cardioprotective effects against oxidative stress, inflammation, and more. Genetic factors further modulate their effects. Tea flavonoids benefit cholesterol, blood pressure, and endothelial function, while coffee constituents impact oxidative stress, metabolism, insulin sensitivity, and gut flora. Moderate consumption of both beverages may offer cardiovascular benefits, but outcomes vary depending on populations and conditions. Tea and coffee consumption may influence CVD recovery by reducing mortality and improving survival, however, it must be noted that it has the potential to be harmful to some individuals. Conclusion Evidence suggests that moderate consumption of these beverages may be linked to reduced cardiovascular mortality, although individual characteristics and pre-existing conditions can influence outcomes. Excessive caffeine consumption, found in both beverages, may pose risks such as arrhythmias, hypertension, and cardiovascular mortality in CVD patients, with a dose-dependent nature. Future research should delve into mechanisms, genetic factors, and diverse cultural impacts of its use. Health care providers should consider individual characteristics when advising on tea and coffee consumption in the context of cardiovascular health.
Collapse
Affiliation(s)
| | - Kristina Nasr
- Faculty of MedicineUniversity of BalamandAl‐KurahLebanon
| | | | | | | |
Collapse
|
4
|
Ali A, Jori C, Kanika, Kumar A, Vyawahare A, Kumar J, Kumar B, Ahmad A, Fareed M, Ali N, Navik U, Khan R. A bioactive and biodegradable vitamin C stearate-based injectable hydrogel alleviates experimental inflammatory arthritis. Biomater Sci 2024; 12:3389-3400. [PMID: 38804911 DOI: 10.1039/d4bm00243a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory joint disorder affecting nearly 1% of the global population. In RA, synovial joints are infiltrated by inflammatory mediators and enzymes, leading to articular cartilage deterioration, joint damage, and bone erosion. Herein, the 9-aminoacridine-6-O-stearoyl-L-ascorbic acid hydrogel (9AA-SAA hydrogel) was formulated by the heat-cool method and further characterized for surface charge, surface morphology, rheology, and cytocompatibility. Furthermore, we evaluated the therapeutic efficacy of the 9AA-SAA hydrogel, an enzyme-responsive drug delivery system with on-and-off switching capabilities based on disease severity against collagen-induced experimental arthritis in Wistar rats. The anti-inflammatory action of the US FDA-approved drug 9-aminoacridine (9AA) was revealed which acted through nuclear receptor subfamily 4 group A member 1 (NR4A1), an anti-inflammatory orphan nuclear receptor that inhibits nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB). Furthermore, we have explored the role of ascorbic acid, an active moiety of 6-O-stearoyl-L-ascorbic acid (SAA), in promoting the production of collagen production through ten-eleven translocation-2 (TET2) upregulation. Targeting through NR4A1 and TET2 could be the probable mechanism for the treatment of experimental arthritis. The combination of 9AA and ascorbic acid demonstrated enhanced therapeutic efficacy in the 9AA-SAA hydrogel, significantly reducing the severity of experimental arthritis. This approach, in contrast to existing treatments with limited effectiveness, presents a promising and more effective strategy for RA treatment by mitigating inflammation in experimental arthritis.
Collapse
Affiliation(s)
- Aneesh Ali
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Chandrashekhar Jori
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Akshay Vyawahare
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Jattin Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Bhuvnesh Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, Foothills Medical Centre, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Mohammad Fareed
- Environmental Health and Clinical Epidemiology Laboratory, Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, Mohali-140306, Punjab, India.
| |
Collapse
|
5
|
Schmidt KE, Höving AL, Kiani Zahrani S, Trevlopoulou K, Kaltschmidt B, Knabbe C, Kaltschmidt C. Serum-Induced Proliferation of Human Cardiac Stem Cells Is Modulated via TGFβRI/II and SMAD2/3. Int J Mol Sci 2024; 25:959. [PMID: 38256034 PMCID: PMC10815425 DOI: 10.3390/ijms25020959] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The ageing phenotype is strongly driven by the exhaustion of adult stem cells (ASCs) and the accumulation of senescent cells. Cardiovascular diseases (CVDs) and heart failure (HF) are strongly linked to the ageing phenotype and are the leading cause of death. As the human heart is considered as an organ with low regenerative capacity, treatments targeting the rejuvenation of human cardiac stem cells (hCSCs) are of great interest. In this study, the beneficial effects of human blood serum on proliferation and senescence of hCSCs have been investigated at the molecular level. We show the induction of a proliferation-related gene expression response by human blood serum at the mRNA level. The concurrent differential expression of the TGFβ target and inhibitor genes indicates the participation of TGFβ signalling in this context. Surprisingly, the application of TGFβ1 as well as the inhibition of TGFβ type I and type II receptor (TGFβRI/II) signalling strongly increased the proliferation of hCSCs. Likewise, both human blood serum and TGFβ1 reduced the senescence in hCSCs. The protective effect of serum on senescence in hCSCs was enhanced by simultaneous TGFβRI/II inhibition. These results strongly indicate a dual role of TGFβ signalling in terms of the serum-mediated effects on hCSCs. Further analysis via RNA sequencing (RNA-Seq) revealed the participation of Ras-inactivating genes wherefore a prevention of hyperproliferation upon serum-treatment in hCSCs via TGFβ signalling and Ras-induced senescence is suggested. These insights may improve treatments of heart failure in the future.
Collapse
Affiliation(s)
- Kazuko E. Schmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
- Institute for Laboratory and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
- Medical Faculty OWL, University of Bielefeld, 33615 Bielefeld, Germany
| | - Anna L. Höving
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
- Institute for Laboratory and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
- Medical Faculty OWL, University of Bielefeld, 33615 Bielefeld, Germany
| | - Sina Kiani Zahrani
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
| | - Katerina Trevlopoulou
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
| | - Barbara Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
- AG Molecular Neurobiology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Cornelius Knabbe
- Institute for Laboratory and Transfusion Medicine, Heart and Diabetes Centre NRW, Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
- Medical Faculty OWL, University of Bielefeld, 33615 Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, 33615 Bielefeld, Germany; (K.E.S.); (S.K.Z.); (K.T.); (B.K.); (C.K.)
| |
Collapse
|
6
|
Khan NG, Tungekar B, Adiga D, Chakrabarty S, Rai PS, Kabekkodu SP. Alterations induced by Bisphenol A on cellular organelles and potential relevance on human health. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119505. [PMID: 37286138 DOI: 10.1016/j.bbamcr.2023.119505] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/29/2023] [Accepted: 05/26/2023] [Indexed: 06/09/2023]
Abstract
Bisphenol A (BPA) is a chemical partially soluble in water and exists in a solid state. Its structural similarity with estrogen makes it an endocrine-disrupting chemical. BPA can disrupt signaling pathways at very low doses and may cause organellar stress. According to in vitro and in vivo studies, BPA interacts with various cell surface receptors to cause organellar stress, producing free radicals, cellular toxicity, structural changes, DNA damage, mitochondrial dysfunction, cytoskeleton remodeling, centriole duplication, and aberrant changes in several cell signaling pathways. The current review summarizes the impact of BPA exposure on the structural and functional aspects of subcellular components of cells such as the nucleus, mitochondria, endoplasmic reticulum, lysosome, ribosome, Golgi apparatus, and microtubules and its consequent impact on human health.
Collapse
Affiliation(s)
- Nadeem G Khan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Bushra Tungekar
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India; Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Padmalatha S Rai
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India; Center for DNA Repair and Genome Stability (CDRGS), Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
7
|
García-Yagüe ÁJ, Cuadrado A. Mechanisms of NURR1 Regulation: Consequences for Its Biological Activity and Involvement in Pathology. Int J Mol Sci 2023; 24:12280. [PMID: 37569656 PMCID: PMC10419244 DOI: 10.3390/ijms241512280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/29/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
NURR1 (Nuclear receptor-related 1 protein or NR4A2) is a nuclear protein receptor transcription factor with an essential role in the development, regulation, and maintenance of dopaminergic neurons and mediates the response to stressful stimuli during the perinatal period in mammalian brain development. The dysregulation of NURR1 activity may play a role in various diseases, including the onset and progression of neurodegenerative diseases, and several other pathologies. NURR1 is regulated by multiple mechanisms, among which phosphorylation by kinases or SUMOylation are the best characterized. Both post-translational modifications can regulate the activity of NURR1, affecting its stability and transcriptional activity. Other non-post-translational regulatory mechanisms include changes in its subcellular distribution or interaction with other protein partners by heterodimerization, also affecting its transcription activity. Here, we summarize the currently known regulatory mechanisms of NURR1 and provide a brief overview of its participation in pathological alterations.
Collapse
Affiliation(s)
- Ángel Juan García-Yagüe
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), 28027 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-CIBERNED), Av. Monforte de Lemos, 3-5. Pabellón 11, Planta, 28029 Madrid, Spain
| | - Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), 28027 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBER-CIBERNED), Av. Monforte de Lemos, 3-5. Pabellón 11, Planta, 28029 Madrid, Spain
| |
Collapse
|
8
|
Ortega R, Liu B, Persaud SJ. Effects of miR-33 Deficiency on Metabolic and Cardiovascular Diseases: Implications for Therapeutic Intervention. Int J Mol Sci 2023; 24:10777. [PMID: 37445956 DOI: 10.3390/ijms241310777] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that post-transcriptionally inhibit gene expression. These small molecules are involved in several biological conditions such as inflammation, cell growth and proliferation, and regulation of energy metabolism. In the context of metabolic and cardiovascular diseases, miR-33 is of particular interest as it has been implicated in the regulation of lipid and glucose metabolism. This miRNA is located in introns harboured in the genes encoding sterol regulatory element-binding protein (SREBP)-1 and SREBP-2, which are key transcription factors involved in lipid biosynthesis and cholesterol efflux. This review outlines the role of miR-33 in a range of metabolic and cardiovascular pathologies, such as dyslipidaemia, nonalcoholic fatty liver disease (NAFLD), obesity, diabetes, atherosclerosis, and abdominal aortic aneurysm (AAA), and it provides discussion about the effectiveness of miR-33 deficiency as a possible therapeutic strategy to prevent the development of these diseases.
Collapse
Affiliation(s)
- Rebeca Ortega
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Bo Liu
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Shanta J Persaud
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
9
|
Viñas JL, Spence M, Porter CJ, Douvris A, Gutsol A, Zimpelmann JA, Campbell PA, Burns KD. micro-RNA-486-5p protects against kidney ischemic injury and modifies the apoptotic transcriptome in proximal tubules. Kidney Int 2021; 100:597-612. [PMID: 34181969 DOI: 10.1016/j.kint.2021.05.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 02/08/2023]
Abstract
Acute kidney injury (AKI) carries high morbidity and mortality, and effective treatments are lacking. Preclinical models support involvement of micro-RNAs (miRs) in AKI pathogenesis, although effects on the kidney transcriptome are unclear. We previously showed that injection of cord blood endothelial colony forming cell-derived exosomes, enriched in miR-486-5p, prevented ischemic AKI in mice. To further define this, we studied direct effects of miR-486-5p in mice with kidney ischemia-reperfusion injury. RNA-Seq was used to compare the impact of miR-486-5p and exosomes on the transcriptome of proximal tubules and kidney endothelial cells 24 hours after ischemia-reperfusion. In mice with AKI, injection of miR-486-5p mimic increased its levels in proximal tubules and endothelial cells, and improved plasma creatinine, histological injury, neutrophil infiltration, and apoptosis. Additionally, miR-486-5p inhibited expression of its target phosphatase and tensin homolog, and activated protein kinase B. In proximal tubules, miR-486-5p or exosomes reduced expression of genes associated with ischemic injury and the tumor necrosis factor (TNF) pathway, and altered distinct apoptotic genes. In endothelial cells, genes associated with metabolic processes were altered by miR-486-5p or exosomes, although TNF pathway genes were not affected. Thus, our results suggest that miR-486-5p may have therapeutic potential in AKI.
Collapse
Affiliation(s)
- Jose L Viñas
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Matthew Spence
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Christopher J Porter
- Ottawa Bioinformatics Core Facility, the Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Adrianna Douvris
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Alex Gutsol
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Joseph A Zimpelmann
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Pearl A Campbell
- Regenerative Medicine Program, the Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Kevin D Burns
- Division of Nephrology, Department of Medicine, Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
10
|
Role of Nurr1 in Carcinogenesis and Tumor Immunology: A State of the Art Review. Cancers (Basel) 2020; 12:cancers12103044. [PMID: 33086676 PMCID: PMC7590204 DOI: 10.3390/cancers12103044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Nuclear receptor related-1 protein (Nurr1) emerges as a therapeutic target in multiple malignancies and immunotherapies. Previous studies have highlighted its association with clinicopathological parameters, tumorigenesis and therapeutic resistance in cancers. In addition, recent studies unraveled its contribution to the suppression of antitumor immunity, suggesting that inhibition of Nurr1 is a potential method to repress cancer aggressiveness and disrupt tumor immune tolerance. In line with this evidence, the present review provides the roles of Nurr1 in tumor progression and the associated underlying molecular mechanisms. Moreover, the significance of Nurr1 in promoting immune tolerance and potential strategies for Nurr1 inhibition are highlighted. Abstract Nuclear receptor related-1 protein (Nurr1), coded by an early response gene, is involved in multiple cellular and physiological functions, including proliferation, survival, and self-renewal. Dysregulation of Nurr1 has been frequently observed in many cancers and is attributed to multiple transcriptional and post-transcriptional mechanisms. Besides, Nurr1 exhibits extensive crosstalk with many oncogenic and tumor suppressor molecules, which contribute to its potential pro-malignant behaviors. Furthermore, Nurr1 is a key player in attenuating antitumor immune responses. It not only potentiates immunosuppressive functions of regulatory T cells but also dampens the activity of cytotoxic T cells. The selective accessibility of chromatin by Nurr1 in T cells is closely associated with cell exhaustion and poor efficacy of cancer immunotherapy. In this review, we summarize the reported findings of Nurr1 in different malignancies, the mechanisms that regulate Nurr1 expression, and the downstream signaling pathways that Nurr1 employs to promote a wide range of malignant phenotypes. We also give an overview of the association between Nurr1 and antitumor immunity and discuss the inhibition of Nurr1 as a potential immunotherapeutic strategy.
Collapse
|
11
|
You X, Guo ZF, Cheng F, Yi B, Yang F, Liu X, Zhu N, Zhao X, Yan G, Ma XL, Sun J. Transcriptional up-regulation of relaxin-3 by Nur77 attenuates β-adrenergic agonist-induced apoptosis in cardiomyocytes. J Biol Chem 2018; 293:14001-14011. [PMID: 30006349 DOI: 10.1074/jbc.ra118.003099] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/12/2018] [Indexed: 12/28/2022] Open
Abstract
The relaxin family peptides have been shown to exert several beneficial effects on the heart, including anti-apoptosis, anti-fibrosis, and anti-hypertrophy activity. Understanding their regulation might provide new opportunities for therapeutic interventions, but the molecular mechanism(s) coordinating relaxin expression in the heart remain largely obscured. Previous work demonstrated a role for the orphan nuclear receptor Nur77 in regulating cardiomyocyte apoptosis. We therefore investigated Nur77 in the hopes of identifying novel relaxin regulators. Quantitative real-time PCR (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA) data indicated that ectopic expression of orphan nuclear receptor Nur77 markedly increased the expression of latexin-3 (RLN3), but not relaxin-1 (RLN1), in neonatal rat ventricular cardiomyocytes (NRVMs). Furthermore, we found that the β-adrenergic agonist isoproterenol (ISO) markedly stimulated RLN3 expression, and this stimulation was significantly attenuated in Nur77 knockdown cardiomyocytes and Nur77 knockout hearts. We showed that Nur77 significantly increased RLN3 promoter activity via specific binding to the RLN3 promoter, as demonstrated by electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assays. Furthermore, we found that Nur77 overexpression potently inhibited ISO-induced cardiomyocyte apoptosis, whereas this protective effect was significantly attenuated in RLN3 knockdown cardiomyocytes, suggesting that Nur77-induced RLN3 expression is an important mediator for the suppression of cardiomyocyte apoptosis. These findings show that Nur77 regulates RLN3 expression, therefore suppressing apoptosis in the heart, and suggest that activation of Nur77 may represent a useful therapeutic strategy for inhibition of cardiac fibrosis and heart failure.
Collapse
Affiliation(s)
- Xiaohua You
- From the Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.,the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, and
| | - Zhi-Fu Guo
- From the Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.,the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, and
| | - Fang Cheng
- the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, and
| | - Bing Yi
- the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, and
| | - Fan Yang
- the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, and
| | - Xinzhu Liu
- the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, and
| | - Ni Zhu
- From the Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Xianxian Zhao
- From the Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Guijun Yan
- the Reproductive Medicine Center, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 211166, China
| | - Xin-Liang Ma
- the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, and
| | - Jianxin Sun
- From the Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China, .,the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, and
| |
Collapse
|
12
|
Won HY, Shin JH, Oh S, Jeong H, Hwang ES. Enhanced CD25 +Foxp3 + regulatory T cell development by amodiaquine through activation of nuclear receptor 4A. Sci Rep 2017; 7:16946. [PMID: 29208963 PMCID: PMC5717225 DOI: 10.1038/s41598-017-17073-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 11/21/2017] [Indexed: 01/10/2023] Open
Abstract
CD4+ T cells play key roles in the regulation of immune responses against pathogenic infectious antigens via development into effector T helper and induced regulatory T (iTreg) cells. Particularly, CD4+CD25+Foxp3+ iTreg cells are crucial for maintaining immune homeostasis and controlling inflammatory diseases. Anti-inflammatory drugs that enhance iTreg cell generation would be effective at preventing and treating inflammatory and autoimmune diseases. In this study, we examined whether anti-malarial and anti-arthritic amodiaquine (AQ) could affect iTreg cell development. Despite the anti-proliferative activity of AQ, AQ only moderately decreased iTreg cell proliferation but substantially increased IL-2 production by iTreg cells. Furthermore, AQ dose-dependently increased iTreg cell development and significantly upregulated iTreg cell markers including CD25. Interestingly, CD25 expression was decreased at later stages of iTreg cell development but was sustained in the presence of AQ, which was independent of IL-2 signaling pathway. AQ directly increased CD25 gene transcription by enhancing the DNA-binding and transcriptional activity of nuclear receptor 4 A. Most importantly, in vivo administration of AQ attenuated inflammatory colitis, resulted in the increased iTreg cells and decreased inflammatory cytokines. The ability of anti-malarial AQ to potentiate iTreg cell development makes it a promising drug for preventing and treating inflammatory and autoimmune diseases.
Collapse
MESH Headings
- Amodiaquine/pharmacology
- Animals
- Cell Proliferation/drug effects
- Colitis/drug therapy
- Colitis/etiology
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Forkhead Transcription Factors/metabolism
- Interleukin-2/metabolism
- Interleukin-2 Receptor alpha Subunit/genetics
- Interleukin-2 Receptor alpha Subunit/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/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, Interleukin-2/metabolism
- Receptors, Steroid/genetics
- Receptors, Steroid/metabolism
- Receptors, Thyroid Hormone/genetics
- Receptors, Thyroid Hormone/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/physiology
- Transforming Growth Factor beta/pharmacology
Collapse
Affiliation(s)
- Hee Yeon Won
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea
| | - Ji Hyun Shin
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea
| | - Sera Oh
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea
| | - Hana Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea
| | - Eun Sook Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Korea.
| |
Collapse
|
13
|
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
|
14
|
Nie X, Tan J, Dai Y, Mao W, Chen Y, Qin G, Li G, Shen C, Zhao J, Chen J. Nur77 downregulation triggers pulmonary artery smooth muscle cell proliferation and migration in mice with hypoxic pulmonary hypertension via the Axin2-β-catenin signaling pathway. Vascul Pharmacol 2016; 87:230-241. [PMID: 27871853 DOI: 10.1016/j.vph.2016.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 11/03/2016] [Indexed: 01/15/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by remodeling of the pulmonary vasculature, including marked proliferation and reduced apoptosis of pulmonary artery smooth muscle cells (PASMCs). Members of the nuclear receptor 4A (NR4A) subfamily are involved in a variety of biological events, such as cell apoptosis, proliferation, inflammation, and metabolism. Activation of Nur77 (an orphan nuclear receptor that belongs to NR4A subfamily) has recently been reported to be as a beneficial agent in the treatment of cardiovascular and metabolic diseases. In the present study, we investigated the effects of NR4A on human PASMCs function in vitro and determined the underlying mechanisms. We found a robust expression of NR4A receptors in lung tissues of PAH patients and hypoxic mice but a highly significant downregulation within pulmonary arteries (PAs) as assessed by quantitative polymerase chain reaction, immunoblotting, and immunohistochemistry. In vitro, NR4A receptors were found significantly decreased in PASMCs derived from PAH patients. To explore the pathological effects of decreased Nur77 in PASMCs, PASMCs were transduced with siRNA against Nur77. The siRNA-mediated knockdown of Nur77 significantly augmented PASMCs proliferation and migration. In contrast, Nur77 overexpression prevented PASMCs from proliferation and migration. Mechanistically, overexpression of Axis inhibition protein 2 (Axin2) or inhibition of β-catenin signaling was shown to be responsible for Nur77 knockdown-induced proliferation of PASMCs. Following hypoxia-induced angiogenesis of the pulmonary artery in C57BL/6 mice, small-molecule Nur77 agonists-Octaketide Cytosporone B (Csn-B) can significantly decreased thickness of vascular wall and markedly attenuated the development of chronic hypoxia-induced PAH in vivo. Therefore, reconstitution of Nur77 levels represents a promising therapeutic option to prevent vascular remodeling processes.
Collapse
Affiliation(s)
- Xiaowei Nie
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China.
| | - Jianxin Tan
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Youai Dai
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Wenjun Mao
- Department of Cardiothoracic Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Yuan Chen
- Department of Cardiothoracic Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Guowei Qin
- Department of Anesthesiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Guirong Li
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Chenyou Shen
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Jingjing Zhao
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China
| | - Jingyu Chen
- Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Department of Cardiothoracic Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China; Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, PR China.
| |
Collapse
|
15
|
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
|
16
|
Giner XC, Cotnoir-White D, Mader S, Lévesque D. Selective ligand activity at Nur/retinoid X receptor complexes revealed by dimer-specific bioluminescence resonance energy transfer-based sensors. FASEB J 2015; 29:4256-67. [PMID: 26148973 DOI: 10.1096/fj.14-259804] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 06/22/2015] [Indexed: 12/16/2022]
Abstract
Retinoid X receptors (RXRs) play a role as master regulators because of their capacity to form heterodimers with other nuclear receptors (NRs). Accordingly, retinoid signaling is involved in multiple biologic processes, including development, cell differentiation, metabolism, and cell death. However, the role and function of RXRs in different heterodimer complexes remain unidentified, mainly because most RXR drugs (called rexinoids) are not selective of specific heterodimer complexes. The lack of selectivity strongly limits the use of rexinoids for specific therapeutic approaches. To better characterize rexinoids at specific NR complexes, we have developed and optimized luciferase (Luc) protein complementation(PCA)-based bioluminescence resonance energy transfer (BRET) assays that can directly measure recruitment of a coactivator (CoA) motif fused to yellow fluorescent protein (YFP) by specific NR dimers. To validate the assays, we compared rexinoid modulation of CoA recruitment by the RXR homodimer and by the heterodimers Nur77/RXR and Nurr1/RXR. Results revealed that some rexinoids display selective CoA recruitment activities with homo- or heterodimer complexes. In particular, SR11237 (BMS649) has stronger potency for recruitment of CoA motif and transcriptional activity with the heterodimer Nur77/RXR than other complexes. This technology should be useful in identifying new compounds with specificity for individual dimeric species formed by NRs.
Collapse
Affiliation(s)
- Xavier C Giner
- *Faculté de Pharmacie and Groupe de Recherche Universitaire sur le Médicament, and Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, Québec, Canada
| | - David Cotnoir-White
- *Faculté de Pharmacie and Groupe de Recherche Universitaire sur le Médicament, and Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, Québec, Canada
| | - Sylvie Mader
- *Faculté de Pharmacie and Groupe de Recherche Universitaire sur le Médicament, and Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, Québec, Canada
| | - Daniel Lévesque
- *Faculté de Pharmacie and Groupe de Recherche Universitaire sur le Médicament, and Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
17
|
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
|
18
|
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]
|
19
|
Paillasse MR, de Medina P. The NR4A nuclear receptors as potential targets for anti-aging interventions. Med Hypotheses 2014; 84:135-40. [PMID: 25543265 DOI: 10.1016/j.mehy.2014.12.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 12/08/2014] [Indexed: 02/06/2023]
Abstract
The development of innovative anti-aging strategy is urgently needed to promote healthy aging and overcome the occurrence of age-related diseases such as cancer, diabetes, cardiovascular and neurodegenerative diseases. Genomic instability, deregulated nutrient sensing and mitochondrial dysfunction are established hallmark of aging. Interestingly, the orphan nuclear receptors NR4A subfamily (NR4A1, NR4A2 and NR4A3) are nutrient sensors that trigger mitochondria biogenesis and improve intrinsic mitochondrial function. In addition, NR4A receptors are components of DNA repair machinery and promote DNA repair. Members of the NR4A subfamily should also be involved in anti-aging properties of hormesis since these receptors are induced by various form of cellular stress and stimulate protective cells response such as anti-oxidative activity and DNA repair. Previous studies reported that NR4A nuclear receptors subfamily is potential therapeutic targets for the treatment of age related disorders (e.g. metabolic syndromes, diabetes and neurodegenerative diseases). Consequently, we propose that targeting NR4A receptors might constitute a new approach to delay aging and the onset of diseases affecting our aging population.
Collapse
|
20
|
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]
|
21
|
Radom-Aizik S, Zaldivar FP, Haddad F, Cooper DM. Impact of brief exercise on circulating monocyte gene and microRNA expression: implications for atherosclerotic vascular disease. Brain Behav Immun 2014; 39:121-9. [PMID: 24423463 PMCID: PMC4101903 DOI: 10.1016/j.bbi.2014.01.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 12/24/2013] [Accepted: 01/04/2014] [Indexed: 12/14/2022] Open
Abstract
Physical activity can prevent and/or attenuate atherosclerosis, a disease clearly linked to inflammation. Paradoxically, even brief exercise induces a stress response and increases inflammatory cells like monocytes in the circulation. We hypothesized that exercise would regulate the expression of genes, gene pathways, and microRNAs in monocytes in a way that could limit pro-inflammatory function and drive monocytes to prevent, rather than contribute to, atherosclerosis. Twelve healthy men (22-30year old) performed ten 2-min bouts of cycle ergometer exercise at a constant work equivalent to an average of 82% of maximum O2 consumption interspersed with 1-min rest. Blood was drawn before and immediately after the exercise. Monocytes were isolated from peripheral blood mononuclear cells. Flow cytometry was used to identify monocyte subtypes. We used Affymetrix U133 + 2.0 arrays for gene expression and Agilent Human miRNA V2 Microarray for miRNAs. A stringent statistical approach (FDR <0.05) was used to determine that exercise significantly altered the expression of 894 annotated genes and 19 miRNAs. We found distinct gene alterations that were likely to direct monocytes in an anti-inflammatory, anti-atherogenic pathway, including the downregulation of monocyte TNF, TLR4, and CD36 genes and the upregulation of EREG and CXCR4. Exercise significantly altered a number of microRNAs that likely influence monocytes involvement in vascular health. Exercise leads to a novel genomic profile of circulating monocytes, which appears to promote cardiovascular health despite the overall stress response.
Collapse
Affiliation(s)
- Shlomit Radom-Aizik
- Pediatric Exercise and Genomics Research Center, Department of Pediatrics, UC Irvine School of Medicine, United States.
| | - Frank P. Zaldivar
- Pediatric Exercise and Genomics Research Center, Department of Pediatrics, UC Irvine School of Medicine
| | - Fadia Haddad
- Pediatric Exercise and Genomics Research Center, Department of Pediatrics, UC Irvine School of Medicine
| | - Dan M. Cooper
- Pediatric Exercise and Genomics Research Center, Department of Pediatrics, UC Irvine School of Medicine
| |
Collapse
|
22
|
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]
|
23
|
Abstract
PURPOSE OF REVIEW To understand chronic inflammatory diseases such as atherosclerosis, we require in-depth knowledge on immune-cell differentiation, function of specific immune-cell subsets and endothelial cell-mediated extravasation. In this review, we summarize a number of very recent observations on the pivotal function of NR4A nuclear receptors in immunity and atherosclerosis. RECENT FINDINGS NR4A nuclear receptors are involved in negative selection of thymocytes, Treg differentiation and the development of Ly6C monocytes. Nur77 and Nurr1 attenuate atherosclerosis in mice whereas NOR-1 aggravates vascular lesion formation. SUMMARY These exciting, novel insights on the function of NR4A nuclear receptors in immunity, vascular cells and atherosclerosis will initiate a plethora of studies to understand the underlying molecular mechanisms, which will culminate in the identification of novel NR4A targets to modulate chronic inflammatory disease.
Collapse
Affiliation(s)
- Anouk A.J. Hamers
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam
| | - Richard N. Hanna
- Division of inflammatory Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Heba Nowyhed
- Division of inflammatory Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Catherine C. Hedrick
- Division of inflammatory Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | - Carlie J.M. de Vries
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam
| |
Collapse
|
24
|
Close AF, Rouillard C, Buteau J. NR4A orphan nuclear receptors in glucose homeostasis: a minireview. DIABETES & METABOLISM 2013; 39:478-84. [PMID: 24075454 DOI: 10.1016/j.diabet.2013.07.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 07/15/2013] [Accepted: 07/17/2013] [Indexed: 01/23/2023]
Abstract
Type 2 diabetes mellitus is a disorder characterized by insulin resistance and a relative deficit in insulin secretion, both of which result in elevated blood glucose. Understanding the molecular mechanisms underlying the pathophysiology of diabetes could lead to the development of new therapeutic approaches. An ever-growing body of evidence suggests that members of the NR4A family of nuclear receptors could play a pivotal role in glucose homeostasis. This review aims to present and discuss advances so far in the evaluation of the potential role of NR4A in the regulation of glucose homeostasis and the development of type 2 diabetes.
Collapse
Affiliation(s)
- A F Close
- Department of AFNS, University of Alberta and Alberta Diabetes Institute, Li Ka Shing Centre, Edmonton, AB, T6G 2E1, Canada
| | | | | |
Collapse
|
25
|
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
|
26
|
Y H, HB L, RW S, AE D. GLP-1-dependent and independent effects and molecular mechanisms of a dipeptidyl peptidase 4 inhibitor in vascular endothelial cells. Mol Biol Rep 2012. [DOI: 10.1007/s11033-012-2290-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Eftedal I, Jørgensen A, Røsbjørgen R, Flatberg A, Brubakk AO. Early genetic responses in rat vascular tissue after simulated diving. Physiol Genomics 2012; 44:1201-7. [PMID: 23132759 DOI: 10.1152/physiolgenomics.00073.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Diving causes a transient reduction of vascular function, but the mechanisms behind this are largely unknown. The aim of this study was therefore to analyze genetic reactions that may be involved in acute changes of vascular function in divers. Rats were exposed to 709 kPa of hyperbaric air (149 kPa Po(2)) for 50 min followed by postdive monitoring of vascular bubble formation and full genome microarray analysis of the aorta from diving rats (n = 8) and unexposed controls (n = 9). Upregulation of 23 genes was observed 1 h after simulated diving. The differential gene expression was characteristic of cellular responses to oxidative stress, with functions of upregulated genes including activation and fine-tuning of stress-responsive transcription, cytokine/cytokine receptor signaling, molecular chaperoning, and coagulation. By qRT-PCR, we verified increased transcription of neuron-derived orphan receptor-1 (Nr4a3), plasminogen activator inhibitor 1 (Serpine1), cytokine TWEAK receptor FN14 (Tnfrsf12a), transcription factor class E basic helix-loop-helix protein 40 (Bhlhe40), and adrenomedullin (Adm). Hypoxia-inducible transcription factor HIF1 subunit HIF1-α was stabilized in the aorta 1 h after diving, and after 4 h there was a fivefold increase in total protein levels of the procoagulant plasminogen activator inhibitor 1 (PAI1) in blood plasma from diving rats. The study did not have sufficient power for individual assessment of effects of hyperoxia and decompression-induced bubbles on postdive gene expression. However, differential gene expression in rats without venous bubbles was similar to that of all the diving rats, indicating that elevated Po(2) instigated the observed genetic reactions.
Collapse
Affiliation(s)
- Ingrid Eftedal
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway.
| | | | | | | | | |
Collapse
|
28
|
Abstract
Inflammation is paradoxical; it is essential for protection following biological, chemical or physical stimuli, but inappropriate or misdirected inflammation is responsible for tissue injury in a variety of inflammatory diseases. The polarization of immune cells is critical in controlling the stages of inflammatory response. The acute phase of inflammation is characterized by a T-lymphocyte:Th2 cytokine profile and involves a co-ordinated migration of immune cells to the site of injury where production of cytokines and acute-phase proteins brings about healing. However, persistent inflammation can result in inappropriate and prolonged T-lymphocyte:Th1 cytokine-mediated action and reaction of self-molecules, leading to a chronic phase in diseases such as RA (rheumatoid arthritis), Ps (psoriasis) and atherosclerosis. The inflammatory response is also controlled by activated macrophage cells, with classically activated (M1) cells producing a wide variety of pro-inflammatory mediators, while alternatively activated (M2) macrophages participate in anti-inflammatory response. Members of the NR4A subfamily (NR4A1/NUR77, NR4A2/NURR1 and NR4A3/NOR1) of orphan NRs (nuclear receptors) have emerged as key transcriptional regulators of cytokine and growth factor action in diseases affecting our aging population. As ligand-independent and constitutively active receptors, the activity of these transcription factors is tightly controlled at the level of expression, post-translational modification and subcellular localization. NR4A subfamily members are aberrantly expressed in inflamed human synovial tissue, psoriatic skin, atherosclerotic lesions, lung and colorectal cancer cells. Significantly, prolonged or inappropriate inflammatory responses contribute to the pathogenesis of these diseases. In activated cells, NR4A receptors are rapidly and potently induced, suggesting that these receptors may act as important transcriptional mediators of inflammatory signals. NR4A receptors may contribute to the cellular processes that control inflammation, playing a critical part in the contribution of chronic inflammation or they may have a protective role, where they may mediate pro-resolution responses. Here, we will review the contribution of the NR4A orphan NRs to integration of cytokine signalling in inflammatory disorders.
Collapse
|
29
|
Berrabah W, Aumercier P, Lefebvre P, Staels B. Control of nuclear receptor activities in metabolism by post-translational modifications. FEBS Lett 2011; 585:1640-50. [PMID: 21486568 DOI: 10.1016/j.febslet.2011.03.066] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 03/30/2011] [Indexed: 12/24/2022]
Abstract
Nuclear receptors (NRs) are molecular transducers of endocrine and dietary signals allowing tissues to adapt their transcriptional responses to endogenous or exogenous cues. These signals act in many cases as specific ligands, converting of NRs into transcriptionally active molecules. This on-off mechanism needs, however, to be finely tuned with respect to the tissue environment and adjusted to the organism needs. These subtle adjustments of NR transcriptional activity are brought about by post-translational modifications (PTMs), which can be, in the case of orphan NRs, the sole regulatory mechanism. The role of PTMs, with a more specific focus on phosphorylation, affecting the functions of NR controlling metabolic events is described in this review.
Collapse
Affiliation(s)
- Wahiba Berrabah
- Université Lille Nord de France, INSERM, U1011, Lille, France
| | | | | | | |
Collapse
|