1
|
Abdelaziz AM. Alpha-Synuclein drives NURR1 and NLRP3 Inflammasome dysregulation in Parkinson's disease: From pathogenesis to potential therapeutic strategies. Int Immunopharmacol 2025; 156:114692. [PMID: 40267723 DOI: 10.1016/j.intimp.2025.114692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
Parkinson's disease (PD), a progressive neurodegenerative disorder, is characterized by the loss of dopaminergic neurons and pathological aggregation of α-synuclein (α-Syn). Emerging evidence highlights the interplay between genetic susceptibility, neuroinflammation, and transcriptional dysregulation in driving PD pathogenesis. This review brings together the latest information on three important players: α-Syn, the transcription factor Orphan nuclear receptor (NURR1), and the NOD-like receptor 3 (NLRP3) inflammasome. Pathogenic α-syn aggregates cause damage to neurons by disrupting mitochondria and lysosomes and spreading in a way similar to prion proteins. They also turn on the NLRP3 inflammasome, which is a key player in neuroinflammation. NLRP3-driven release of pro-inflammatory cytokines exacerbates neurodegeneration and creates a self-sustaining inflammatory milieu. Meanwhile, reduced NURR1 activity, a pivotal modulator of dopaminergic neuron survival and development, exposes neurons to oxidative stress, neuroinflammation, and α-Syn toxicity, hence exacerbating disease progression. So, targeting this trio exhibits transformative potential against PD pathogenesis.
Collapse
Affiliation(s)
- Ahmed M Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish 45511, Egypt.
| |
Collapse
|
2
|
da Silva AR, Gunawan F, Boezio GLM, Faure E, Théron A, Avierinos JF, Lim S, Jha SG, Ramadass R, Guenther S, Looso M, Zaffran S, Juan T, Stainier DYR. egr3 is a mechanosensitive transcription factor gene required for cardiac valve morphogenesis. SCIENCE ADVANCES 2024; 10:eadl0633. [PMID: 38748804 PMCID: PMC11095463 DOI: 10.1126/sciadv.adl0633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/11/2024] [Indexed: 05/19/2024]
Abstract
Biomechanical forces, and their molecular transducers, including key mechanosensitive transcription factor genes, such as KLF2, are required for cardiac valve morphogenesis. However, klf2 mutants fail to completely recapitulate the valveless phenotype observed under no-flow conditions. Here, we identify the transcription factor EGR3 as a conserved biomechanical force transducer critical for cardiac valve formation. We first show that egr3 null zebrafish display a complete and highly penetrant loss of valve leaflets, leading to severe blood regurgitation. Using tissue-specific loss- and gain-of-function tools, we find that during cardiac valve formation, Egr3 functions cell-autonomously in endothelial cells, and identify one of its effectors, the nuclear receptor Nr4a2b. We further find that mechanical forces up-regulate egr3/EGR3 expression in the developing zebrafish heart and in porcine valvular endothelial cells, as well as during human aortic valve remodeling. Altogether, these findings reveal that EGR3 is necessary to transduce the biomechanical cues required for zebrafish cardiac valve morphogenesis, and potentially for pathological aortic valve remodeling in humans.
Collapse
Affiliation(s)
- Agatha Ribeiro da Silva
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Felix Gunawan
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Giulia L. M. Boezio
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Emilie Faure
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
| | - Alexis Théron
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
- Service de Chirurgie Cardiaque, AP-HM, Hôpital de la Timone, 13005 Marseille, France
| | - Jean-François Avierinos
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
- Service de Cardiologie, AP-HM, Hôpital de la Timone, 13005 Marseille, France
| | - SoEun Lim
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
| | - Shivam Govind Jha
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
| | - Radhan Ramadass
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
| | - Stefan Guenther
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mario Looso
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stéphane Zaffran
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
| | - Thomas Juan
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Didier Y. R. Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| |
Collapse
|
3
|
Abbey CA, Duran CL, Chen Z, Chen Y, Roy S, Coffell A, Sveeggen TM, Chakraborty S, Wells GB, Chang J, Bayless KJ. Identification of New Markers of Angiogenic Sprouting Using Transcriptomics: New Role for RND3. Arterioscler Thromb Vasc Biol 2024; 44:e145-e167. [PMID: 38482696 PMCID: PMC11043006 DOI: 10.1161/atvbaha.123.320599] [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: 12/18/2023] [Accepted: 02/28/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND New blood vessel formation requires endothelial cells to transition from a quiescent to an invasive phenotype. Transcriptional changes are vital for this switch, but a comprehensive genome-wide approach focused exclusively on endothelial cell sprout initiation has not been reported. METHODS Using a model of human endothelial cell sprout initiation, we developed a protocol to physically separate cells that initiate the process of new blood vessel formation (invading cells) from noninvading cells. We used this model to perform multiple transcriptomics analyses from independent donors to monitor endothelial gene expression changes. RESULTS Single-cell population analyses, single-cell cluster analyses, and bulk RNA sequencing revealed common transcriptomic changes associated with invading cells. We also found that collagenase digestion used to isolate single cells upregulated the Fos proto-oncogene transcription factor. Exclusion of Fos proto-oncogene expressing cells revealed a gene signature consistent with activation of signal transduction, morphogenesis, and immune responses. Many of the genes were previously shown to regulate angiogenesis and included multiple tip cell markers. Upregulation of SNAI1 (snail family transcriptional repressor 1), PTGS2 (prostaglandin synthase 2), and JUNB (JunB proto-oncogene) protein expression was confirmed in invading cells, and silencing JunB and SNAI1 significantly reduced invasion responses. Separate studies investigated rounding 3, also known as RhoE, which has not yet been implicated in angiogenesis. Silencing rounding 3 reduced endothelial invasion distance as well as filopodia length, fitting with a pathfinding role for rounding 3 via regulation of filopodial extensions. Analysis of in vivo retinal angiogenesis in Rnd3 heterozygous mice confirmed a decrease in filopodial length compared with wild-type littermates. CONCLUSIONS Validation of multiple genes, including rounding 3, revealed a functional role for this gene signature early in the angiogenic process. This study expands the list of genes associated with the acquisition of a tip cell phenotype during endothelial cell sprout initiation.
Collapse
Affiliation(s)
- Colette A. Abbey
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Camille L. Duran
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Zhishi Chen
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Houston, TX
| | - Yanping Chen
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Houston, TX
| | - Sukanya Roy
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
| | - Ashley Coffell
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Timothy M. Sveeggen
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Sanjukta Chakraborty
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
| | - Gregg B. Wells
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, TX
| | - Jiang Chang
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Houston, TX
| | - Kayla J. Bayless
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| |
Collapse
|
4
|
Choi M, Lee SM, Lee JW, Kim I, Pack CG, Ha CH. Yeast beta-glucan mediates histone deacetylase 5-induced angiogenesis in vascular endothelial cells. Int J Biol Macromol 2022; 211:556-567. [PMID: 35569678 DOI: 10.1016/j.ijbiomac.2022.05.057] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/28/2022] [Accepted: 05/08/2022] [Indexed: 12/13/2022]
Abstract
The role of yeast-derived β-glucan in angiogenesis has not been elucidated because there have been few specific studies on its clinical and physiological significance. Therefore, this study investigated the correlation between β-glucan and histone deacetylase 5 (HDAC5) in human umbilical vein endothelial cells (HUVECs), revealing the role of β-glucan in angiogenesis. We confirmed that HDAC5 was phosphorylated by β-glucan stimulation and released from the nucleus to the cytoplasm. Furthermore, we found that β-glucan-stimulated HDAC5 translocation mediates the transcriptional activation of MEF2. As a result, the expression of KLF2, EGR2, and NR4A2, whose expression is MEF2-dependent and involved in angiogenesis, increased. Thus, we showed the activity of β-glucan in angiogenesis through in vitro and ex vivo assays including cell migration, tube formation, and aortic ring analyses. Specifically, application of an HDAC5 inhibitor repressed MEF2 transcriptional activation in both in vitro and ex vivo angiogenesis. HDAC5 inhibitor LMK235 inhibited the proangiogenic activity of beta-glucan, suggesting that β-glucan induces angiogenesis through HDAC5. These findings suggest that HDAC5 is essential for angiogenesis, and that β-glucan induces angiogenesis. In conclusion, this study demonstrates that β-glucan induces angiogenesis through HDAC5. It also suggests that β-glucan has potential value as a novel therapeutic agent for modulating angiogenesis.
Collapse
Affiliation(s)
- Min Choi
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung Min Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin Woo Lee
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Inki Kim
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chan-Gi Pack
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chang Hoon Ha
- Department of Convergence Medicine and Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Wu SC, Kuo PJ, Rau CS, Huang LH, Lin CW, Wu YC, Wu CJ, Tsai CW, Hsieh TM, Liu HT, Huang CY, Hsieh CH. Increased Angiogenesis by Exosomes Secreted by Adipose-Derived Stem Cells upon Lipopolysaccharide Stimulation. Int J Mol Sci 2021; 22:8877. [PMID: 34445582 PMCID: PMC8396299 DOI: 10.3390/ijms22168877] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/19/2022] Open
Abstract
Exosomes secreted by adipose-derived stem cells (ADSCs) enhance angiogenesis and wound healing. However, in clinical settings, wounds may be infected by various bacteria or pathogens. We investigated whether human ADSCs stimulated with lipopolysaccharide (LPS) secrete exosomes (ADSC-LPS-exo) that augment the angiogenesis of human umbilical vein endothelial cells (HUVECs). ExoQuick-TC exosome precipitation solution was used to purify exosomes from human ADSC culture media in the presence or absence of 1 µg/mL LPS treatment for 24 h. The uptake of ADSC-LPS-exo significantly induced the activation of cAMP response element binding protein (CREB), activating protein 1 (AP-1), and nuclear factor-κB (NF-κB) signaling pathways and increased the migration of and tube formation in HUVECs. RNA interference with CREB, AP-1, or NF-κB1 significantly reduced the migration of and tube formation in HUVECs treated with ADSC-LPS-exo. An experiment with an antibody array for 25 angiogenesis-related proteins revealed that only interleukin-8 expression was significantly upregulated in HUVECs treated with ADSC-LPS-exo. In addition, proteomic analysis revealed that eukaryotic translation initiation factor 4E, amyloid beta A4 protein, integrin beta-1, and ras-related C3 botulinum toxin substrate 1 may be potential candidates involved in ADSC-LPS-exo-mediated enhanced angiogenesis.
Collapse
Affiliation(s)
- Shao-Chun Wu
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan;
| | - Pao-Jen Kuo
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Cheng-Shyuan Rau
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (C.-S.R.); (L.-H.H.)
| | - Lien-Hung Huang
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (C.-S.R.); (L.-H.H.)
| | - Chia-Wei Lin
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Yi-Chan Wu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Chia-Jung Wu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Chia-Wen Tsai
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Ting-Min Hsieh
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (T.-M.H.); (H.-T.L.)
| | - Hang-Tsung Liu
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (T.-M.H.); (H.-T.L.)
| | - Chun-Ying Huang
- Department of Trauma Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (T.-M.H.); (H.-T.L.)
| | - Ching-Hua Hsieh
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kahosiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| |
Collapse
|
6
|
Zhang X, Connelly J, Chao Y, Wang QJ. Multifaceted Functions of Protein Kinase D in Pathological Processes and Human Diseases. Biomolecules 2021; 11:biom11030483. [PMID: 33807058 PMCID: PMC8005150 DOI: 10.3390/biom11030483] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023] Open
Abstract
Protein kinase D (PKD) is a family of serine/threonine protein kinases operating in the signaling network of the second messenger diacylglycerol. The three family members, PKD1, PKD2, and PKD3, are activated by a variety of extracellular stimuli and transduce cell signals affecting many aspects of basic cell functions including secretion, migration, proliferation, survival, angiogenesis, and immune response. Dysregulation of PKD in expression and activity has been detected in many human diseases. Further loss- or gain-of-function studies at cellular levels and in animal models provide strong support for crucial roles of PKD in many pathological conditions, including cancer, metabolic disorders, cardiac diseases, central nervous system disorders, inflammatory diseases, and immune dysregulation. Complexity in enzymatic regulation and function is evident as PKD isoforms may act differently in different biological systems and disease models, and understanding the molecular mechanisms underlying these differences and their biological significance in vivo is essential for the development of safer and more effective PKD-targeted therapies. In this review, to provide a global understanding of PKD function, we present an overview of the PKD family in several major human diseases with more focus on cancer-associated biological processes.
Collapse
|
7
|
Hepatic stellate cells promote intrahepatic cholangiocarcinoma progression via NR4A2/osteopontin/Wnt signaling axis. Oncogene 2021; 40:2910-2922. [PMID: 33742120 DOI: 10.1038/s41388-021-01705-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 01/26/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022]
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a highly fatal malignancy characterized by a vast amount of intra-tumoral fibroblasts. These fibroblasts are potentially implicated in maintaining the high aggressiveness of ICC, whereas its pro-cancer mechanisms remain scarcely reported. Here, by establishing co-culture models of ICC cells and hepatic stellate cells (HSCs), we identified that HSCs triggered the expression of nuclear receptor family 4 subgroup A member 2 (NR4A2), a transcription factor previously reported as a molecular switch between inflammation and cancer, in ICC cells. Functionally, NR4A2 promotes tumor proliferation, metastatic potentiality and represents an independent prognostic indicator for overall survival in ICC patients. Mechanistically, NR4A2 upregulates osteopontin (OPN) expression through transcriptional activation and thereby augments the activity of Wnt/β-catenin signaling. Intriguingly, in the context of co-culture, vascular endothelial growth factor (VEGF), a previously proved NR4A2 stimulus, not only enhances NR4A2 expression, but also can be blunted by the interference of the NR4A2-OPN axis. Altogether, this study suggests the NR4A2/OPN/Wnt signaling axis to be a pivotal executor of HSC-instigated cancer-promoting roles in ICC, and the NR4A2/OPN/VEGF positive feedback loop may help to reinforce the effect.
Collapse
|
8
|
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
|
9
|
Goyal D, Goyal R. Angiogenic Transformation in Human Brain Micro Endothelial Cells: Whole Genome DNA Methylation and Transcriptomic Analysis. Front Physiol 2019; 10:1502. [PMID: 31920707 PMCID: PMC6917667 DOI: 10.3389/fphys.2019.01502] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/26/2019] [Indexed: 01/22/2023] Open
Abstract
We tested the hypothesis that endothelial capillary tube formation in 3D cultures in basement membrane extract (BME) is secondary to the altered DNA promoter methylation and mRNA expression in human brain micro endothelial cells (HBMECs). We conducted a whole-genome transcriptomic and methylation microarray and CRISPR/Cas9-mediated gene knockdown to test our hypothesis. The data demonstrated that with angiogenic transformation 1318 and 1490 genes were significantly (p < 0.05) upregulated and downregulated, respectively. We compared our gene expression data with the published databases on GEO and found several genes in common. PTGS2, SELE, ID2, HSPA6, DLX2, HEY2, FOSB, SMAD6, SMAD7, and SMAD9 showed a very high level of expression during capillary tube formation. Among downregulated gene were ITGB4, TNNT1, PRSS35, TXNIP, IGFBP5. The most affected canonical pathways were ATM signaling and cell cycle G2/M DNA damage checkpoint regulation. The top upstream regulators of angiogenic transformation were identified to be VEGF, TP53, HGF, ESR1, and CDKN1A. We compared the changes in gene expression with the change in gene methylation and found hypomethylation of the CpG sites was associated with upregulation of 515 genes and hypermethylation was associated with the downregulation of 31 genes. Furthermore, the silencing of FOSB, FZD7, HEY2, HSPA6, NR4A3, SELE, PTGS2, SMAD6, SMAD7, and SMAD9 significantly inhibited angiogenic transformation as well as cell migration of HBMECs. We conclude that the angiogenic transformation is associated with altered DNA methylation and gene expression changes.
Collapse
Affiliation(s)
- Dipali Goyal
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
| | - Ravi Goyal
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
10
|
Schneller D, Hofer-Warbinek R, Sturtzel C, Lipnik K, Gencelli B, Seltenhammer M, Wen M, Testori J, Bilban M, Borowski A, Windwarder M, Kapel SS, Besemfelder E, Cejka P, Habertheuer A, Schlechta B, Majdic O, Altmann F, Kocher A, Augustin HG, Luttmann W, Hofer E. Cytokine-Like 1 Is a Novel Proangiogenic Factor Secreted by and Mediating Functions of Endothelial Progenitor Cells. Circ Res 2019; 124:243-255. [PMID: 30582450 DOI: 10.1161/circresaha.118.313645] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RATIONALE Endothelial colony forming cells (ECFCs) or late blood outgrowth endothelial cells can be isolated from human cord or peripheral blood, display properties of endothelial progenitors, home into ischemic tissues and support neovascularization in ischemic disease models. OBJECTIVE To assess the functions of CYTL1 (cytokine-like 1), a factor we found preferentially produced by ECFCs, in regard of vessel formation. METHODS AND RESULTS We show by transcriptomic analysis that ECFCs are distinguished from endothelial cells of the vessel wall by production of high amounts of CYTL1. Modulation of expression demonstrates that the factor confers increased angiogenic sprouting capabilities to ECFCs and can also trigger sprouting of mature endothelial cells. The data further display that CYTL1 can be induced by hypoxia and that it functions largely independent of VEGF-A (vascular endothelial growth factor-A). By recombinant production of CYTL1 we confirm that the peptide is indeed a strong proangiogenic factor and induces sprouting in cellular assays and functional vessel formation in animal models comparable to VEGF-A. Mass spectroscopy corroborates that CYTL1 is specifically O-glycosylated on 2 neighboring threonines in the C-terminal part and this modification is important for its proangiogenic bioactivity. Further analyses show that the factor does not upregulate proinflammatory genes and strongly induces several metallothionein genes encoding anti-inflammatory and antiapoptotic proteins. CONCLUSIONS We conclude that CYTL1 can mediate proangiogenic functions ascribed to endothelial progenitors such as ECFCs in vivo and may be a candidate to support vessel formation and tissue regeneration in ischemic pathologies.
Collapse
Affiliation(s)
- Doris Schneller
- From the Department of Vascular Biology and Thrombosis Research (D.S., C.S., K.L., B.G., M.S., M. Wen, J.T., E.H.), Medical University of Vienna, Austria.,Division Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), Heidelberg (D.S.)
| | - Renate Hofer-Warbinek
- Clinical Department for Heart Surgery (R.H.-W., A.H., A.K.), Medical University of Vienna, Austria
| | - Caterina Sturtzel
- From the Department of Vascular Biology and Thrombosis Research (D.S., C.S., K.L., B.G., M.S., M. Wen, J.T., E.H.), Medical University of Vienna, Austria
| | - Karoline Lipnik
- From the Department of Vascular Biology and Thrombosis Research (D.S., C.S., K.L., B.G., M.S., M. Wen, J.T., E.H.), Medical University of Vienna, Austria
| | - Burcu Gencelli
- From the Department of Vascular Biology and Thrombosis Research (D.S., C.S., K.L., B.G., M.S., M. Wen, J.T., E.H.), Medical University of Vienna, Austria
| | - Monika Seltenhammer
- From the Department of Vascular Biology and Thrombosis Research (D.S., C.S., K.L., B.G., M.S., M. Wen, J.T., E.H.), Medical University of Vienna, Austria
| | - Mingjie Wen
- From the Department of Vascular Biology and Thrombosis Research (D.S., C.S., K.L., B.G., M.S., M. Wen, J.T., E.H.), Medical University of Vienna, Austria
| | - Julia Testori
- From the Department of Vascular Biology and Thrombosis Research (D.S., C.S., K.L., B.G., M.S., M. Wen, J.T., E.H.), Medical University of Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine (M.B.), Medical University of Vienna, Austria
| | | | - Markus Windwarder
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria (M. Windwarder, F.A.)
| | - Stephanie S Kapel
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (S.S.K., E.B., H.G.A.).,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Germany (S.S.K., H.G.A.)
| | - Eva Besemfelder
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (S.S.K., E.B., H.G.A.)
| | - Petra Cejka
- Department of Immunology (P.C., O.M.), Medical University of Vienna, Austria
| | - Andreas Habertheuer
- Clinical Department for Heart Surgery (R.H.-W., A.H., A.K.), Medical University of Vienna, Austria
| | - Bernhard Schlechta
- Department of Gynecology and Obstetrics (B.S.), Medical University of Vienna, Austria
| | - Otto Majdic
- Department of Immunology (P.C., O.M.), Medical University of Vienna, Austria
| | - Friedrich Altmann
- Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria (M. Windwarder, F.A.)
| | - Alfred Kocher
- Clinical Department for Heart Surgery (R.H.-W., A.H., A.K.), Medical University of Vienna, Austria
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (S.S.K., E.B., H.G.A.).,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Germany (S.S.K., H.G.A.)
| | | | - Erhard Hofer
- From the Department of Vascular Biology and Thrombosis Research (D.S., C.S., K.L., B.G., M.S., M. Wen, J.T., E.H.), Medical University of Vienna, Austria
| |
Collapse
|
11
|
Function of Nr4a Orphan Nuclear Receptors in Proliferation, Apoptosis and Fuel Utilization Across Tissues. Cells 2019; 8:cells8111373. [PMID: 31683815 PMCID: PMC6912296 DOI: 10.3390/cells8111373] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/24/2019] [Accepted: 10/30/2019] [Indexed: 12/21/2022] Open
Abstract
The Nr4a family of nuclear hormone receptors is composed of three members-Nr4a1/Nur77, Nr4a2/Nurr1 and Nr4a3/Nor1. While currently defined as ligandless, these transcription factors have been shown to regulate varied processes across a host of tissues. Of particular interest, the Nr4a family impinge, in a tissue dependent fashion, on cellular proliferation, apoptosis and fuel utilization. The regulation of these processes occurs through both nuclear and non-genomic pathways. The purpose of this review is to provide a balanced perspective of the tissue specific and Nr4a family member specific, effects on cellular proliferation, apoptosis and fuel utilization.
Collapse
|
12
|
Liu DH, Zhao ZR, Lin YB, Zhou WJ, Hou JY, Ye ZH, Long H. Prognostic Effect of TP53 and PKD Co-Mutations in Patients with Resected Epidermal Growth Factor Receptor-Mutated Lung Adenocarcinoma. Ann Surg Oncol 2019; 26:1934-1941. [PMID: 30820786 DOI: 10.1245/s10434-019-07254-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND The impact of specific co-mutations in epidermal growth factor receptor (EGFR)-mutated lung adenocarcinoma is unclear. METHODS Tissues from 147 consecutive patients with resected EGFR-mutated lung adenocarcinomas treated at Sun Yat-Sen University Cancer Center were analyzed by next-generation sequencing (NGS). Associations between mutation status, patient baseline characteristics, and survival outcomes (disease-free survival [DFS] and overall survival [OS]) after surgical resection were analyzed. RESULTS TP53 and protein kinase D (PKD) mutations were the two most frequently observed co-mutations in this cohort. Dual PKD/EGFR and TP53/EGFR mutations were found in 39 (27%) and 72 patients (49%), respectively, with dual TP53/EGFR mutations more commonly observed in male patients (P = 0.021). Both TP53 (hazard ratio [HR] 2.08, 95% confidence interval [CI] 1.23-3.54, P = 0.007) and PKD co-mutations (HR 1.72, 95% CI 1.01-2.93, P = 0.044) were associated with shorter DFS, but not OS, in univariate analysis. In multivariate analysis, patients harboring PKD/TP53 co-mutations had shorter DFS compared with PKD-/TP53- cases (HR 2.49, 95% CI 1.15-5.37, P = 0.02). In a subgroup of never-smokers, TP53 co-mutations were associated with significantly worse OS (HR 50.11, 95% CI 2.39-1049.83, P = 0.012). CONCLUSION TP53 and PKD mutations were the two most frequently observed co-mutations in resected EGFR-mutated lung adenocarcinoma. Both mutations were associated with poorer prognoses in affected patients.
Collapse
Affiliation(s)
- Di-Han Liu
- Department of Thoracic Surgery, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.,Lung Cancer Research Center, Sun Yat-Sen University, Guangzhou, China
| | - Ze-Rui Zhao
- Department of Thoracic Surgery, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.,Lung Cancer Research Center, Sun Yat-Sen University, Guangzhou, China
| | - Yao-Bin Lin
- Department of Thoracic Surgery, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.,Lung Cancer Research Center, Sun Yat-Sen University, Guangzhou, China
| | - Wen-Jie Zhou
- Department of Thoracic Surgery, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.,Lung Cancer Research Center, Sun Yat-Sen University, Guangzhou, China
| | - Jing-Yu Hou
- Department of Thoracic Surgery, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.,Lung Cancer Research Center, Sun Yat-Sen University, Guangzhou, China
| | - Zheng-Hao Ye
- Department of Thoracic Surgery, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.,Lung Cancer Research Center, Sun Yat-Sen University, Guangzhou, China
| | - Hao Long
- Department of Thoracic Surgery, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China. .,Lung Cancer Research Center, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
13
|
Zhao S, Li P, Wang P, Yang J, Song P, Zhang D, Zhou G. Nurr1 Promotes Lung Cancer Apoptosis Via Enhancing Mitochondrial Stress and p53-Drp1 Pathway. Open Life Sci 2019; 14:262-274. [PMID: 33817160 PMCID: PMC7874811 DOI: 10.1515/biol-2019-0030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Mitochondrial homeostasis is vital for the progression of lung cancer. Nurr1 has been identified as a novel mediator of mitochondrial homeostasis in several types of cancers. The aim of our study was to investigate whether Nurr1 modulates the viability of A549 lung cancer cells by inducing mitochondrial dysfunction, with a focus on the p53-Drp1 signaling pathway. METHODS western blotting, ELISA and immunofluorescence assay was used to verify the alterations of cell death. siRNA was used to determine the role of p53-Drp1 pathway in lung cancer death. RESULTS Nurr1 was downregulated in A549 lung cancer cells compared to normal pulmonary epithelial cells. Interestingly, overexpression of Nurr1 reduced the viability of A549 lung cancer cells by activating apoptosis and mitochondrial stress. At the molecular level, we provide data to support the regulatory effects of Nurr1 on the p53-Drp1 signaling pathway. Blockade of the p53-Drp1 signaling pathway abolished the proapoptotic action of Nurr1 on A549 cells and sustained mitochondrial homeostasis. CONCLUSION Taken together, our results depict the tumor-suppressive role played by Nurr1 in A549 lung cancer in vitro and show that the anticancer effects of Nurr1 are executed via triggering of mitochondrial dysfunction and activation of the p53-Drp1 signaling pathway.
Collapse
Affiliation(s)
- Shu Zhao
- Department of Oncology, the Second Medical Center, Chinese PLA (People’s Liberation Army)General Hospital, Beijing, 100853,China
| | - Peng Li
- Department of Oncology, the Second Medical Center, Chinese PLA (People’s Liberation Army)General Hospital, Beijing, 100853,China
| | - Peng Wang
- Department of Oncology, the Second Medical Center, Chinese PLA (People’s Liberation Army)General Hospital, Beijing, 100853,China
| | - Jing Yang
- Department of Oncology, the Second Medical Center, Chinese PLA (People’s Liberation Army)General Hospital, Beijing, 100853,China
| | - Peng Song
- Department of Oncology, the Second Medical Center, Chinese PLA (People’s Liberation Army)General Hospital, Beijing, 100853,China
| | - Dong Zhang
- Department of Oncology, the Second Medical Center, Chinese PLA (People’s Liberation Army)General Hospital, Beijing, 100853,China
| | - Gang Zhou
- Department of Oncology, the Second Medical Center, Chinese PLA (People’s Liberation Army)General Hospital, Beijing, 100853,China
| |
Collapse
|
14
|
Zou J, Chen Z, Wei X, Chen Z, Fu Y, Yang X, Chen D, Wang R, Jenner P, Lu JH, Li M, Zhang Z, Tang B, Jin K, Wang Q. Cystatin C as a potential therapeutic mediator against Parkinson's disease via VEGF-induced angiogenesis and enhanced neuronal autophagy in neurovascular units. Cell Death Dis 2017; 8:e2854. [PMID: 28569795 PMCID: PMC5520899 DOI: 10.1038/cddis.2017.240] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/18/2017] [Accepted: 04/26/2017] [Indexed: 02/06/2023]
Abstract
Cystatin C (CYS C, Cst3) is an endogenous cysteine protease inhibitor that plays neuroprotective roles in neurodegenerative diseases. We aimed to explore the association of CYS C with Parkinson’s disease (PD) models and investigate its involvement in the role of neurovascular units (NVUs) in PD neuro-pathogenesis. We used A53T α-synuclein (SNCA) transgenic mice and 6-hydroxydopamine-lesioned DAergic PC12 cells as experimental PD models to investigate the mechanisms behind this association. The injections of CYS C were administered to the right substantia nigra (SN) of A53T SNCA transgenic mice to measure the effects of CYS C in transgenic A53T SNCA mice. To explore the angiogenesis in vivo and in vitro, we used the chick embryo chorioallantoic membrane (CAM) assay and tube formation (TF) assay. We found that CYS C has a neuroprotective effect in this in vivo PD model. We observed increased VEGF, NURR1 and autophagy markers LC3B and decreased SNCA and apoptosis marker cleaved CASP3 in different brain regions of CYS C-treated A53T SNCA transgenic mice. In vitro, we observed that CYS C-induced VEGF, a secreted protein, attenuated 6-OHDA-lesioned DAergic PC12 cell degeneration by regulating p-PKC-α/p-ERK1/2-Nurr1 signaling and inducing autophagy. VEGF-mediated angiogenesis was markedly enhanced in the conditioned media of 6-OHDA-lesioned PC12 cells with CYS C-overexpression, whereas blockage of autophagy in CYS C-overexpressing PC12 cells significantly downregulated VEGF expression and the associated angiogenesis. Our data indicate that CYS C displays dual neuronal–vascular functions, promoting PC12 cell survival and angiogenesis via regulating the level of secreted VEGF in NVUs. Our study provides evidence that may aid in the development of an alternative approach for the treatment of PD through modulation of CYS C-mediated neuronal-vascular pathways.
Collapse
Affiliation(s)
- Jing Zou
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Zhaoyu Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Xiaobo Wei
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Zhigang Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Yongmei Fu
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Xiaoyan Yang
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Dan Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Rui Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Peter Jenner
- Neurodegenerative Diseases Research Group, Faculty of Health Sciences and Medicine, King's College, London SE1 1UL, UK
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, China
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Zhuohua Zhang
- Department of Neurology, Xiangya School of Medicine and The State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410078, China
| | - Beisha Tang
- Department of Neurology, Xiangya School of Medicine and The State Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan 410078, China
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Qing Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| |
Collapse
|
15
|
Adenovirus-mediated expression of orphan nuclear receptor NR4A2 targeting hepatic stellate cell attenuates liver fibrosis in rats. Sci Rep 2016; 6:33593. [PMID: 27646469 PMCID: PMC5028713 DOI: 10.1038/srep33593] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/24/2016] [Indexed: 01/20/2023] Open
Abstract
Liver fibrosis is a wound-healing response characterized with the accumulation of extracellular matrix (ECM). And hepatic stellate cells (HSCs) are the principal cell source of ECM. NR4A2 (Nurr1) is a member of orphan nuclear receptor NR4A family and acts as transcription factor. It participates in regulating cell differentiation, proliferation and apoptosis. We previously demonstrated that NR4A2 expression in fibrotic liver reduced significantly compared with normal liver and NR4A2 knockout in HSCs promoted ECM production. In the present study we explored the role of NR4A2 on liver fibrosis. Studies in cultured HSCs demonstrated that NR4A2 over-expression suppressed the activation of HSCs, such as ECM production and invasion ability. Moreover cell cycle was arrested, cell apoptosis was promoted and cell signaling pathway was influenced. Adenovirus-mediated delivery of NR4A2 in rats ameliorated significantly dimethylnitrosamine (DMN) induced liver fibrosis. The In vivo experiments produced results consistent with in vitro experiments. Taken together these results demonstrate NR4A2 enhancement attenuates liver fibrosis via suppressing the activation of HSCs and NR4A2 may be an ideal target for anti-fibrotic therapy.
Collapse
|
16
|
Wang J, Yang ZH, Chen H, Li HH, Chen LY, Zhu Z, Zou Y, Ding CC, Yang J, He ZW. Nemo-like kinase as a negative regulator of nuclear receptor Nurr1 gene transcription in prostate cancer. BMC Cancer 2016; 16:257. [PMID: 27036119 PMCID: PMC4815267 DOI: 10.1186/s12885-016-2291-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 03/22/2016] [Indexed: 02/07/2023] Open
Abstract
Background Nurr1, a member of the orphan receptor family, plays an important role in several types of cancer. Our previous work demonstrated that increased expression of Nurr1 plays a significant role in the initiation and progression of prostate cancer (PCa), though the mechanisms for regulation of Nurr1 expression remain unknown. In this study, we investigated the hypothesis that Nemo-like kinase (NLK) is a key regulator of Nurr1 expression in PCa. Methods Immunohistochemistry and Western blot analysis were used to evaluate levels of NLK and Nurr1 in prostatic tissues and cell lines. The effects of overexpression or knockdown of Nurr1 were evaluated in PCa cells through use of PCR, Western blots and promoter reporter assays. The role of Nurr1 promoter cis element was studied by creation of two mutant Nurr1 promoter luciferase constructs, one with a mutated NF-κB binding site and one with a mutated CREB binding site. In addition, three specific inhibitors were used to investigate the roles of these proteins in transcriptional activation of Nurr1, including BAY 11–7082 (NF-κB inhibitor), KG-501 (CREB inhibitor) and ICG-001 (CREB binding protein, CBP, inhibitor). The function of CBP in NLK-mediated regulation of Nurr1 expression was investigated using immunofluorescence, co-immunoprecipitation (Co-IP) and chromatin immunoprecipitation assays (ChIPs). Results NLK expression was inversely correlated with Nurr1 expression in prostate cancer tissues and cell lines. Overexpression of NLK suppressed Nurr1 promoter activity, leading to downregulation of Nurr1 expression. In contrast, knockdown of NLK demonstrated opposite results, leading to upregulation of Nurr1. When compared with the wild-type Nurr1 promoter, mutation of NF-κB- and CREB-binding sites of the Nurr1 promoter region significantly reduced the upregulation of Nurr1 induced by knockdown of NLK in LNCaP cells; treatment with inhibitors of CREB, CBP and NF-κB led to similar results. We also found that NLK directly interacts with CBP, that knockdown of NLK significantly increases the recruitment of CBP to both NF-κB- and CREB-binding sites, and that regulation of NLK on Nurr1 expression is abrogated by knockdown of CBP. Conclusions Our results suggest that NLK inhibits transcriptional activation of Nurr1 gene by impeding CBP’s role as a co-activator of NF-κB and CREB in prostate cancer.
Collapse
Affiliation(s)
- Jian Wang
- Sino-American Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, 1 Xincheng Road, Dongguan, 523808, China
| | - Zhi-Hong Yang
- Department of Obstetrics and Gynecology, Longgang District Central Hospital of Shenzhen, 1228 Longgang Road, Shenzhen, 518116, China
| | - Hua Chen
- Sino-American Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, 1 Xincheng Road, Dongguan, 523808, China
| | - Hua-Hui Li
- Sino-American Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, 1 Xincheng Road, Dongguan, 523808, China
| | - Li-Yong Chen
- Sino-American Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, 1 Xincheng Road, Dongguan, 523808, China
| | - Zhu Zhu
- Sino-American Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, 1 Xincheng Road, Dongguan, 523808, China
| | - Ying Zou
- Sino-American Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, 1 Xincheng Road, Dongguan, 523808, China
| | - Cong-Cong Ding
- Sino-American Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, 1 Xincheng Road, Dongguan, 523808, China
| | - Jing Yang
- Department of Biochemistry, Liaoning Medical University, 40 Songpo Road, Jinzhou, 121001, China.
| | - Zhi-Wei He
- Sino-American Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, 1 Xincheng Road, Dongguan, 523808, China.
| |
Collapse
|
17
|
Dong J, Li S, Mo JL, Cai HB, Le WD. Nurr1-Based Therapies for Parkinson's Disease. CNS Neurosci Ther 2016; 22:351-9. [PMID: 27012974 DOI: 10.1111/cns.12536] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/12/2016] [Accepted: 02/12/2016] [Indexed: 12/13/2022] Open
Abstract
Previous studies have documented that orphan nuclear receptor Nurr1 (also known as NR4A2) plays important roles in the midbrain dopamine (DA) neuron development, differentiation, and survival. Furthermore, it has been reported that the defects in Nurr1 are associated with Parkinson's disease (PD). Thus, Nurr1 might be a potential therapeutic target for PD. Emerging evidence from in vitro and in vivo studies has recently demonstrated that Nurr1-activating compounds and Nurr1 gene therapy are able not only to enhance DA neurotransmission but also to protect DA neurons from cell injury induced by environmental toxin or microglia-mediated neuroinflammation. Moreover, modulators that interact with Nurr1 or regulate its function, such as retinoid X receptor, cyclic AMP-responsive element-binding protein, glial cell line-derived neurotrophic factor, and Wnt/β-catenin pathway, have the potential to enhance the effects of Nurr1-based therapies in PD. This review highlights the recent progress in preclinical studies of Nurr1-based therapies and discusses the outlook of this emerging therapy as a promising new generation of PD medication.
Collapse
Affiliation(s)
- Jie Dong
- The Center for Translational Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Song Li
- The Center for Translational Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jing-Lin Mo
- The Center for Translational Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Huai-Bin Cai
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Wei-Dong Le
- The Center for Translational Research on Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, China.,Institute of Health Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
18
|
Zhang LM, Sun CC, Mo MS, Cen L, Wei L, Luo FF, Li Y, Li GF, Zhang SY, Yi L, Huang W, Liu ZL, Le WD, Xu PY. Dopamine Agonists Exert Nurr1-inducing Effect in Peripheral Blood Mononuclear Cells of Patients with Parkinson's Disease. Chin Med J (Engl) 2016; 128:1755-60. [PMID: 26112716 PMCID: PMC4733729 DOI: 10.4103/0366-6999.159349] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background: Nurr1 plays an essential role in the development, survival, and function maintenance of midbrain dopaminergic (DA) neurons, and it is a potential target for Parkinson's disease (PD). Nurr1 mRNA can be detected in peripheral blood mononuclear cells (PBMCs), but whether there is any association of altered Nurr1 expression in PBMC with the disease and DA drug treatments remains elusive. This study aimed to measure the Nurr1 mRNA level in PBMC and evaluate the effect of Nurr1 expression by DA agents in vivo and in vitro. Methods: The mRNA levels of Nurr1 in PBMC of four subgroups of 362 PD patients and 193 healthy controls (HCs) using real-time polymerase chain reaction were measured. The nonparametric Mann-Whitney U-test and Kruskal-Wallis test were performed to evaluate the differences between PD and HC, as well as the subgroups of PD. Multivariate linear regression analysis was used to evaluate the independent association of Nurr1 expression with Hoehn and Yahr scale, age, and drug treatments. Besides, the Nurr1 expression in cultured PBMC was measured to determine whether DA agonist pramipexole affects its mRNA level. Results: The relative Nurr1 mRNA levels in DA agonists treated subgroup were significant higher than those in recent-onset cases without any anti-PD treatments (de novo) (P < 0.001) and HC groups (P < 0.010), respectively. Furthermore, the increase in Nurr1 mRNA expression was seen in DA agonist and L-dopa group. Multivariate linear regression showed DA agonists, L-dopa, and DA agonists were independent predictors correlated with Nurr1 mRNA expression level in PBMC. In vitro, in the cultured PBMC treated with 10 μmol/L pramipexole, the Nurr1 mRNA levels were significantly increased by 99.61%, 71.75%, 73.16% in 2, 4, and 8 h, respectively (P < 0.001). Conclusions: DA agonists can induce Nurr1 expression in PBMC, and such effect may contribute to DA agonists-mediated neuroprotection on DA neurons.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Ping-Yi Xu
- Department of Neurology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080; Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangdong 510120, China
| |
Collapse
|
19
|
Jia J, Ye T, Cui P, Hua Q, Zeng H, Zhao D. AP-1 transcription factor mediates VEGF-induced endothelial cell migration and proliferation. Microvasc Res 2016; 105:103-8. [PMID: 26860974 DOI: 10.1016/j.mvr.2016.02.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 01/21/2016] [Accepted: 02/05/2016] [Indexed: 11/16/2022]
Abstract
VEGF, upon binding to its endothelial cell specific receptors VEGF-R1 and VEGF-R2, can induce endothelial cell migration, proliferation and angiogenesis. However, the molecular mechanism of these effects still remains unclear. In this study, we investigated whether VEGF promotes human umbilical vascular endothelial cell (HUVEC) migration and proliferation through activator protein-1 transcription factor (AP-1) family. We first showed that VEGF induces immediate-early genes AP-1 family gene expression differentially with the profound induction of JunB (both mRNA and protein) under various conditions (PBS, DMSO or control adenoviruses). The increase in AP-1 mRNA expression occurs primarily at the transcriptional level. Inhibition of AP-1 DNA binding activity by adenovirus expressing a potent dominant negative form of c-Fos (Afos) significantly attenuated VEGF-induced HUVEC migration and proliferation and cyclin D1 expression. Knockdown of JunB with adenovirus expressing JunB shRNA reduces VEGF-induced JunB expression and attenuated HUVEC migration. However the shJunB-expressing virus has no effect on VEGF-induced cyclin D1 protein expression and proliferation. These results suggest that VEGF-induced endothelial migration is mediated primarily by induction of JunB whereas the promotion of endothelial proliferation by VEGF is mediated by JunB-independent AP-1 family members.
Collapse
Affiliation(s)
- Jing Jia
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02460, USA; Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02460, USA; School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Taiyang Ye
- Divison of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02460, USA; Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02460, USA; Department of Obstetrics and Gynecology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Pengfei Cui
- Divison of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02460, USA; Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02460, USA; Department of General Surgery, Pancreatic Disease Institute, Union Hospital, Huazhong University of Science & Technology, Wuhan, China
| | - Qian Hua
- School of Preclinical Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huiyan Zeng
- Divison of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02460, USA; Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02460, USA
| | - Dezheng Zhao
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02460, USA; Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02460, USA.
| |
Collapse
|
20
|
Chen P, Li J, Huo Y, Lu J, Wan L, Li B, Gan R, Guo C. Orphan nuclear receptor NR4A2 inhibits hepatic stellate cell proliferation through MAPK pathway in liver fibrosis. PeerJ 2015; 3:e1518. [PMID: 26713258 PMCID: PMC4690364 DOI: 10.7717/peerj.1518] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/28/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatic stellate cells (HSCs) play a crucial role in liver fibrosis, which is a pathological process characterized by extracellular matrix accumulation. NR4A2 is a nuclear receptor belonging to the NR4A subfamily and vital in regulating cell growth, metabolism, inflammation and other biological functions. However, its role in HSCs is unclear. We analyzed NR4A2 expression in fibrotic liver and stimulated HSCs compared with control group and studied the influence on cell proliferation, cell cycle, cell apoptosis and MAPK pathway after NR4A2 knockdown. NR4A2 expression was examined by real-time polymerase chain reaction, Western blotting, immunohistochemistry and immunofluorescence analyses. NR4A2 expression was significantly lower in fibrotic liver tissues and PDGF BB or TGF-β stimulated HSCs compared with control group. After NR4A2 knockdown α-smooth muscle actin and Col1 expression increased. In addition, NR4A2 silencing led to the promotion of cell proliferation, increase of cell percentage in S phase and reduced phosphorylation of ERK1/2, P38 and JNK in HSCs. These results indicate that NR4A2 can inhibit HSC proliferation through MAPK pathway and decrease extracellular matrix in liver fibrogenesis. NR4A2 may be a promising therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Pengguo Chen
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China ; Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Jie Li
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Yan Huo
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Jin Lu
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Lili Wan
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Bin Li
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Run Gan
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Cheng Guo
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China ; Shanghai Jiao Tong University School of Medicine , Shanghai , China
| |
Collapse
|
21
|
Singh NK, Kotla S, Kumar R, Rao GN. Cyclic AMP Response Element Binding Protein Mediates Pathological Retinal Neovascularization via Modulating DLL4-NOTCH1 Signaling. EBioMedicine 2015; 2:1767-84. [PMID: 26870802 PMCID: PMC4740322 DOI: 10.1016/j.ebiom.2015.09.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/08/2015] [Accepted: 09/23/2015] [Indexed: 11/29/2022] Open
Abstract
Retinal neovascularization is the most common cause of moderate to severe vision loss in all age groups. Despite the use of anti-VEGFA therapies, this complication continues to cause blindness, suggesting a role for additional molecules in retinal neovascularization. Besides VEGFA and VEGFB, hypoxia induced VEGFC expression robustly. Based on this finding, we tested the role of VEGFC in pathological retinal angiogenesis. VEGFC induced proliferation, migration, sprouting and tube formation of human retinal microvascular endothelial cells (HRMVECs) and these responses require CREB-mediated DLL4 expression and NOTCH1 activation. Furthermore, down regulation of VEGFC levels substantially reduced tip cell formation and retinal neovascularization in vivo. In addition, we observed that CREB via modulating the DLL4-NOTCH1 signaling mediates VEGFC-induced tip cell formation and retinal neovascularization. In regard to upstream mechanism, we found that down regulation of p38β levels inhibited hypoxia-induced CREB-DLL4-NOTCH1 activation, tip cell formation, sprouting and retinal neovascularization. Based on these findings, it may be suggested that VEGFC besides its role in the regulation of lymphangiogenesis also plays a role in pathological retinal angiogenesis and this effect depends on p38β and CREB-mediated activation of DLL4-NOTCH1 signaling.
Collapse
Affiliation(s)
- Nikhlesh K Singh
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sivareddy Kotla
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Raj Kumar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
22
|
Molecular Interactions between NR4A Orphan Nuclear Receptors and NF-κB Are Required for Appropriate Inflammatory Responses and Immune Cell Homeostasis. Biomolecules 2015; 5:1302-18. [PMID: 26131976 PMCID: PMC4598753 DOI: 10.3390/biom5031302] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/16/2015] [Accepted: 06/16/2015] [Indexed: 12/22/2022] Open
Abstract
Appropriate innate and adaptive immune responses are essential for protection and resolution against chemical, physical or biological insults. Immune cell polarization is fundamental in orchestrating distinct phases of inflammation, specifically acute phase responses followed by resolution and tissue repair. Dysregulation of immune cell and inflammatory responses is a hallmark of multiple diseases encompassing atherosclerosis, rheumatoid arthritis, psoriasis and metabolic syndromes. A master transcriptional mediator of diverse inflammatory signaling and immune cell function is NF-κB, and altered control of this key regulator can lead to an effective switch from acute to chronic inflammatory responses. Members of the nuclear receptor (NR) superfamily of ligand-dependent transcription factors crosstalk with NF-κB to regulate immune cell function(s). Within the NR superfamily the NR4A1-3 orphan receptors have emerged as important regulators of immune cell polarization and NF-κB signaling. NR4A receptors modulate NF-κB activity in a dynamic fashion, either repressing or enhancing target gene expression leading to altered inflammatory outcome. Here we will discuss the pivotal role NR4A’s receptors play in orchestrating immune cell homeostasis through molecular crosstalk with NF-κB. Specifically, we will examine such NR4A/NF-κB interactions within the context of distinct cell phenotypes, including monocyte, macrophage, T cells, endothelial, and mesenchymal cells, which play a role in inflammation-associated disease. Finally, we review the therapeutic potential of altering NR4A/NF-κB interactions to limit hyper-inflammatory responses in vivo.
Collapse
|
23
|
Clinicopathological significance of orphan nuclear receptor Nurr1 expression in gastric cancer. Clin Transl Oncol 2015; 17:788-94. [PMID: 26022133 DOI: 10.1007/s12094-015-1305-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/16/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Gastric cancer is the fourth most common cancer and the second leading cause of cancer-related deaths worldwide. Gastric cancer is characterized by high levels of invasion and metastasis. Increasing attention is being focused on discovering molecular markers for the diagnosis of gastric cancer and for predicting its prognosis. The objective of the present study was to evaluate Nurr1 expression in gastric cancer and to assess its correlation with clinicopathological parameters and prognosis in gastric cancer patients. METHODS Tissue samples were obtained from 120 gastric cancer patients. We investigated Nurr1 expression in human normal and gastric cancer tissues using real-time reverse transcription polymerase chain reaction (qRT-PCR), western blotting, and immunohistochemistry. We determined the association between Nurr1 and recurrence, prognosis and patient clinicopathological parameters. Univariate and multivariate survival analyses with a Cox's proportional hazards regression model were used to identify independent factors related to recurrence and prognosis. RESULTS The immunohistochemical, qRT-PCR and western blot analyses revealed that Nurr1 expression was increased in gastric cancer tissues compared with normal gastric tissue (P < 0.05). Nurr1 expression was significantly correlated with the tumor size, depth of tumor invasion, lymph node metastasis, recurrence, and distant metastasis of gastric cancer (P < 0.05). Moreover, Nurr1-high patients also exhibited poorer overall survival (OS) and disease-free survival compared with Nurr1-low patients (P < 0.01). The univariate and multivariate survival analyses suggested that Nurr1 expression (P = 0.011), histology (P = 0.018), depth of tumor invasion (P = 0.037), and presence of lymph node metastasis (P = 0.031) were independent prognostic factors for recurrence. In addition, Nurr1 expression (P = 0.007), depth of tumor invasion (P = 0.014), lymph node metastasis (P = 0.044), distant metastasis (P = 0.023), and recurrence (P = 0.011) were independent prognostic factors of OS in gastric cancer patients. CONCLUSIONS The Nurr1 protein may be useful as a marker of recurrence, metastasis, and poor prognosis following curative resection in patients with gastric cancer.
Collapse
|
24
|
Xie L, Mouillet JF, Chu T, Parks WT, Sadovsky E, Knöfler M, Sadovsky Y. C19MC microRNAs regulate the migration of human trophoblasts. Endocrinology 2014; 155:4975-85. [PMID: 25211593 PMCID: PMC4239420 DOI: 10.1210/en.2014-1501] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Early in pregnancy, trophoblast invasion into the decidua and inner myometrium is essential for establishment of proper implantation, maternal-fetal exchange, and immunological tolerance of the feto-placental allograft. Unlike villous trophoblasts (VTs), extravillous trophoblasts (EVTs) are unique in their capacity to invade the maternal decidua and myometrium. The largest human microRNA (miRNA) gene cluster, the chromosome 19 miRNA cluster (C19MC), is expressed almost exclusively in the placenta and, rarely, in certain tumors and undifferentiated cells. In the work reported here, we found that the expression of C19MC miRNAs is higher in VTs than in EVTs. Using a bacterial artificial chromosome (BAC)-mediated overexpression of C19MC miRNAs in an EVT-derived cell line, which does not naturally express these miRNAs, we found that C19MC miRNAs selectively attenuate cell migration without affecting cell proliferation or apoptosis. A microarray analysis revealed that C19MC miRNAs regulate target transcripts related to cellular movement. Our data also implicated a specific C19MC member, miR-519d, indirectly regulating the EVT invasive phenotype by targeting CXCL6, NR4A2 and FOXL2 transcripts through a 3'UTR miRNA-responsive element. Together, our data suggest a role for C19MC miRNAs in modulating the migration of EVTs.
Collapse
Affiliation(s)
- Lan Xie
- Magee-Womens Research Institute (L.X., J.-F.M., T.C., W.T.P., E.S., Y.S.), Department of OBGYN and Reproductive Sciences, University of Pittsburgh, Pennsylvania 15213; Medical Systems Biology Research Center (L.X.), Department of Biomedical Engineering, Tsinghua University School of Medicine, Beijing 100084 China; Department of Obstetrics and Fetal-Maternal Medicine (M.K.), Reproductive Biology Unit, Medical University of Vienna, Vienna, A-1090 Austria; and Department of Microbiology and Molecular Genetics (Y.S.), University of Pittsburgh, Pennsylvania 15213
| | | | | | | | | | | | | |
Collapse
|
25
|
The interplay of NR4A receptors and the oncogene-tumor suppressor networks in cancer. Cell Signal 2014; 27:257-66. [PMID: 25446259 DOI: 10.1016/j.cellsig.2014.11.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 10/25/2014] [Accepted: 11/08/2014] [Indexed: 12/11/2022]
Abstract
Nuclear receptor (NR) subfamily 4 group A (NR4A) is a family of three highly homologous orphan nuclear receptors that have multiple physiological and pathological roles, including some in cancer. These NRs are reportedly dysregulated in multiple cancer types, with many studies demonstrating pro-oncogenic roles for NR4A1 (Nur77) and NR4A2 (Nurr1). Additionally, NR4A1 and NR4A3 (Nor-1) are described as tumor suppressors in leukemia. The dysregulation and functions of the NR4A members are due to many factors, including transcriptional regulation, protein-protein interactions, and post-translational modifications. These various levels of intracellular regulation result from the signaling cross-talk of the NR4A members with various signaling pathways, many of which are relevant to cancer and likely explain the family members' functions in oncogenesis and tumor suppression. In this review, we discuss the multiple functions of the NR4A receptors in cancer and summarize a growing body of scientific literature that describes the interconnectedness of the NR4A receptors with various oncogene and tumor suppressor pathways.
Collapse
|
26
|
The p38 MAPK-regulated PKD1/CREB/Bcl-2 pathway contributes to selenite-induced colorectal cancer cell apoptosis in vitro and in vivo. Cancer Lett 2014; 354:189-99. [DOI: 10.1016/j.canlet.2014.08.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 08/05/2014] [Accepted: 08/06/2014] [Indexed: 12/20/2022]
|
27
|
Sturtzel C, Testori J, Schweighofer B, Bilban M, Hofer E. The transcription factor MEF2C negatively controls angiogenic sprouting of endothelial cells depending on oxygen. PLoS One 2014; 9:e101521. [PMID: 24988463 PMCID: PMC4079651 DOI: 10.1371/journal.pone.0101521] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 06/09/2014] [Indexed: 11/18/2022] Open
Abstract
The MADS box transcription factor MEF2C has been detected by us to be upregulated by the angiogenic factors VEGF-A and bFGF in endothelial cells. We have here investigated its potential role for angiogenesis. MEF2C was surprisingly found to strongly inhibit angiogenic sprouting, whereas a dominant negative mutant rather induced sprouting. The factor mainly affected migratory processes of endothelial cells, but not proliferation. In gene profiling experiments we delineated the alpha-2-macroglobulin gene to be highly upregulated by MEF2C. Further data confirmed that MEF2C in endothelial cells indeed induces alpha-2-macroglobulin mRNA as well as the secretion of alpha-2-macroglobulin and that conditioned supernatants of cells overexpressing MEF2C inhibit sprouting. Alpha-2-macroglobulin mediates, at least to a large extent, the inhibitory effects of MEF2C as is shown by knockdown of alpha-2-macroglobulin mRNA by lentiviral shRNA expression which reduces the inhibitory effect. However, under hypoxic conditions the VEGF-A/bFGF-mediated upregulation of MEF2C is reduced and the production of alpha-2-macroglobulin largely abolished. Taken together, this suggests that the MEF2C/alpha-2-macroglobulin axis functions in endothelial cells as a negative feed-back mechanism that adapts sprouting activity to the oxygen concentration thus diminishing inappropriate and excess angiogenesis.
Collapse
Affiliation(s)
- Caterina Sturtzel
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Julia Testori
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Schweighofer
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Erhard Hofer
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
28
|
Camós S, Gubern C, Sobrado M, Rodríguez R, Romera V, Moro M, Lizasoain I, Serena J, Mallolas J, Castellanos M. The high-mobility group I-Y transcription factor is involved in cerebral ischemia and modulates the expression of angiogenic proteins. Neuroscience 2014; 269:112-30. [DOI: 10.1016/j.neuroscience.2014.03.036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 03/07/2014] [Accepted: 03/18/2014] [Indexed: 12/24/2022]
|
29
|
Theranostic tumor homing nanocarriers for the treatment of lung cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 10:1053-63. [PMID: 24355163 DOI: 10.1016/j.nano.2013.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 11/25/2013] [Accepted: 12/07/2013] [Indexed: 12/31/2022]
Abstract
UNLABELLED The drugs/strategies to selectively inhibit tumor blood supply have generated interest in recent years for enhancement of cancer therapeutics. The objective of this study was to formulate tumor homing PEGylated CREKA peptide conjugated theranostic nanoparticles of DIM-C-pPhC6H5 (DIM-P) and investigate in vivo antitumor activity as well as evaluate the targeted efficiency to lung tumors using imaging techniques. DIM-P loaded Nanoparticles (NCs-D) were prepared using lipids, and DOGS-NTA-Ni and the surface of NCs-D were modified with PEGylated CREKA peptide (PCNCs-D). PCNCs-D showed 3 fold higher binding to clotted plasma proteins in tumor vasculature compared to NCs-D. PCNCs-D showed 26%±4% and 22%±5% increase in tumor reduction compared to NCs-D in metastatic and orthotopic models respectively. In-vivo imaging studies showed ~40 folds higher migration of PCNCs-Di in tumor vasculature than NCs-Di. Our studies demonstrate the role of PCNCs-D as theranostic tumor homing drug delivery and imaging systems for lung cancer diagnosis and treatment. FROM THE CLINICAL EDITOR This study demonstrates a very efficient delivery system to address lung cancer growth through blood supply inhibition.
Collapse
|
30
|
Corsini M, Moroni E, Ravelli C, Andrés G, Grillo E, Ali IH, Brazil DP, Presta M, Mitola S. Cyclic adenosine monophosphate-response element-binding protein mediates the proangiogenic or proinflammatory activity of gremlin. Arterioscler Thromb Vasc Biol 2013; 34:136-45. [PMID: 24233491 DOI: 10.1161/atvbaha.113.302517] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Angiogenesis and inflammation are closely related processes. Gremlin is a novel noncanonical vascular endothelial growth factor receptor-2 (VEGFR2) ligand that induces a proangiogenic response in endothelial cells (ECs). Here, we investigated the role of the cyclic adenosine monophosphate-response element (CRE)-binding protein (CREB) in mediating the proinflammatory and proangiogenic responses of ECs to gremlin. APPROACH AND RESULTS Gremlin induces a proinflammatory response in ECs, leading to reactive oxygen species and cyclic adenosine monophosphate production and the upregulation of proinflammatory molecules involved in leukocyte extravasation, including chemokine (C-C motif) ligand-2 (Ccl2) and Ccl7, chemokine (C-X-C motif) ligand-1 (Cxcl1), vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1). Accordingly, gremlin induces the VEGFR2-dependent phosphorylation, nuclear translocation, and transactivating activity of CREB in ECs. CREB activation mediates the early phases of the angiogenic response to gremlin, including stimulation of EC motility and permeability, and leads to monocyte/macrophage adhesion to ECs and their extravasation. All these effects are inhibited by EC transfection with a dominant-negative CREB mutant or with a CREB-binding protein-CREB interaction inhibitor that competes for CREB/CRE binding. Also, both recombinant gremlin and gremlin-expressing tumor cells induce proinflammatory/proangiogenic responses in vivo that are suppressed by the anti-inflammatory drug hydrocortisone. Similar effects were induced by the canonical VEGFR2 ligand VEGF-A165. CONCLUSIONS Together, the results underline the tight cross-talk between angiogenesis and inflammation and demonstrate a crucial role of CREB activation in the modulation of the VEGFR2-mediated proinflammatory/proangiogenic response of ECs to gremlin.
Collapse
Affiliation(s)
- Michela Corsini
- From the Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (M.C., E.M., C.R., E.G., M.P., S.M.); Electron Microscopy Unit, Centro de Biologia Molecular Severo Ochoa, Campus Cantoblanco, Madrid, Spain (G.A.); and Centre for Experimental Medicine, Queen's University Belfast, ICS-A, Grosvenor Road, Belfast BT12 6BA, UK (I.H.A., D.P.B.)
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
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
|
32
|
McMorrow JP, Crean D, Gogarty M, Smyth A, Connolly M, Cummins E, Veale D, Fearon U, Tak PP, Fitzgerald O, Murphy EP. Tumor necrosis factor inhibition modulates thrombospondin-1 expression in human inflammatory joint disease through altered NR4A2 activity. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1243-1257. [PMID: 23933487 DOI: 10.1016/j.ajpath.2013.06.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 06/19/2013] [Accepted: 06/24/2013] [Indexed: 01/07/2023]
Abstract
We examined thrombospondin-1 (THBS1, alias TSP-1) expression in human synovial tissue (ST) during the resolution phase of chronic inflammation and elucidated its transcriptional regulation by the orphan receptor 4A2 (NR4A2). In vivo, rheumatoid arthritis (RA) serum and ST revealed altered expression levels and tissue distribution of TSP-1. After anti-tumor necrosis factor therapy, a reciprocal relationship between TSP-1 and NR4A2 expression levels was measured in patients with clinical and ST responses to biological treatment. In vitro, primary RA fibroblast-like synoviocytes (FLSs) expressed minimal TSP-1 mRNA levels with high transcript levels of NR4A2, vascular endothelial growth factor (VEGF), and IL-8 measured. Hypoxic modulation of RA FLSs resulted in inverse expression levels of TSP-1 compared with NR4A2, IL-8, and VEGF. Ectopic NR4A2 expression led to reduced TSP-1 mRNA and protein levels with concomitant increases in proangiogenic mediators. NR4A2 transcriptional activity, independent of DNA binding, repressed the hTSP-1 promoter leading to reduced mRNA and protein release in immortalized K4IM FLSs. Bioinformatic and deletion studies identified a 5' region of the TSP-1 promoter repressed by NR4A2 and proangiogenic transcription factors, including NF-κB and Ets1/2. Stable depletion of NR4A2 levels resulted in a shift in the TSP-1/VEGF expression ratio. Thus, modulation of TSP-1 expression is achieved through anti-tumor necrosis factor therapy effects on specific transcriptional networks, suggesting that enhanced TSP-1 expression may help restore tissue homeostasis during resolution of inflammation.
Collapse
Affiliation(s)
- Jason P McMorrow
- UCD Veterinary Sciences Centre, University College Dublin, Belfield, Ireland
| | - Daniel Crean
- UCD Veterinary Sciences Centre, University College Dublin, Belfield, Ireland
| | - Martina Gogarty
- UCD Veterinary Sciences Centre, University College Dublin, Belfield, Ireland
| | - Aisling Smyth
- UCD Veterinary Sciences Centre, University College Dublin, Belfield, Ireland
| | - Mary Connolly
- Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland; Department of Rheumatology, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Eoin Cummins
- Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland
| | - Douglas Veale
- Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland; Department of Rheumatology, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Ursula Fearon
- Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland; Department of Rheumatology, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Paul P Tak
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Oliver Fitzgerald
- Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland; Department of Rheumatology, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Evelyn P Murphy
- UCD Veterinary Sciences Centre, University College Dublin, Belfield, Ireland; Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Ireland.
| |
Collapse
|
33
|
Kurakula K, Hamers AAJ, de Waard V, de Vries CJM. Nuclear Receptors in atherosclerosis: a superfamily with many 'Goodfellas'. Mol Cell Endocrinol 2013; 368:71-84. [PMID: 22664910 DOI: 10.1016/j.mce.2012.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 05/23/2012] [Accepted: 05/25/2012] [Indexed: 01/07/2023]
Abstract
Nuclear Receptors form a superfamily of 48 transcription factors that exhibit a plethora of functions in steroid hormone signaling, regulation of metabolism, circadian rhythm and cellular differentiation. In this review, we describe our current knowledge on the role of Nuclear Receptors in atherosclerosis, which is a multifactorial disease of the vessel wall. Various cell types are involved in this chronic inflammatory pathology in which multiple cellular processes and numerous genes are dysregulated. Systemic risk factors for atherosclerosis are among others adverse blood lipid profiles, enhanced circulating cytokine levels, as well as increased blood pressure. Since many Nuclear Receptors modulate lipid profiles or regulate blood pressure they indirectly affect atherosclerosis. In the present review, we focus on the functional involvement of Nuclear Receptors within the atherosclerotic vessel wall, more specifically on their modulation of cellular functions in endothelial cells, smooth muscle cells and macrophages. Collectively, this overview shows that most of the Nuclear Receptors are athero-protective in atherosclerotic lesions.
Collapse
Affiliation(s)
- Kondababu Kurakula
- Department of Medical Biochemistry, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
34
|
Han YF, Cao GW. Role of nuclear receptor NR4A2 in gastrointestinal inflammation and cancers. World J Gastroenterol 2012; 18:6865-73. [PMID: 23322982 PMCID: PMC3531668 DOI: 10.3748/wjg.v18.i47.6865] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 08/27/2012] [Accepted: 09/12/2012] [Indexed: 02/06/2023] Open
Abstract
NR4A2 is a transcription factor belonging to the steroid orphan nuclear receptor superfamily. It was originally considered to be essential in the generation and maintenance of dopaminergic neurons, and associated with neurological disorders such as Parkinson’s disease. Recently, NR4A2 has been found to play a critical role in some inflammatory diseases and cancer. NR4A2 can be efficiently trans-activated by some proinflammatory cytokines, such as tumor necrosis factor-α, interleukin-1β, and vascular endothelial growth factor (VEGF). The nuclear factor-κB signaling pathway serves as a principal regulator of inducible NR4A expression in immune cells. NR4A2 can trans-activate Foxp3, a hallmark specifically expressed in regulatory T (Treg) cells, and plays a critical role in the differentiation, maintenance, and function of Treg cells. NR4A2 in T lymphocytes is pivotal for Treg cell induction and suppression of aberrant induction of Th1 under physiological and pathological conditions. High density of Foxp3+ Treg cells is significantly associated with gastrointestinal inflammation, tumor immune escape, and disease progression. NR4A2 is produced at high levels in CD133+ colorectal carcinoma (CRC) cells and significantly upregulated by cyclooxygenase-2-derived prostaglandin E2 in a cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)-dependent manner in CRC cells. The cAMP/PKA signaling pathway is the common pathway of NR4A2-related inflammation and cancer. NR4A2 trans-activates osteopontin, a direct target of the Wnt/β-catenin pathway associated with CRC invasion, metastasis, and poor prognosis. Knockdown of endogenous NR4A2 expression attenuates VEGF-induced endothelial cell proliferation, migration and in vivo angiogenesis. Taken together, NR4A2 emerges as an important nuclear factor linking gastrointestinal inflammation and cancer, especially CRC, and should serve as a candidate therapeutic target for inflammation-related gastrointestinal cancer.
Collapse
|
35
|
Abstract
Bone marrow (BM) cells depend on their niche for growth and survival. However, the genes modulated by niche stimuli have not been discriminated yet. For this purpose, we investigated BM aspirations from patients with various hematological malignancies. Each aspirate was fractionated, and the various samples were fixed at different time points and analyzed by microarray. Identification of niche-modulated genes relied on sustained change in expression following loss of niche regulation. Compared with the reference (‘authentic') samples, which were fixed immediately following aspiration, the BM samples fixed after longer stay out-of-niche acquired numerous changes in gene-expression profile (GEP). The overall genes modulated included a common subset of functionally diverse genes displaying prompt and sustained ‘switch' in expression irrespective of the tumor type. Interestingly, the ‘switch' in GEP was reversible and turned ‘off-and-on' again in culture conditions, resuming cell–cell–matrix contact versus respread into suspension, respectively. Moreover, the resuming of contact prolonged the survival of tumor cells out-of-niche, and the regression of the ‘contactless switch' was followed by induction of a new set of genes, this time mainly encoding extracellular proteins including angiogenic factors and extracellular matrix proteins. Our data set, being unique in authentic expression design, uncovered niche-modulated and niche-modulating genes capable of controlling homing, expansion and angiogenesis.
Collapse
|
36
|
Mix KS, McMahon K, McMorrow JP, Walkenhorst DE, Smyth AM, Petrella BL, Gogarty M, Fearon U, Veale D, Attur MG, Abramson SB, Murphy EP. Orphan nuclear receptor NR4A2 induces synoviocyte proliferation, invasion, and matrix metalloproteinase 13 transcription. ACTA ACUST UNITED AC 2012; 64:2126-36. [PMID: 22275273 DOI: 10.1002/art.34399] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To address the role of the nuclear receptor 4A (NR4A) family of orphan nuclear receptors in synoviocyte transformation, hyperplasia, and regulation of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) in models of inflammatory arthritis. METHODS NR4A messenger RNA levels in synovial tissue and primary synoviocytes were measured by quantitative reverse transcription-polymerase chain reaction (RT-PCR). NR4A2 was stably overexpressed in normal synoviocytes, and cell proliferation, survival, anchorage-independent growth, migration, and invasion were monitored in vitro. MMP and TIMP expression levels were analyzed by quantitative RT-PCR, and MMP-13 promoter activity was measured using reporter assays. Stable depletion of endogenous NR4A levels was achieved by lentiviral transduction of NR4A short hairpin RNA (shRNA), and the effects on proliferation, migration, and MMP-13 expression were analyzed. RESULTS NR4A2 was expressed at elevated levels in normal, OA, and RA synovial tissue and in primary RA synoviocytes. Tumor necrosis factor α (TNFα) rapidly and selectively induced expression of NR4A2 in synoviocytes. Ectopic expression of NR4A2 in normal synoviocytes significantly increased proliferation and survival, promoted anchorage-independent growth, and induced migration and invasion. MMP-13 gene expression was synergistically induced by NR4A2 and TNFα, while expression of TIMP-2 was antagonized. NR4A2 directly transactivated the proximal MMP-13 promoter, and a point mutation in the DNA binding domain of NR4A2 abolished transcriptional activation. Depletion of endogenous NR4A receptors with shRNA reduced synoviocyte proliferation, migration, and MMP-13 expression. CONCLUSION The orphan nuclear receptor NR4A2 is a downstream mediator of TNFα signaling in synovial tissue. NR4A2 transcriptional activity contributes to the hyperplastic and invasive phenotype of synoviocytes that leads to cartilage destruction, suggesting that this receptor may show promise as a therapeutic target in inflammatory arthritis.
Collapse
Affiliation(s)
- Kimberlee S Mix
- Loyola University New Orleans, Department of Biological Sciences, New Orleans, Louisiana 70118, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Mohan HM, Aherne CM, Rogers AC, Baird AW, Winter DC, Murphy EP. Molecular pathways: the role of NR4A orphan nuclear receptors in cancer. Clin Cancer Res 2012; 18:3223-8. [PMID: 22566377 DOI: 10.1158/1078-0432.ccr-11-2953] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nuclear receptors are of integral importance in carcinogenesis. Manipulation of classic ligand-activated nuclear receptors, such as estrogen receptor blockade in breast cancer, is an important established cancer therapy. Orphan nuclear receptors, such as nuclear family 4 subgroup A (NR4A) receptors, have no known natural ligand(s). These elusive receptors are increasingly recognized as molecular switches in cell survival and a molecular link between inflammation and cancer. NR4A receptors act as transcription factors, altering expression of downstream genes in apoptosis (Fas-ligand, TRAIL), proliferation, DNA repair, metabolism, cell migration, inflammation (interleukin-8), and angiogenesis (VEGF). NR4A receptors are modulated by multiple cell-signaling pathways, including protein kinase A/CREB, NF-κB, phosphoinositide 3-kinase/AKT, c-jun-NH(2)-kinase, Wnt, and mitogen-activated protein kinase pathways. NR4A receptor effects are context and tissue specific, influenced by their levels of expression, posttranslational modification, and interaction with other transcription factors (RXR, PPAR-Υ). The subcellular location of NR4A "nuclear receptors" is also important functionally; novel roles have been described in the cytoplasm where NR4A proteins act both indirectly and directly on the mitochondria to promote apoptosis via Bcl-2. NR4A receptors are implicated in a wide variety of malignancies, including breast, lung, colon, bladder, and prostate cancer; glioblastoma multiforme; sarcoma; and acute and/or chronic myeloid leukemia. NR4A receptors modulate response to conventional chemotherapy and represent an exciting frontier for chemotherapeutic intervention, as novel agents targeting NR4A receptors have now been developed. This review provides a concise clinical overview of current knowledge of NR4A signaling in cancer and the potential for therapeutic manipulation.
Collapse
Affiliation(s)
- Helen M Mohan
- UCD Veterinary Sciences Centre, University College Dublin, Department of Surgery, St Vincent's University Hospital, Elm Park, Dublin, Ireland.
| | | | | | | | | | | |
Collapse
|
38
|
Maijenburg MW, Gilissen C, Melief SM, Kleijer M, Weijer K, Ten Brinke A, Roelofs H, Van Tiel CM, Veltman JA, de Vries CJM, van der Schoot CE, Voermans C. Nuclear receptors Nur77 and Nurr1 modulate mesenchymal stromal cell migration. Stem Cells Dev 2011; 21:228-38. [PMID: 21480782 DOI: 10.1089/scd.2011.0076] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Detailed understanding of mesenchymal stromal cells (MSC) migration is imperative for future cellular therapies. To identify genes involved in the process of MSC migration, we generated gene expression profiles of migrating and nonmigrating fetal bone marrow MSC (FBMSC). Only 12 genes showed differential expression in migrating versus nonmigrating FBMSC. The nuclear receptors Nur77 and Nurr1 showed the highest expression in migratory MSC. Nur77 and Nurr1 are members of NR4A nuclear orphan receptor family, and we found that their expression is rapidly increased upon exposure of FBMSC to the migratory stimuli stromal-derived factor-1α (SDF-1α) and platelet-derived growth factor-BB. Lentiviral expression of Nur77 or Nurr1 resulted in enhanced migration of FBMSC toward SDF-1α compared with mock-transduced FBMSC. Analysis of the cell cycle, known to be involved in MSC migration, revealed that expression of Nur77 and Nurr1 decreases the proportion of cells in S-phase compared with control cells. Further, gain-of-function experiments showed increased hepatocyte growth factor expression and interleukin (IL)-6 and IL-8 production in MSC. Despite the altered cytokine profile, FBMSC expressing Nur77 or Nurr1 maintained the capacity to inhibit T-cell proliferation in a mixed lymphocyte reaction. Our results demonstrate that Nur77 and Nurr1 promote FBMSC migration. Modulation of Nur77 and Nurr1 activity may therefore offer perspectives to enhance the migratory potential of FBMSC which may specifically regulate the local immune response.
Collapse
Affiliation(s)
- Marijke W Maijenburg
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|