101
|
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
|
102
|
Requirement of novel amino acid fragments of orphan nuclear receptor TR3/Nur77 for its functions in angiogenesis. Oncotarget 2016; 6:24261-76. [PMID: 26155943 PMCID: PMC4695184 DOI: 10.18632/oncotarget.4637] [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: 04/30/2015] [Accepted: 06/05/2015] [Indexed: 01/08/2023] Open
Abstract
Pathological angiogenesis is a hallmark of many diseases. We demonstrated that TR3/Nur77 is an excellent target for pro-angiogenesis and anti-angiogenesis therapies. Here, we report that TR3 transcriptionally regulates endothelial cell migration, permeability and the formation of actin stress fibers that is independent of RhoA GTPase. 1) Amino acid residues 344-GRR-346 and de-phosphorylation of amino acid residue serine 351 in the DNA binding domain, and 2) phosphorylation of amino acid residues in the 41-61 amino acid fragment of the transactivation domain, of TR3 are required for its induction of the formation of actin stress fibers, cell proliferation, migration and permeability. The 41-61 amino acid fragment contains one of the three potential protein interaction motifs in the transactivation domain of TR3, predicted by computational modeling and analysis. These studies further our understanding of the molecular mechanism, by which TR3 regulates angiogenesis, identify novel therapeutic targeted sites of TR3, and set the foundation for the development of high-throughput screening assays to identify compounds targeting TR3/Nur77 for pro-angiogenesis and anti-angiogenesis therapies.
Collapse
|
103
|
Guo PD, Lu XX, Gan WJ, Li XM, He XS, Zhang S, Ji QH, Zhou F, Cao Y, Wang JR, Li JM, Wu H. RARγ Downregulation Contributes to Colorectal Tumorigenesis and Metastasis by Derepressing the Hippo-Yap Pathway. Cancer Res 2016; 76:3813-25. [PMID: 27325643 DOI: 10.1158/0008-5472.can-15-2882] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/15/2016] [Indexed: 11/16/2022]
Abstract
The Hippo-Yap pathway conveys oncogenic signals, but its regulation during cancer development is not well understood. Here, we identify the nuclear receptor RARγ as a regulator of the Hippo-Yap pathway in colorectal tumorigenesis and metastasis. RARγ is downregulated in human colorectal cancer tissues, where its expression correlates inversely with tumor size, TNM stage, and distant metastasis. Functional studies established that silencing of RARγ drove colorectal cancer cell growth, invasion, and metastatic properties both in vitro and in vivo Mechanistically, RARγ controlled Hippo-Yap signaling to inhibit colorectal cancer development, acting to promote phosphorylation and binding of Lats1 to its transcriptional coactivator Yap and thereby inactivating Yap target gene expression. In clinical specimens, RARγ expression correlated with overall survival outcomes and expression of critical Hippo-Yap pathway effector molecules in colorectal cancer patients. Collectively, our results defined RARγ as tumor suppressor in colorectal cancer that acts by restricting oncogenic signaling by the Hippo-Yap pathway, with potential implications for new approaches to colorectal cancer therapy. Cancer Res; 76(13); 3813-25. ©2016 AACR.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Cell Movement
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Hippo Signaling Pathway
- Humans
- Immunoenzyme Techniques
- Lymphatic Metastasis
- Male
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Staging
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- Prognosis
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Survival Rate
- Transcription Factors
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
- YAP-Signaling Proteins
- Retinoic Acid Receptor gamma
Collapse
Affiliation(s)
- Peng-Da Guo
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Xing-Xing Lu
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Wen-Juan Gan
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China. The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiu-Ming Li
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Xiao-Shun He
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China. The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shen Zhang
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Qing-Hua Ji
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Feng Zhou
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Yue Cao
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Jing-Ru Wang
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China
| | - Jian-Ming Li
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China.
| | - Hua Wu
- Pathology Center and Department of Pathology, Soochow University, Suzhou, China.
| |
Collapse
|
104
|
Roles of d-Amino Acids on the Bioactivity of Host Defense Peptides. Int J Mol Sci 2016; 17:ijms17071023. [PMID: 27376281 PMCID: PMC4964399 DOI: 10.3390/ijms17071023] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 12/15/2022] Open
Abstract
Host defense peptides (HDPs) are positively-charged and amphipathic components of the innate immune system that have demonstrated great potential to become the next generation of broad spectrum therapeutic agents effective against a vast array of pathogens and tumor. As such, many approaches have been taken to improve the therapeutic efficacy of HDPs. Amongst these methods, the incorporation of d-amino acids (d-AA) is an approach that has demonstrated consistent success in improving HDPs. Although, virtually all HDP review articles briefly mentioned about the role of d-AA, however it is rather surprising that no systematic review specifically dedicated to this topic exists. Given the impact that d-AA incorporation has on HDPs, this review aims to fill that void with a systematic discussion of the impact of d-AA on HDPs.
Collapse
|
105
|
Delgado E, Boisen MM, Laskey R, Chen R, Song C, Sallit J, Yochum ZA, Andersen CL, Sikora MJ, Wagner J, Safe S, Elishaev E, Lee A, Edwards RP, Haluska P, Tseng G, Schurdak M, Oesterreich S. High expression of orphan nuclear receptor NR4A1 in a subset of ovarian tumors with worse outcome. Gynecol Oncol 2016; 141:348-356. [PMID: 26946093 PMCID: PMC5154956 DOI: 10.1016/j.ygyno.2016.02.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 02/18/2016] [Accepted: 02/22/2016] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Nuclear receptors (NRs) play a vital role in the development and progression of several cancers including breast and prostate. Using TCGA data, we sought to identify critical nuclear receptors in high grade serous ovarian cancers (HGSOC) and to confirm these findings using in vitro approaches. METHODS In silico analysis of TCGA data was performed to identify relevant NRs in HGSOC. Ovarian cancer cell lines were screened for NR expression and functional studies were performed to determine the significance of these NRs in ovarian cancers. NR expression was analyzed in ovarian cancer tissue samples using immunohistochemistry to identify correlations with histology and stage of disease. RESULTS The NR4A family of NRs was identified as a potential driver of ovarian cancer pathogenesis. Overexpression of NR4A1 in particular correlated with worse progression free survival. Endogenous expression of NR4A1 in normal ovarian samples was relatively high compared to that of other tissue types, suggesting a unique role for this orphan receptor in the ovary. Expression of NR4A1 in HGSOC cell lines as well as in patient samples was variable. NR4A1 primarily localized to the nucleus in normal ovarian tissue while co-localization within the cytoplasm and nucleus was noted in ovarian cancer cell lines and patient tissues. CONCLUSIONS NR4A1 is highly expressed in a subset of HGSOC samples from patients that have a worse progression free survival. Studies to target NR4A1 for therapeutic intervention should include HGSOC.
Collapse
MESH Headings
- Animals
- Carcinoma, Ovarian Epithelial
- Cell Line, Tumor
- Female
- Genome
- Heterografts
- Humans
- Immunohistochemistry
- Mice
- Mice, SCID
- Neoplasms, Glandular and Epithelial/genetics
- Neoplasms, Glandular and Epithelial/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/biosynthesis
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Prognosis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
Collapse
Affiliation(s)
- Evan Delgado
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
| | - Michelle M Boisen
- Division of Gynecologic Oncology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| | - Robin Laskey
- Division of Gynecologic Oncology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Rui Chen
- Department of Biostatistics and Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chi Song
- Department of Biostatistics and Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Zachary A Yochum
- Department of Medicine, Division of Hematology Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Courtney L Andersen
- Department of Pharmacology and Chemical Biology, Womens Cancer Research Center, Magee-Womens Research Institute, and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA; Molecular Pharmacology Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Matthew J Sikora
- Department of Pharmacology and Chemical Biology, Womens Cancer Research Center, Magee-Womens Research Institute, and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Jacob Wagner
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Esther Elishaev
- Department of Pathology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Adrian Lee
- Department of Pharmacology and Chemical Biology, Womens Cancer Research Center, Magee-Womens Research Institute, and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Robert P Edwards
- Division of Gynecologic Oncology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Paul Haluska
- Department of Oncology and Pharmacology, Mayo Clinic, Rochester, MN, USA
| | - George Tseng
- Department of Biostatistics and Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark Schurdak
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, PA, USA
| | - Steffi Oesterreich
- Department of Pharmacology and Chemical Biology, Womens Cancer Research Center, Magee-Womens Research Institute, and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
106
|
Duong DT, Singh S, Bagheri M, Verma NK, Schmidtchen A, Malmsten M. Pronounced peptide selectivity for melanoma through tryptophan end-tagging. Sci Rep 2016; 6:24952. [PMID: 27117225 PMCID: PMC4847013 DOI: 10.1038/srep24952] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/08/2016] [Indexed: 12/18/2022] Open
Abstract
Effects of oligotryptophan end-tagging on the uptake of arginine-rich peptides into melanoma cells was investigated under various conditions and compared to that into non-malignant keratinocytes, fibroblasts, and erythrocytes, also monitoring resulting cell toxicity. In parallel, biophysical studies on peptide binding to, and destabilization of, model lipid membranes provided mechanistic insight into the origin of the selectivity between melanoma and non-malignant cells. Collectively, the results demonstrate that W-tagging represents a powerful way to increase selective peptide internalization in melanoma cells, resulting in toxicity against these, but not against the non-malignant cells. These effects were shown to be due to increased peptide adsorption to the outer membrane in melanoma cells, caused by the presence of anionic lipids such as phosphatidylserine and ganglioside GM1, and to peptide effects on mitochondria membranes and resulting apoptosis. In addition, the possibility of using W-tagged peptides for targeted uptake of nanoparticles/drug carriers in melanoma was demonstrated, as was the possibility to open up the outer membrane of melanoma cells in order to facilitate uptake of low Mw anticancer drugs, here demonstrated for doxorubicin.
Collapse
Affiliation(s)
- Dinh Thuy Duong
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232
| | - Shalini Singh
- Department of Pharmacy, Uppsala University, SE-75123, Uppsala, Sweden
| | - Mojtaba Bagheri
- Department of Pharmacy, Uppsala University, SE-75123, Uppsala, Sweden
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232
| | - Artur Schmidtchen
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, SE-221 84 Lund, Sweden
| | - Martin Malmsten
- Department of Pharmacy, Uppsala University, SE-75123, Uppsala, Sweden
| |
Collapse
|
107
|
Godoi PHC, Wilkie-Grantham RP, Hishiki A, Sano R, Matsuzawa Y, Yanagi H, Munte CE, Chen Y, Yao Y, Marassi FM, Kalbitzer HR, Matsuzawa SI, Reed JC. Orphan Nuclear Receptor NR4A1 Binds a Novel Protein Interaction Site on Anti-apoptotic B Cell Lymphoma Gene 2 Family Proteins. J Biol Chem 2016; 291:14072-14084. [PMID: 27129202 DOI: 10.1074/jbc.m116.715235] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Indexed: 11/06/2022] Open
Abstract
B cell lymphoma gene 2 (Bcl-2) family proteins are key regulators of programmed cell death and important targets for drug discovery. Pro-apoptotic and anti-apoptotic Bcl-2 family proteins reciprocally modulate their activities in large part through protein interactions involving a motif known as BH3 (Bcl-2 homology 3). Nur77 is an orphan member of the nuclear receptor family that lacks a BH3 domain but nevertheless binds certain anti-apoptotic Bcl-2 family proteins (Bcl-2, Bfl-1, and Bcl-B), modulating their effects on apoptosis and autophagy. We used a combination of NMR spectroscopy-based methods, mutagenesis, and functional studies to define the interaction site of a Nur77 peptide on anti-apoptotic Bcl-2 family proteins and reveal a novel interaction surface. Nur77 binds adjacent to the BH3 peptide-binding crevice, suggesting the possibility of cross-talk between these discrete binding sites. Mutagenesis of residues lining the identified interaction site on Bcl-B negated the interaction with Nur77 protein in cells and prevented Nur77-mediated modulation of apoptosis and autophagy. The findings establish a new protein interaction site with the potential to modulate the apoptosis and autophagy mechanisms governed by Bcl-2 family proteins.
Collapse
Affiliation(s)
- Paulo H C Godoi
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037
| | | | - Asami Hishiki
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Renata Sano
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Yasuko Matsuzawa
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Hiroko Yanagi
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Claudia E Munte
- Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Ya Chen
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Yong Yao
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Francesca M Marassi
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Hans R Kalbitzer
- Institute of Biophysics and Physical Biochemistry, University of Regensburg, Universitätsstr. 31, 93040 Regensburg, Germany
| | - Shu-Ichi Matsuzawa
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037,.
| | - John C Reed
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California 92037,; Roche, Pharma Research and Early Development, Basel 4070, Switzerland.
| |
Collapse
|
108
|
Toxoplasma gondii ROP18: potential to manipulate host cell mitochondrial apoptosis. Parasitol Res 2016; 115:2415-22. [PMID: 27021182 DOI: 10.1007/s00436-016-4993-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/09/2016] [Indexed: 10/22/2022]
Abstract
Toxoplasma gondii is an obligate intracellular parasite that may manipulate host cell mitochondrial apoptosis pathways. In our experiment, 293T cells were transfected with the p3×FLAG-CMV-Myc-ROP18 vector and expressed the ROP18-Myc fusion protein. Cell apoptosis was induced by 0.5 μg/mL actinomycin D (ActD) and was detected by Annexin V-FITC/PI assay. The cell mitochondrial membrane potential was determined by JC-1. Cytochrome c (Cyto-c) from mitochondria and the cytoplasm was measured by Western blot. The Bcl-2 and Bax coding gene expression levels were detected by real-time PCR. We found, in vitro, that T. gondii ROP18 significantly suppressed 293T cell apoptosis induced by ActD and maintained mitochondrial membrane potential and integrity, thereby preventing the release of Cyto-c from mitochondria into the cytoplasm. The ratio of Bcl-2/Bax in ROP18-overexpressing cells was significantly higher than that of the negative control. Therefore, we speculate that ROP18 could suppress host cell apoptosis via the mitochondrial apoptosis pathway in vitro.
Collapse
|
109
|
Safe S, Jin UH, Morpurgo B, Abudayyeh A, Singh M, Tjalkens RB. Nuclear receptor 4A (NR4A) family - orphans no more. J Steroid Biochem Mol Biol 2016; 157:48-60. [PMID: 25917081 PMCID: PMC4618773 DOI: 10.1016/j.jsbmb.2015.04.016] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 03/26/2015] [Accepted: 04/21/2015] [Indexed: 01/17/2023]
Abstract
The orphan nuclear receptors NR4A1, NR4A2 and NR4A3 are immediate early genes induced by multiple stressors, and the NR4A receptors play an important role in maintaining cellular homeostasis and disease. There is increasing evidence for the role of these receptors in metabolic, cardiovascular and neurological functions and also in inflammation and inflammatory diseases and in immune functions and cancer. Despite the similarities of NR4A1, NR4A2 and NR4A3 and their interactions with common cis-genomic elements, they exhibit unique activities and cell-/tissue-specific functions. Although endogenous ligands for NR4A receptors have not been identified, there is increasing evidence that structurally-diverse synthetic molecules can directly interact with the ligand binding domain of NR4A1 and act as agonists or antagonists, and ligands for NR4A2 and NR4A3 have also been identified. Since NR4A receptors are key factors in multiple diseases, there are opportunities for the future development of NR4A ligands for clinical applications in treating multiple health problems including metabolic, neurologic and cardiovascular diseases, other inflammatory conditions, and cancer.
Collapse
MESH Headings
- Arthritis/metabolism
- Cardiovascular Diseases/metabolism
- DNA-Binding Proteins/metabolism
- Homeostasis
- Humans
- Immunity, Cellular
- Inflammation/metabolism
- Ligands
- Metabolic Diseases/genetics
- Metabolic Diseases/metabolism
- Neoplasms/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Receptors, Steroid/metabolism
- Receptors, Thyroid Hormone/metabolism
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA.
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Benjamin Morpurgo
- Texas A&M Institute for Genomic Medicine, Texas A&M University, 670 Raymond Stotzer Pkwy, College Station, TX 77843, USA
| | - Ala Abudayyeh
- Department of General Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mandip Singh
- Department of Pharmaceutics, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Ronald B Tjalkens
- Department of Toxicology and Neuroscience, Colorado State University, 1680Campus Delivery, Fort Collins, CO 80523-1680, USA
| |
Collapse
|
110
|
Zhang X, Zhou H, Su Y. Targeting truncated RXRα for cancer therapy. Acta Biochim Biophys Sin (Shanghai) 2016; 48:49-59. [PMID: 26494413 DOI: 10.1093/abbs/gmv104] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/24/2015] [Indexed: 01/08/2023] Open
Abstract
Retinoid X receptor-alpha (RXRα), a unique member of the nuclear receptor superfamily, is a well-established drug target, representing one of the most important targets for pharmacologic interventions and therapeutic applications for cancer. However, how RXRα regulates cancer cell growth and how RXRα modulators suppress tumorigenesis are poorly understood. Altered expression and aberrant function of RXRα are implicated in the development of cancer. Previously, several studies had demonstrated the presence of N-terminally truncated RXRα (tRXRα) proteins resulted from limited proteolysis of RXRα in tumor cells. Recently, we discovered that overexpression of tRXRα can promote tumor growth by interacting with tumor necrosis factor-alpha-induced phosphoinositide 3-kinase and NF-κB signal transduction pathways. We also identified nonsteroidal anti-inflammatory drug Sulindac and analogs as effective inhibitors of tRXRα activities via a unique binding mechanism. This review discusses the emerging roles of tRXRα and modulators in the regulation of cancer cell survival and death as well as inflammation and our recent understanding of tRXRα regulation by targeting the alternate binding sites on its surface.
Collapse
Affiliation(s)
- Xiaokun Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China Sanford Burnham Prebys Medical Discovery Institute, Cancer Center, La Jolla, CA 92037, USA
| | - Hu Zhou
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Ying Su
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China Sanford Burnham Prebys Medical Discovery Institute, Cancer Center, La Jolla, CA 92037, USA
| |
Collapse
|
111
|
Cui M, Cai Z, Chu S, Sun Z, Wang X, Hu L, Yi J, Shen L, He B. Orphan Nuclear Receptor Nur77 Inhibits Angiotensin II–Induced Vascular Remodeling via Downregulation of β-Catenin. Hypertension 2016; 67:153-62. [DOI: 10.1161/hypertensionaha.115.06114] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Mingli Cui
- From the Department of Cardiology, Renji Hospital (M.C., Z.C., S.C., Z.S., X.W., L.H., L.S., B.H.) and Department of Cell Biology, Key Laboratory of the Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciences (J.Y.), School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhaohua Cai
- From the Department of Cardiology, Renji Hospital (M.C., Z.C., S.C., Z.S., X.W., L.H., L.S., B.H.) and Department of Cell Biology, Key Laboratory of the Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciences (J.Y.), School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shichun Chu
- From the Department of Cardiology, Renji Hospital (M.C., Z.C., S.C., Z.S., X.W., L.H., L.S., B.H.) and Department of Cell Biology, Key Laboratory of the Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciences (J.Y.), School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhe Sun
- From the Department of Cardiology, Renji Hospital (M.C., Z.C., S.C., Z.S., X.W., L.H., L.S., B.H.) and Department of Cell Biology, Key Laboratory of the Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciences (J.Y.), School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaolei Wang
- From the Department of Cardiology, Renji Hospital (M.C., Z.C., S.C., Z.S., X.W., L.H., L.S., B.H.) and Department of Cell Biology, Key Laboratory of the Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciences (J.Y.), School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Liuhua Hu
- From the Department of Cardiology, Renji Hospital (M.C., Z.C., S.C., Z.S., X.W., L.H., L.S., B.H.) and Department of Cell Biology, Key Laboratory of the Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciences (J.Y.), School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jing Yi
- From the Department of Cardiology, Renji Hospital (M.C., Z.C., S.C., Z.S., X.W., L.H., L.S., B.H.) and Department of Cell Biology, Key Laboratory of the Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciences (J.Y.), School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Linghong Shen
- From the Department of Cardiology, Renji Hospital (M.C., Z.C., S.C., Z.S., X.W., L.H., L.S., B.H.) and Department of Cell Biology, Key Laboratory of the Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciences (J.Y.), School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ben He
- From the Department of Cardiology, Renji Hospital (M.C., Z.C., S.C., Z.S., X.W., L.H., L.S., B.H.) and Department of Cell Biology, Key Laboratory of the Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciences (J.Y.), School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
112
|
Xie L, Yang R, Liu S, Lyle S, Cotsarelis G, Xiang L, Zhang L, Li B, Wan M, Xu X. TR3 is preferentially expressed by bulge epithelial stem cells in human hair follicles. J Transl Med 2016; 96:81-8. [PMID: 26707825 PMCID: PMC4915568 DOI: 10.1038/labinvest.2015.125] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 09/23/2015] [Accepted: 09/24/2015] [Indexed: 12/12/2022] Open
Abstract
TR3 is an orphan member of the steroid/thyroid/retinoid nuclear receptor superfamily of transcription factors and it plays a pivotal role in regulating cell growth and apoptosis. The expression and function of TR3 in skin have not been well investigated. Using a cDNA expression assay, we discover that TR3 is significantly enriched in human telogen bulge compared with anagen bulb. Immunohistochemical staining confirms that TR3 is highly expressed in the bulge region of human hair follicles and it colocalizes with cytokeratin 15 (K15), an epithelial stem cell marker. To study the function of TR3 in the effect of androgens in keratinocytes, we treat HaCaT keratinocytes and primary human keratinocytes with dihydrotestosterone (DHT) and testosterone (T). The treated keratinocytes show a dose-dependent growth reduction to DHT and T. DHT increases the expression of TR3 in keratinocytes, associated with a concomitant increase of BAD and decrease of Bcl-2 expression. Knockdown TR3 expression by siRNA blocks the inhibitory effect of DHT on keratinocyte proliferation. Our results demonstrate that TR3 is localized to the stem cell compartment in the human hair follicles. Androgen increases TR3 expression in cultured keratinocytes. Our data suggest that TR3 mediates at least part of the inhibitory effect of androgens on keratinocytes.
Collapse
Affiliation(s)
- Lin Xie
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA,Department of Dermatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China,Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ruifeng Yang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Shujing Liu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Stephen Lyle
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - George Cotsarelis
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Leihong Xiang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Litao Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Bin Li
- Department of Dermatology, Yueyang Hospital, Shanghai, China
| | - Miaojian Wan
- Department of Dermatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
113
|
Sinoporphyrin sodium mediated photodynamic therapy inhibits the migration associated with collapse of F-actin filaments cytoskeleton in MDA-MB-231 cells. Photodiagnosis Photodyn Ther 2015; 13:58-65. [PMID: 26742781 DOI: 10.1016/j.pdpdt.2015.12.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 11/25/2015] [Accepted: 12/22/2015] [Indexed: 01/10/2023]
Abstract
OBJECTIVE We previously demonstrated that the photosensitizer sinoporphyrin sodium (DVDMS) mediated photodynamic therapy (PDT) had potential advantages in inhibiting tumor growth and metastasis. However, details regarding the mechanism of cell migration inhibition remain unclear. Therefore, in this study, we aimed to investigate the effects of DVDMS-PDT on F-actin filaments, cell migration, apoptotic response and the possible interactions between them in human breast cancer MDA-MB-231 cells. MATERIALS AND METHODS The cell viability was evaluated by MTT and Guava ViaCount assays. The subcellular localization of DVDMS and reactive oxygen species (ROS) generation were analyzed by fluorescence microscope and flow cytometry. FITC-phalloidin was used to evaluate the changes of F-actin filaments. Cell migration was analyzed by scratch assay and Transwell assay. Cell apoptosis was determined by nuclear TUNEL staining and Annexin V-PE/7-AAD assay. Jasplakinolide, an F-actin stabilizer, was applied to dissect the influences of F-actin filaments disruption on cell migration and apoptosis. RESULTS DVDMS-PDT significantly suppressed cell proliferation, promoted early apoptotic response, triggered collapse of F-actin filaments and inhibited cell migration in MDA-MB-231 cells. Cell migration significantly increased when cells were pretreated with F-actin stabilizer jasplakinolide after PDT, while cell apoptosis was not obviously affected. Moreover, ROS was a key factor in causing collapse of F-actin filaments. CONCLUSION We demonstrated that DVDMS-PDT triggered cell apoptosis and collapse of F-actin filaments through the induction of ROS in MDA-MB-231 cells. F-actin filaments contributed to cell migration but produced no obvious effect on cell apoptosis.
Collapse
|
114
|
Wu H, Li XM, Wang JR, Gan WJ, Jiang FQ, Liu Y, Zhang XD, He XS, Zhao YY, Lu XX, Guo YB, Zhang XK, Li JM. NUR77 exerts a protective effect against inflammatory bowel disease by negatively regulating the TRAF6/TLR-IL-1R signalling axis. J Pathol 2015; 238:457-69. [PMID: 26564988 DOI: 10.1002/path.4670] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 11/02/2015] [Accepted: 11/06/2015] [Indexed: 01/05/2023]
Abstract
Nur77, an immediate-early response gene, participates in a wide range of biological functions. Its human homologue, NUR77, is known by several names and has the HGNC-approved gene symbol NR4A1. However, the role of Nur77 in inflammatory bowel disease (IBD) and its underlying mechanisms remain elusive. Here, using public data from the International Inflammatory Bowel Disease Genetics Consortium (IIBDGC) on the most recent genome-wide association studies (GWAS) for ulcerative colitis (UC) and Crohn's disease (CD), we found that genetic variants of the NUR77 gene are associated with increased risk for both UC and CD. Accordingly, Nur77 expression was significantly reduced in colon tissues from patients with UC or CD and mice treated with DSS. Nur77 deficiency increased the susceptibility of mice to DSS-induced experimental colitis and prevented intestinal recovery, whereas treatment with cytosporone B (Csn-B), an agonist for Nur77, significantly attenuated excessive inflammatory response in the DSS-induced colitis mouse model. Mechanistically, NUR77 acts as a negative regulator of TLR-IL-1R signalling by interacting with TRAF6. This interaction prevented auto-ubiquitination and oligomerization of TRAF6 and subsequently inhibited NF-κB activation and pro-inflammatory cytokine production. Taken together, our GWAS-based analysis and in vitro and in vivo studies have demonstrated that Nur77 is an important regulator of TRAF6/TLR-IL-1R-initiated inflammatory signalling, and loss of Nur77 may contribute to the development of IBD, suggesting Nur77 as a potential target for the prevention and treatment of IBD.
Collapse
Affiliation(s)
- Hua Wu
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Xiu-Ming Li
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Jing-Ru Wang
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Wen-Juan Gan
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China.,First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fu-Quan Jiang
- School of Pharmaceutical Sciences, Xiamen University, People's Republic of China
| | - Yao Liu
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Xin-Dao Zhang
- School of Pharmaceutical Sciences, Xiamen University, People's Republic of China
| | - Xiao-Shun He
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Yuan-Yuan Zhao
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Xing-Xing Lu
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| | - Yan-Bing Guo
- School of Pharmaceutical Sciences, Xiamen University, People's Republic of China.,Sanford-Burnham Medical Research Institute, Cancer Center, La Jolla, CA, USA
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Xiamen University, People's Republic of China.,Sanford-Burnham Medical Research Institute, Cancer Center, La Jolla, CA, USA
| | - Jian-Ming Li
- Pathology Centre and Department of Pathology, Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
115
|
Xie L, Jiang F, Zhang X, Alitongbieke G, Shi X, Meng M, Xu Y, Ren A, Wang J, Cai L, Zhou Y, Xu Y, Su Y, Liu J, Zeng Z, Wang G, Zhou H, Chen QC, Zhang XK. Honokiol sensitizes breast cancer cells to TNF-α induction of apoptosis by inhibiting Nur77 expression. Br J Pharmacol 2015; 173:344-56. [PMID: 26505879 DOI: 10.1111/bph.13375] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 10/01/2015] [Accepted: 10/13/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE The orphan nuclear receptor Nur77 is implicated in the survival and apoptosis of cancer cells. The purpose of this study was to determine whether and how Nur77 serves to mediate the effect of the inflammatory cytokine TNF-α in cancer cells and to identify and characterize new agents targeting Nur77 for cancer therapy. EXPERIMENTAL APPROACH The effects of TNF-α on the expression and function of Nur77 were studied using in vitro and in vivo models. Nur77 expression was evaluated in tumour tissues from breast cancer patients. The anticancer effects of honokiol and its mechanism of action were assessed by in vitro, cell-based and animal studies. KEY RESULTS TNF-α rapidly and potently induced the expression of Nur77 in breast cancer cells through activation of IκB kinase and JNK. Knocking down Nur77 resulted in TNF-α-dependent apoptosis, while ectopic Nur77 expression in MCF-7 cells promoted their growth in animals. Levels of Nur77 were higher in tumour tissues than the corresponding tissues surrounding the tumour in about 50% breast cancer patients studied. Our in vitro and animal studies also identified honokiol as an effective sensitizer of TNF-α-induced apoptosis by inhibiting TNF-α-induced Nur77 mRNA expression, which could be attributed to its interference of TNFR1's interaction with receptor-interacting protein 1 (RIPK1). CONCLUSIONS AND IMPLICATIONS TNF-α-induced Nur77 serves as a survival factor to attenuate the death effect of TNF-α in cancer cells. With its proven human safety profile, honokiol represents a promising agent that warrants further clinical development.
Collapse
Affiliation(s)
- Lei Xie
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Fuquan Jiang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Xindao Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | | | - Xinlei Shi
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - MinJun Meng
- Zhongshan Hospital, Xiamen University, Xiamen, 361102, China
| | - Yiming Xu
- Zhongshan Hospital, Xiamen University, Xiamen, 361102, China
| | - Anshi Ren
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Jing Wang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Lijun Cai
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Yunxia Zhou
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Yang Xu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Ying Su
- Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, 92037, USA
| | - Jie Liu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Guanghui Wang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Hu Zhou
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Quan Cheng Chen
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China.,Sanford Burnham Prebys Medical Discovery Institute Cancer Center, La Jolla, CA, 92037, USA
| |
Collapse
|
116
|
Prokhorova EA, Zamaraev AV, Kopeina GS, Zhivotovsky B, Lavrik IN. Role of the nucleus in apoptosis: signaling and execution. Cell Mol Life Sci 2015; 72:4593-612. [PMID: 26346492 PMCID: PMC11113907 DOI: 10.1007/s00018-015-2031-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/06/2015] [Accepted: 08/27/2015] [Indexed: 02/06/2023]
Abstract
Since their establishment in the early 1970s, the nuclear changes upon apoptosis induction, such as the condensation of chromatin, disassembly of nuclear scaffold proteins and degradation of DNA, were, and still are, considered as the essential steps and hallmarks of apoptosis. These are the characteristics of the execution phase of apoptotic cell death. In addition, accumulating data clearly show that some nuclear events can lead to the induction of apoptosis. In particular, if DNA lesions resulting from deregulation during the cell cycle or DNA damage induced by chemotherapeutic drugs or viral infection cannot be efficiently eliminated, apoptotic mechanisms, which enable cellular transformation to be avoided, are activated in the nucleus. The functional heterogeneity of the nuclear organization allows the tight regulation of these signaling events that involve the movement of various nuclear proteins to other intracellular compartments (and vice versa) to initiate and govern apoptosis. Here, we discuss how these events are coordinated to execute apoptotic cell death.
Collapse
Affiliation(s)
- Evgeniia A Prokhorova
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Alexey V Zamaraev
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Gelina S Kopeina
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Boris Zhivotovsky
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia.
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden.
| | - Inna N Lavrik
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia
- Department of Translational Inflammation, Institute of Experimental Internal Medicine, Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
117
|
Agostini-Dreyer A, Jetzt AE, Stires H, Cohick WS. Endogenous IGFBP-3 Mediates Intrinsic Apoptosis Through Modulation of Nur77 Phosphorylation and Nuclear Export. Endocrinology 2015; 156:4141-51. [PMID: 26340041 DOI: 10.1210/en.2015-1215] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In nontransformed bovine mammary epithelial cells, the intrinsic apoptosis inducer anisomycin (ANS) induces IGFBP-3 expression and nuclear localization and knockdown of IGFBP-3 attenuates ANS-induced apoptosis. Others have shown in prostate cancer cells that exogenous IGFBP-3 induces apoptosis by facilitating nuclear export of the orphan nuclear receptor Nur77 and its binding partner, retinoid X receptor-α (RXRα). The goal of the present work was to determine whether endogenous IGFBP-3 plays a role in ANS-induced apoptosis by facilitating nuclear transport of Nur77 and/or RXRα in nontransformed cells. Knockdown of Nur77 with siRNA decreased ANS-induced cleavage of caspase-3 and -7 and their downstream target, PARP, indicating a role for Nur77 in ANS-induced apoptosis. In cells transfected with IGFBP-3, IGFBP-3 associated with RXRα but not Nur77 under basal conditions, however, IGFBP-3 co-precipitated with phosphorylated forms of both proteins in ANS-treated cells. Indirect immunofluorescence and cell fractionation techniques showed that ANS induced phosphorylation and transport of Nur77 from the nucleus to the cytoplasm and these effects were attenuated by knockdown of IGFBP-3. These data suggest that endogenous IGFBP-3 plays a role in intrinsic apoptosis by facilitating phosphorylation and nuclear export of Nur77 to the cytoplasm where it exerts its apoptotic effect. Whether this mechanism involves a physical association between endogenous IGFBP-3 and Nur77 or RXRα remains to be determined.
Collapse
Affiliation(s)
- Allyson Agostini-Dreyer
- Graduate Program in Nutritional Sciences (A.A.-D., W.S.C.), Department of Animal Sciences, Rutgers (A.E.J., W.S.C.), and Graduate Program in Endocrinology and Animal Biosciences (H.S., W.S.C.), Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901-8520
| | - Amanda E Jetzt
- Graduate Program in Nutritional Sciences (A.A.-D., W.S.C.), Department of Animal Sciences, Rutgers (A.E.J., W.S.C.), and Graduate Program in Endocrinology and Animal Biosciences (H.S., W.S.C.), Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901-8520
| | - Hillary Stires
- Graduate Program in Nutritional Sciences (A.A.-D., W.S.C.), Department of Animal Sciences, Rutgers (A.E.J., W.S.C.), and Graduate Program in Endocrinology and Animal Biosciences (H.S., W.S.C.), Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901-8520
| | - Wendie S Cohick
- Graduate Program in Nutritional Sciences (A.A.-D., W.S.C.), Department of Animal Sciences, Rutgers (A.E.J., W.S.C.), and Graduate Program in Endocrinology and Animal Biosciences (H.S., W.S.C.), Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901-8520
| |
Collapse
|
118
|
Hedrick E, Lee SO, Doddapaneni R, Singh M, Safe S. Nuclear receptor 4A1 as a drug target for breast cancer chemotherapy. Endocr Relat Cancer 2015; 22:831-840. [PMID: 26229035 DOI: 10.1530/erc-15-0063] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/30/2015] [Indexed: 12/26/2022]
Abstract
The orphan nuclear receptor 4A1 (NR4A1) is overexpressed in mammary tumors and breast cancer cell lines. The functional activity of this receptor was investigated by RNA interference with oligonucleotides targeted to NR4A1 (siNR4A1) and by treatment with NR4A1 antagonists. Breast cancer cells were treated with NR4A1 antagonists or transfected with siNR4A. Effects on cell proliferation and apoptosis as well as specific genes associated with these responses were investigated in MCF-7, SKBR3, and MDA-MB-231 cells, and in athymic nude mice bearing MDA-MB-231 cells as xenografts. Transfection of MCF-7, MDA-MB-231, and SKBR3 breast cancer cells with siNR4A1 decreased cell proliferation and induced apoptosis in these cell lines. Transfection of breast cancer cells with siNR4A1 also decreased expression of Sp-regulated genes including survivin, bcl-2, and epidermal growth factor receptor, inhibited mTOR signaling in MCF-7 cells that express WT p53, and activated oxidative and endoplasmic reticulum stress through downregulation of thioredoxin domain-containing 5 and isocitrate dehydrogenase 1. 1,1-Bis(3'-indolyl)-1-(p-substituted phenyl)methanes (C-DIMs) are NR4A1 ligands that act as NR4A1 antagonists. Treatment with selected analogs also inhibited breast cancer cell and tumor growth and induced apoptosis. The effects of C-DIM/NR4A1 antagonists were comparable to those observed after NR4A1 knockdown. Results with siNR4A1 or C-DIMs/NR4A1 antagonists in breast cancer cells and tumors were similar to those previously reported in pancreatic, lung, and colon cancer cells. They demonstrate the potential clinical applications of NR4A1 antagonists in patients with tumors that overexpress this receptor.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Blotting, Western
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Proliferation/drug effects
- Endoplasmic Reticulum Stress/drug effects
- Female
- Humans
- Indoles/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Phenols/pharmacology
- RNA, Small Interfering/genetics
- Reactive Oxygen Species/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Erik Hedrick
- Department of Veterinary Physiology and Pharmacology Texas A&M University, 4466 TAMU, College Station, Texas 77843-4466, USA Department of Food Science and Technology Keimyung University, Daegu 704701, Republic of Korea Department of Pharmaceutics College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Syng-Ook Lee
- Department of Veterinary Physiology and Pharmacology Texas A&M University, 4466 TAMU, College Station, Texas 77843-4466, USA Department of Food Science and Technology Keimyung University, Daegu 704701, Republic of Korea Department of Pharmaceutics College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Ravi Doddapaneni
- Department of Veterinary Physiology and Pharmacology Texas A&M University, 4466 TAMU, College Station, Texas 77843-4466, USA Department of Food Science and Technology Keimyung University, Daegu 704701, Republic of Korea Department of Pharmaceutics College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Mandip Singh
- Department of Veterinary Physiology and Pharmacology Texas A&M University, 4466 TAMU, College Station, Texas 77843-4466, USA Department of Food Science and Technology Keimyung University, Daegu 704701, Republic of Korea Department of Pharmaceutics College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology Texas A&M University, 4466 TAMU, College Station, Texas 77843-4466, USA Department of Food Science and Technology Keimyung University, Daegu 704701, Republic of Korea Department of Pharmaceutics College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| |
Collapse
|
119
|
Land RH, Rayne AK, Vanderbeck AN, Barlowe TS, Manjunath S, Gross M, Eiger S, Klein PS, Cunningham NR, Huang J, Emerson SG, Punt JA. The orphan nuclear receptor NR4A1 specifies a distinct subpopulation of quiescent myeloid-biased long-term HSCs. Stem Cells 2015; 33:278-88. [PMID: 25284014 DOI: 10.1002/stem.1852] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/29/2014] [Indexed: 01/09/2023]
Abstract
Hematopoiesis is maintained throughout life by self-renewing hematopoietic stem cells (HSCs) that differentiate to produce both myeloid and lymphoid cells. The NR4A family of orphan nuclear receptors, which regulates cell fate in many tissues, appears to play a key role in HSC proliferation and differentiation. Using a NR4A1(GFP) BAC transgenic reporter mouse we have investigated NR4A1 expression and its regulation in early hematopoiesis. We show that NR4A1 is most highly expressed in a subset of Lin(-) Sca-1(+) c-Kit(+) CD48(-) CD150(+) long-term (LT) HSCs, and its expression is tightly associated with HSC quiescence. We also show that NR4A1 expression in HSCs is induced by PGE2, a known enhancer of stem cell engraftment potential. Finally, we find that both NR4A1(GFP+) and NR4A1(GFP-) HSCs successfully engraft primary and secondary irradiated hosts; however, NR4A1(GFP+) HSCs are distinctly myeloid-biased. These results show that NR4A1 expression identifies a highly quiescent and distinct population of myeloid-biased LT-HSCs.
Collapse
Affiliation(s)
- Ruben H Land
- Department of Microbiology and Immunology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Nur77 is involved in graft infiltrating T lymphocyte apoptosis in rat cardiac transplantation model. Pathol Res Pract 2015; 211:633-40. [DOI: 10.1016/j.prp.2015.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 04/05/2015] [Accepted: 04/17/2015] [Indexed: 12/12/2022]
|
121
|
Wang WJ, Wang Y, Hou PP, Li FW, Zhou B, Chen HZ, Bian XL, Cai QX, Xing YZ, He JP, Zhang H, Huang PQ, Lin T, Wu Q. Induction of Autophagic Death in Cancer Cells by Agonizing TR3 and Attenuating Akt2 Activity. CHEMISTRY & BIOLOGY 2015; 22:1040-1051. [PMID: 26235054 DOI: 10.1016/j.chembiol.2015.06.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 05/27/2015] [Accepted: 06/15/2015] [Indexed: 11/16/2022]
Abstract
Apoptotic resistance is becoming a significant obstacle for cancer therapy as the majority of treatment takes the route of apoptotic induction. It is of great importance to develop an alternative strategy to induce cancer cell death. We previously reported that autophagic cell death mediated by nuclear receptor TR3 and driven by a chemical agonist, 1-(3,4,5-trihydroxyphenyl)nonan-1-one (THPN), is highly effective in the therapy of melanoma but not any other cancer types. Here, we discovered that the insensitivity of cancer cells to THPN originated from a high cellular Akt2 activity. Akt2 phosphorylation interferes with TR3 export to cytoplasm and targeting to mitochondria, which lead to the autophagic induction. Therefore, the TR3-mediated autophagy could be effectively induced in the otherwise insensitive cells by downregulating Akt2 activity. Highly effective antineoplastic compounds are developed through optimizing the structure of THPN. This study implicates a general strategy for cancer therapy by the induction of autophagic cell death.
Collapse
Affiliation(s)
- Wei-jia Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Yuan Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Pei-pei Hou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Feng-wei Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Bo Zhou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Hang-zi Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Xue-li Bian
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Qi-xu Cai
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Yong-zhen Xing
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Jian-ping He
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China
| | - Hongkui Zhang
- Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, Fujian Province, P.R. China
| | - Pei-qiang Huang
- Key Laboratory for Chemical Biology of Fujian Province, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, Fujian Province, P.R. China
| | - Tianwei Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China.
| | - Qiao Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, State-Province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian Province, P.R. China.
| |
Collapse
|
122
|
Chen F, Chen J, Lin J, Cheltsov AV, Xu L, Chen Y, Zeng Z, Chen L, Huang M, Hu M, Ye X, Zhou Y, Wang G, Su Y, Zhang L, Zhou F, Zhang XK, Zhou H. NSC-640358 acts as RXRα ligand to promote TNFα-mediated apoptosis of cancer cell. Protein Cell 2015; 6:654-666. [PMID: 26156677 PMCID: PMC4537469 DOI: 10.1007/s13238-015-0178-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 05/03/2015] [Indexed: 12/18/2022] Open
Abstract
Retinoid X receptor α (RXRα) and its N-terminally truncated version tRXRα play important roles in tumorigenesis, while some RXRα ligands possess potent anti-cancer activities by targeting and modulating the tumorigenic effects of RXRα and tRXRα. Here we describe NSC-640358 (N-6), a thiazolyl-pyrazole derived compound, acts as a selective RXRα ligand to promote TNFα-mediated apoptosis of cancer cell. N-6 binds to RXRα and inhibits the transactivation of RXRα homodimer and RXRα/TR3 heterodimer. Using mutational analysis and computational study, we determine that Arg316 in RXRα, essential for 9-cis-retinoic acid binding and activating RXRα transactivation, is not required for antagonist effects of N-6, whereas Trp305 and Phe313 are crucial for N-6 binding to RXRα by forming extra π–π stacking interactions with N-6, indicating a distinct RXRα binding mode of N-6. N-6 inhibits TR3-stimulated transactivation of Gal4-DBD-RXRα-LBD by binding to the ligand binding pocket of RXRα-LBD, suggesting a strategy to regulate TR3 activity indirectly by using small molecules to target its interacting partner RXRα. For its physiological activities, we show that N-6 strongly inhibits tumor necrosis factor α (TNFα)-induced AKT activation and stimulates TNFα-mediated apoptosis in cancer cells in an RXRα/tRXRα dependent manner. The inhibition of TNFα-induced tRXRα/p85α complex formation by N-6 implies that N-6 targets tRXRα to inhibit TNFα-induced AKT activation and to induce cancer cell apoptosis. Together, our data illustrate a new RXRα ligand with a unique RXRα binding mode and the abilities to regulate TR3 activity indirectly and to induce TNFα-mediated cancer cell apoptosis by targeting RXRα/tRXRα.
Collapse
Affiliation(s)
- Fan Chen
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
- />School of Biological Science and Biotechnology, Minnan Normal University, Zhangzhou, 363000 China
| | - Jiebo Chen
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
| | - Jiacheng Lin
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
| | | | - Lin Xu
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
| | - Ya Chen
- />Cancer Center, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037 USA
| | - Zhiping Zeng
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
| | - Liqun Chen
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
| | - Mingfeng Huang
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
| | - Mengjie Hu
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
| | - Xiaohong Ye
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
| | - Yuqi Zhou
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
| | - Guanghui Wang
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
| | - Ying Su
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
- />Cancer Center, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037 USA
| | - Long Zhang
- />Life Science Institute, Zhejiang University, Hangzhou, 310058 China
| | - Fangfang Zhou
- />Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123 China
| | - Xiao-kun Zhang
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
- />Cancer Center, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037 USA
| | - Hu Zhou
- />School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102 China
| |
Collapse
|
123
|
Yu C, Cui S, Zong C, Gao W, Xu T, Gao P, Chen J, Qin D, Guan Q, Liu Y, Fu Y, Li X, Wang X. The Orphan Nuclear Receptor NR4A1 Protects Pancreatic β-Cells from Endoplasmic Reticulum (ER) Stress-mediated Apoptosis. J Biol Chem 2015; 290:20687-20699. [PMID: 26157144 PMCID: PMC4543630 DOI: 10.1074/jbc.m115.654863] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Indexed: 11/06/2022] Open
Abstract
The role of NR4A1 in apoptosis is controversial. Pancreatic β-cells often face endoplasmic reticulum (ER) stress under adverse conditions such as high free fatty acid (FFA) concentrations and sustained hyperglycemia. Severe ER stress results in β-cell apoptosis. The aim of this study was to analyze the role of NR4A1 in ER stress-mediated β-cell apoptosis and to characterize the related mechanisms. We confirmed that upon treatment with the ER stress inducers thapsigargin (TG) or palmitic acid (PA), the mRNA and protein levels of NR4A1 rapidly increased in both MIN6 cells and mouse islets. NR4A1 overexpression in MIN6 cells conferred resistance to cell loss induced by TG or PA, as assessed by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, and TUNEL assays indicated that NR4A1 overexpression also protected against ER stress-induced apoptosis. This conclusion was further confirmed by experiments exploiting siRNA to knockdown NR4A1 expression in MIN6 cells or exploiting NR4A1 knock-out mice. NR4A1 overexpression in MIN6 cells reduced C/EBP homologous protein (CHOP) expression and Caspase3 activation induced by TG or PA. NR4A1 overexpression in MIN6 cells or mouse islets resulted in Survivin up-regulation. A critical regulatory element was identified in Survivin promoter (-1872 bp to -1866 bp) with a putative NR4A1 binding site; ChIP assays demonstrated that NR4A1 physically associates with the Survivin promoter. In conclusion, NR4A1 protects pancreatic β-cells against ER stress-mediated apoptosis by up-regulating Survivin expression and down-regulating CHOP expression, which we termed as "positive and negative regulation."
Collapse
Affiliation(s)
- Cong Yu
- The Department of Cell Biology, Shandong University School of Medicine, Jinan, China, 250012
| | - Shang Cui
- The Department of Cell Biology, Shandong University School of Medicine, Jinan, China, 250012
| | - Chen Zong
- The Department of Cell Biology, Shandong University School of Medicine, Jinan, China, 250012
| | - Weina Gao
- The Department of Cell Biology, Shandong University School of Medicine, Jinan, China, 250012
| | - Tongfu Xu
- The Department of Cell Biology, Shandong University School of Medicine, Jinan, China, 250012
| | - Peng Gao
- The Department of Cell Biology, Shandong University School of Medicine, Jinan, China, 250012
| | - Jicui Chen
- The Department of Cell Biology, Shandong University School of Medicine, Jinan, China, 250012
| | - Dandan Qin
- The Department of Cell Biology, Shandong University School of Medicine, Jinan, China, 250012
| | - Qingbo Guan
- The Department of Endocrinology, Provincial Hospital affiliated to Shandong University, Jinan, China, 250021
| | - Yuantao Liu
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao, China, 266071
| | - Yuchang Fu
- The Department of Nutrition Sciences, University of Alabama at Birmingham, Alabama 35294
| | - Xia Li
- The Department of Cell Biology, Shandong University School of Medicine, Jinan, China, 250012.
| | - Xiangdong Wang
- The Department of Cell Biology, Shandong University School of Medicine, Jinan, China, 250012; Key Laboratory of Protein Sciences for Chronic Degenerative Diseases in Universities of Shandong (Shandong University), Jinan, China 250012.
| |
Collapse
|
124
|
Hedrick E, Lee SO, Kim G, Abdelrahim M, Jin UH, Safe S, Abudayyeh A. Nuclear Receptor 4A1 (NR4A1) as a Drug Target for Renal Cell Adenocarcinoma. PLoS One 2015; 10:e0128308. [PMID: 26035713 PMCID: PMC4452731 DOI: 10.1371/journal.pone.0128308] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 04/24/2015] [Indexed: 12/20/2022] Open
Abstract
The orphan nuclear receptor NR4A1 exhibits pro-oncogenic activity in cancer cell lines. NR4A1 activates mTOR signaling, regulates genes such as thioredoxin domain containing 5 and isocitrate dehydrogenase 1 that maintain low oxidative stress, and coactivates specificity protein 1 (Sp1)-regulated pro-survival and growth promoting genes. Transfection of renal cell carcinoma (RCC) ACHN and 786-O cells with oligonucleotides that target NR4A1 results in a 40-60% decrease in cell proliferation and induction of apoptosis. Moreover, knockdown of NR4A1 in RCC cells decreased bcl-2, survivin and epidermal growth factor receptor expression, inhibited of mTOR signaling, induced oxidative and endoplasmic reticulum stress, and decreased TXNDC5 and IDH1. We have recently demonstrated that selected 1,1-bis(3'-indolyl)-1-(p-substituted phenyl)methane (C-DIM) compounds including the p-hydroxyphenyl (DIM-C-pPhOH) and p-carboxymethyl (DIM-C-pPhCO2Me) analogs bind NR4A1 and act as antagonists. Both DIM-C-pPhOH and DIM-C-pPhCO2Me inhibited growth and induced apoptosis in ACHN and 786-O cells, and the functional and genomic effects of the NR4A1 antagonists were comparable to those observed after NR4A1 knockdown. These results indicate that NR4A1 antagonists target multiple growth promoting and pro-survival pathways in RCC cells and in tumors (xenograft) and represent a novel chemotherapy for treating RCC.
Collapse
MESH Headings
- Animals
- Apoptosis
- Blotting, Western
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/therapy
- Cell Proliferation
- Fluorescent Antibody Technique
- Humans
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Kidney Neoplasms/therapy
- Male
- Mice
- Mice, Nude
- Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Oligonucleotides, Antisense/genetics
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Erik Hedrick
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, United States of America
| | - Syng-Ook Lee
- Department of Food Science and Technology, Keimyung University, Daegu, Republic of Korea
| | - Gyungeun Kim
- Institute of Biosciences and Technology, Texas A&M Health Sciences Center, Houston, TX, United States of America
| | - Maen Abdelrahim
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, United States of America
- Institute of Biosciences and Technology, Texas A&M Health Sciences Center, Houston, TX, United States of America
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, United States of America
- Institute of Biosciences and Technology, Texas A&M Health Sciences Center, Houston, TX, United States of America
| | - Ala Abudayyeh
- Department of General Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| |
Collapse
|
125
|
Roshan-Moniri M, Hsing M, Butler MS, Cherkasov A, Rennie PS. Orphan nuclear receptors as drug targets for the treatment of prostate and breast cancers. Cancer Treat Rev 2015; 40:1137-52. [PMID: 25455729 DOI: 10.1016/j.ctrv.2014.10.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 10/10/2014] [Accepted: 10/13/2014] [Indexed: 02/06/2023]
Abstract
Nuclear receptors (NRs), a family of 48 transcriptional factors, have been studied intensively for their roles in cancer development and progression. The presence of distinctive ligand binding sites capable of interacting with small molecules has made NRs attractive targets for developing cancer therapeutics. In particular, a number of drugs have been developed over the years to target human androgen- and estrogen receptors for the treatment of prostate cancer and breast cancer. In contrast, orphan nuclear receptors (ONRs), which in many cases lack known biological functions or ligands, are still largely under investigated. This review is a summary on ONRs that have been implicated in prostate and breast cancers, specifically retinoic acid-receptor-related orphan receptors (RORs), liver X receptors (LXRs), chicken ovalbumin upstream promoter transcription factors (COUP-TFs), estrogen related receptors (ERRs), nerve growth factor 1B-like receptors, and ‘‘dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1’’ (DAX1). Discovery and development of small molecules that can bind at various functional sites on these ONRs will help determine their biological functions. In addition, these molecules have the potential to act as prototypes for future drug development. Ultimately, the therapeutic value of targeting the ONRs may go well beyond prostate and breast cancers.
Collapse
|
126
|
Ma W, Zhao R, Yang R, Liu B, Chen X, Wu L, Qi H. Nuclear receptors of the NR4a family are not required for the development and function of follicular T helper cells. Int Immunopharmacol 2015; 28:841-5. [PMID: 25899083 DOI: 10.1016/j.intimp.2015.04.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/05/2015] [Accepted: 04/06/2015] [Indexed: 11/15/2022]
Abstract
Follicular T helper (Tfh) cells promote germinal center (GC) reaction and high-affinity antibody production. The molecular mechanisms that regulate development and function of Tfh cells are not fully understood. Here we report that ligand-independent nuclear receptors of the Nr4a family are highly expressed in Tfh cells. In a well-established adoptive transfer model, enforced expression of Nr4a receptors reduces helper T cell expansion but apparently increased the T cell capacity to promote the GC response. On the other hand, deletion of all Nr4a receptors in T cells did not significantly affect expansion or differentiation of Tfh cells or the development of GC reaction. These findings suggest that Nr4a receptors may promote but are not necessary for Tfh development or function in vivo.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cells, Cultured
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/immunology
- Germinal Center
- Mice, Inbred C57BL
- Mice, Transgenic
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/immunology
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/immunology
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/immunology
- RNA, Messenger/metabolism
- Receptors, Steroid/genetics
- Receptors, Steroid/immunology
- Receptors, Thyroid Hormone/genetics
- Receptors, Thyroid Hormone/immunology
- T-Lymphocytes, Helper-Inducer/cytology
- T-Lymphocytes, Helper-Inducer/immunology
Collapse
Affiliation(s)
- Weiwei Ma
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, PR China.
| | - Ruozhu Zhao
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, PR China
| | - Runqing Yang
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, PR China
| | - Bo Liu
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, PR China
| | - Xin Chen
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, PR China
| | - Longyan Wu
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, PR China
| | - Hai Qi
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology Institute for Immunology, School of Medicine, Tsinghua University, Beijing 100084, PR China.
| |
Collapse
|
127
|
Watanabe T, Sekine S, Naguro I, Sekine Y, Ichijo H. Apoptosis Signal-regulating Kinase 1 (ASK1)-p38 Pathway-dependent Cytoplasmic Translocation of the Orphan Nuclear Receptor NR4A2 Is Required for Oxidative Stress-induced Necrosis. J Biol Chem 2015; 290:10791-803. [PMID: 25752609 DOI: 10.1074/jbc.m114.623280] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Indexed: 11/06/2022] Open
Abstract
p38 mitogen-activated protein kinases (MAPKs) play important roles in various cellular stress responses, including cell death, which is roughly categorized into apoptosis and necrosis. Although p38 signaling has been extensively studied, the molecular mechanisms of p38-mediated cell death are unclear. ASK1 is a stress-responsive MAP3K that acts as an upstream kinase of p38 and is activated by various stresses, such as oxidative stress. Here, we show that NR4A2, a member of the NR4A nuclear receptor family, acts as a necrosis promoter downstream of ASK1-p38 pathway during oxidative stress. Although NR4A2 is well known as a nucleus-localized transcription factor, we found that it is translocated into the cytosol after phosphorylation by p38. Because the phosphorylation site mutants of NR4A2 cannot rescue the cell death-promoting activity, ASK1-p38 pathway-dependent phosphorylation and subsequent cytoplasmic translocation of NR4A2 may be required for oxidative stress-induced cell death. In addition, NR4A2-mediated cell death does not depend on caspases and receptor-interacting protein 1 (RIP1)-RIP3 complex, suggesting that NR4A2 promotes an RIP kinase-independent necrotic type of cell death. Our findings may enable a more precise understanding of molecular mechanisms that regulate oxidative stress-induced and p38-mediated necrosis.
Collapse
Affiliation(s)
- Takeshi Watanabe
- From the Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, the Department of Cellular Physiological Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Yushima, 1-5-45 Bunkyo-ku, Tokyo 113-8510, Japan, and
| | - Shiori Sekine
- From the Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Isao Naguro
- From the Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yusuke Sekine
- From the Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan, the Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Hidenori Ichijo
- From the Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan,
| |
Collapse
|
128
|
Pawlak A, Strzadala L, Kalas W. Non-genomic effects of the NR4A1/Nur77/TR3/NGFIB orphan nuclear receptor. Steroids 2015; 95:1-6. [PMID: 25555471 DOI: 10.1016/j.steroids.2014.12.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 10/07/2014] [Accepted: 12/18/2014] [Indexed: 11/30/2022]
Abstract
The orphan nuclear receptor NR4A1/Nur77/TR3/NGFIB acts primarily as a transcription factor to regulate the expression of multiple genes. However, increasing research attention has recently been given to non-genomic activities of NR4A1. The first description of a non-genomic action of NR4A1 referred to the conversion of anti-apoptotic Bcl-2 into a pro-apoptotic protein by direct interaction with NR4A1. In response to certain apoptotic stimuli, NR4A1 translocates from the nucleus to the mitochondrial outer membrane (MOM) where it associates with Bcl-2 and thereby causes apoptosis. Afterwards, it appeared that NR4A1 could also bind and convert other anti-apoptotic Bcl-2 family members. The latest studies indicate a significant role of NR4A1 in the process of autophagy. For example, a new NR4A1-mediated pathway specific for melanoma cells has been described where NR4A1 interacts with the adenine nucleotide translocase 1 (ANT1) on the mitochondrial inner membrane (MIM) leading to induction of the autophagy pathway. Moreover, NR4A1 interaction with cytoplasmic p53 may also contribute to the induction of autophagy. In addition to mitochondria, NR4A1 could be translocated to the outer membrane of the endoplasmic reticulum (ER) and associate with Bcl-2 or translocon-associated protein subunit γ (TRAPγ) causing ER stress-induced apoptosis. NR4A1 also contributes to the proteasomal degradation of β-catenin in colon cancer cells in vitro and in vivo, as well as to the stabilization of hypoxia-inducible factor-1α (HIF-1α) under non-hypoxic conditions. This review summarizes research findings on non-genomic effects of NR4A1 in normal and cancer cells.
Collapse
Affiliation(s)
- Alicja Pawlak
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wroclaw, Poland.
| | - Leon Strzadala
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wroclaw, Poland.
| | - Wojciech Kalas
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wroclaw, Poland; Jan Dlugosz University in Czestochowa, Waszyngtona 4/8, 42-200 Czestochowa, Poland.
| |
Collapse
|
129
|
Palumbo-Zerr K, Zerr P, Distler A, Fliehr J, Mancuso R, Huang J, Mielenz D, Tomcik M, Fürnrohr BG, Scholtysek C, Dees C, Beyer C, Krönke G, Metzger D, Distler O, Schett G, Distler JHW. Orphan nuclear receptor NR4A1 regulates transforming growth factor-β signaling and fibrosis. Nat Med 2015; 21:150-8. [DOI: 10.1038/nm.3777] [Citation(s) in RCA: 272] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023]
|
130
|
Wang J, Yang J, Li BB, He ZW. High cytoplasmic expression of the orphan nuclear receptor NR4A2 predicts poor survival in nasopharyngeal carcinoma. Asian Pac J Cancer Prev 2015; 14:2805-9. [PMID: 23803035 DOI: 10.7314/apjcp.2013.14.5.2805] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE This study aimed at investigating whether the orphan nuclear receptor NR4A2 is significantly associated with clinicopathologic features and overall survival of patients with nasopharyngeal carcinoma (NPC). METHODS Immunohistochemistry was performed to determine NR4A2 protein expression in 84 NPC tissues and 20 non-cancerous nasopharyngeal (NP) tissues. The prognostic significance of NR4A2 protein expression was evaluated using Cox proportional hazards regression models and Kaplan-Meier survival analysis. RESULTS We did not find a significant association between total NR4A2 expression and clinicopathological variables in 84 patients with NPC. However, we observed that high cytoplasmic expression of NR4A2 was significantly associated with tumor size (T classification) (P = 0.006), lymph node metastasis (N classification) (P = 0.002) and clinical stage (P = 0.017). Patients with higher cytoplasmic NR4A2 expression had a significantly lower survival rate than those with lower cytoplasmic NR4A2 expression (P = 0.004). Multivariate Cox regression analysis analysis suggested that the level of cytoplasmic NR4A2 expression was an independent prognostic indicator for overall survival of patients with NPC (P = 0.033). CONCLUSIONS High cytoplasmic expression of NR4A2 is a potential unfavorable prognostic factor for patients with NPC.
Collapse
Affiliation(s)
- Jian Wang
- Sino-American Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Guangdong Medical College, Dongguan, China
| | | | | | | |
Collapse
|
131
|
Abstract
While all herpesviruses can switch between lytic and latent life cycle, which are both driven by specific transcription programs, a unique feature of latent EBV infection is the expression of several distinct and well-defined viral latent transcription programs called latency I, II, and III. Growth transformation of B-cells by EBV in vitro is based on the concerted action of Epstein-Barr virus nuclear antigens (EBNAs) and latent membrane proteins(LMPs). EBV growth-transformed B-cells express a viral transcriptional program, termed latency III, which is characterized by the coexpression of EBNA2 and EBNA-LP with EBNA1, EBNA3A, -3B, and -3C as well as LMP1, LMP2A, and LMP2B. The focus of this review will be to discuss the current understanding of how two of these proteins, EBNA2 and EBNA-LP, contribute to EBV-mediated B-cell growth transformation.
Collapse
Affiliation(s)
- Bettina Kempkes
- Department of Gene Vectors, Helmholtz Center Munich, German Research Center for Environmental Health, Marchioninistr. 25, 81377, Munich, Germany.
| | - Paul D Ling
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
132
|
Zhang XK, Su Y, Chen L, Chen F, Liu J, Zhou H. Regulation of the nongenomic actions of retinoid X receptor-α by targeting the coregulator-binding sites. Acta Pharmacol Sin 2015; 36:102-12. [PMID: 25434990 DOI: 10.1038/aps.2014.109] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 09/28/2014] [Indexed: 12/31/2022]
Abstract
Retinoid X receptor-α (RXRα), a unique member of the nuclear receptor superfamily, represents an intriguing and unusual target for pharmacologic interventions and therapeutic applications in cancer, metabolic disorders and neurodegenerative diseases. Despite the fact that the RXR-based drug Targretin (bexarotene) is currently used for treating human cutaneous T-cell lymphoma and the fact that RXRα ligands (rexinoids) show beneficial effects in the treatment of cancer and diseases, the therapeutic potential of RXRα remains unexplored. In addition to its conventional transcription regulation activity in the nucleus, RXRα can act in the cytoplasm to modulate important biological processes, such as mitochondria-dependent apoptosis, inflammation, and phosphatidylinositol 3-kinase (PI3K)/AKT-mediated cell survival. Recently, new small-molecule-binding sites on the surface of RXRα have been identified, which mediate the regulation of the nongenomic actions of RXRα by a class of small molecules derived from the nonsteroidal anti-inflammatory drug (NSAID) Sulindac. This review discusses the emerging roles of the nongenomic actions of RXRα in the RXRα signaling network, and their possible implications in cancer, metabolic and neurodegenerative disorders, as well as our current understanding of RXRα regulation by targeting alternate binding sites on its surface.
Collapse
|
133
|
Deng X, Qiu Q, Yang B, Wang X, Huang W, Qian H. Design, synthesis and biological evaluation of novel peptides with anti-cancer and drug resistance-reversing activities. Eur J Med Chem 2015; 89:540-8. [DOI: 10.1016/j.ejmech.2014.10.072] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 10/24/2014] [Accepted: 10/25/2014] [Indexed: 10/24/2022]
|
134
|
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: 72] [Impact Index Per Article: 6.5] [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
|
135
|
Tabassum S, Yadav S, Arjmand F. Exploration of glycosylated-organotin(IV) complexes as anticancer drug candidates. Inorganica Chim Acta 2014; 423:38-45. [DOI: 10.1016/j.ica.2014.07.080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
136
|
Niu L, Li Y, Li Q. Medicinal properties of organotin compounds and their limitations caused by toxicity. Inorganica Chim Acta 2014. [DOI: 10.1016/j.ica.2014.05.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
137
|
Arjmand F, Parveen S, Tabassum S, Pettinari C. Organo-tin antitumor compounds: Their present status in drug development and future perspectives. Inorganica Chim Acta 2014; 423:26-37. [DOI: 10.1016/j.ica.2014.07.066] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
138
|
Lee SO, Li X, Hedrick E, Jin UH, Tjalkens RB, Backos DS, Li L, Zhang Y, Wu Q, Safe S. Diindolylmethane analogs bind NR4A1 and are NR4A1 antagonists in colon cancer cells. Mol Endocrinol 2014; 28:1729-1739. [PMID: 25099012 PMCID: PMC4179635 DOI: 10.1210/me.2014-1102] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 07/29/2014] [Indexed: 12/20/2022] Open
Abstract
1,1-Bis(3'-indolyl)-1-(p-substituted phenyl)methane (C-DIM) compounds exhibit antineoplastic activity in multiple cancer cell lines and the p-hydroxyphenyl analog (DIM-C-pPhOH) inactivates nuclear receptor 4A1 (NR4A1) in lung and pancreatic cancer cell lines. Using a series of 14 different p-substituted phenyl C-DIMs, we show that several compounds including DIM-C-pPhOH directly interacted with the ligand binding domain of NR4A1. Computational-based molecular modeling studies showed high-affinity interactions of DIM-C-pPhOH and related compounds within the ligand binding pocket of NR4A1, and these same compounds decreased NR4A1-dependent transactivation in colon cancer cells transfected with a construct containing 3 tandem Nur77 binding response elements linked to a luciferase reporter gene. Moreover, we also show that knockdown of NR4A1 by RNA interference (small interfering NR4A1) or treatment with DIM-C-pPhOH and related compounds decreased colon cancer cell growth, induced apoptosis, decreased expression of survivin and other Sp-regulated genes, and inhibited mammalian target of rapamycin signaling. Thus, C-DIMs such as DIM-C-pPhOH directly bind NR4A1 and are NR4A1 antagonists in colon cancer cells, and their antineoplastic activity is due, in part, to their interactions with nuclear NR4A1.
Collapse
Affiliation(s)
- Syng-Ook Lee
- Department of Food Science and Technology (S.-O.L.), Keimyung University, Daegu 704-701, Republic of Korea; College of Medicine (X.L.), Texas A&M Health Science Center, and Department of Veterinary Physiology and Pharmacology (E.H., S.S.), Texas A&M University, College Station, TX 77843; Institute of Bioscience and Technology (U.-H.J., S.S.), Texas A&M Health Science Center, Houston, Texas 77030; Center for Environmental Medicine (R.B.T.), Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523; Department of Pharmaceutical Sciences (D.S.B.), University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045; and School of Life Sciences (L.L., Y.Z., Q.W.), University of Xiamen, Xiamen, 361005 Fujian, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Zhou Y, Zhao W, Xie G, Huang M, Hu M, Jiang X, Zeng D, Liu J, Zhou H, Chen H, Wang GH, Zhang XK. Induction of Nur77-dependent apoptotic pathway by a coumarin derivative through activation of JNK and p38 MAPK. Carcinogenesis 2014; 35:2660-9. [PMID: 25187486 DOI: 10.1093/carcin/bgu186] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Coumarins are plant-derived natural products with a broad range of known pharmacological activities including anticancer effects. However, the molecular mechanisms by which this class of promising compounds exerts their anticancer effects remain largely unknown. We report here that a furanocoumarin named apaensin could effectively induce apoptosis of cancer cells through its activation of Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). Apoptosis induction by apaensin in cancer cells was suppressed by chemical inhibitors of JNK and p38 MAPK. Inhibition of the expression of orphan nuclear receptor Nur77 by small interfering RNA (siRNA) approach also abrogated the death effect of apaensin. Molecular analysis demonstrated that JNK activation was required for the nuclear export of Nur77, a known apoptotic event in cancer cells. Although p38 MAPK activation was not involved in Nur77 nuclear export, it was essential for Nur77 mitochondrial targeting through induction of Nur77 interaction with Bcl-2, which is also known to convert Bcl-2 from an antiapoptotic to a proapoptotic molecule. Together, our results identify a new natural product that targets orphan nuclear receptor Nur77 through its unique activation of JNK and p38 MAPK and provide insight into the complex regulation of the Nur77-Bcl-2 apoptotic pathway.
Collapse
Affiliation(s)
- Yuqi Zhou
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Wei Zhao
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Guobin Xie
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Mingfeng Huang
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Mengjie Hu
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Xin Jiang
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Dequan Zeng
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Jie Liu
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Hu Zhou
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and Cancer Center, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Haifeng Chen
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Guang-Hui Wang
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and
| | - Xiao-Kun Zhang
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian 361005, China and Cancer Center, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
140
|
Dai Y, Zhang W, Sun Q, Zhang X, Zhou X, Hu Y, Shi J. Nuclear receptor nur77 promotes cerebral cell apoptosis and induces early brain injury after experimental subarachnoid hemorrhage in rats. J Neurosci Res 2014; 92:1110-21. [PMID: 24737679 DOI: 10.1002/jnr.23392] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/14/2014] [Accepted: 03/14/2014] [Indexed: 01/11/2023]
Abstract
Nur77 is a potent proapoptotic member of the nuclear receptor superfamily that is expressed predominantly in brain tissue. It has been demonstrated that Nur77 mediates apoptosis in multiple organs. Nur77-mediated early brain injury (EBI) involves a conformational change in BCL-2 and triggers cytochrome C (cytoC) release resulting in cellular apoptosis. This study investigates whether Nur77 can promote cerebral cell apoptosis after experimentally induced subarachnoid hemorrhage (SAH) in rats. Sprague Dawley rats were randomly assigned to three groups: 1) untreated group, 2) treatment control group, and 3) SAH group. The experimental SAH group was divided into four subgroups, corresponding to 12 hr, 24 hr, 48 hr, and 72 hr after experimentally induced SAH. It remains unclear whether Nur77 can play an important role during EBI after SAH as a proapoptotic protein in cerebral cells. Cytosporone B (Csn-B) was used to demonstrate that Nur77 could be enriched and used to aggravate EBI after SAH. Rats treated with Csn-B were given an intraperitoneal injection (13 mg/kg) 30 min after experimentally induced SAH. We found that Nur77 promotes cerebral cell apoptosis by mediating EBI and triggering a conformational change in BCL-2, resulting in cytoC release. Nur77 activity, along with cerebral cell apoptosis, peaked at 24 hr after SAH onset. After induction of SAH, an injection of Csn-B, an agonist for Nur77, enhanced the expression and function of Nur77. In summary, we have demonstrated the proapoptotic effect of Nur77 within cerebral cells, an effect that can be further exacerbated with Csn-B stimulation.
Collapse
Affiliation(s)
- Yuxiang Dai
- Jinling Hospital, School of Medicine, Department of Neurosurgery, Nanjing University, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
141
|
Rollano Peñaloza OM, Lewandowska M, Stetefeld J, Ossysek K, Madej M, Bereta J, Sobczak M, Shojaei S, Ghavami S, Łos MJ. Apoptins: selective anticancer agents. Trends Mol Med 2014; 20:519-28. [PMID: 25164066 DOI: 10.1016/j.molmed.2014.07.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/17/2014] [Accepted: 07/17/2014] [Indexed: 12/20/2022]
Abstract
Therapies that selectively target cancer cells for death have been the center of intense research recently. One potential therapy may involve apoptin proteins, which are able to induce apoptosis in cancer cells leaving normal cells unharmed. Apoptin was originally discovered in the Chicken anemia virus (CAV); however, human gyroviruses (HGyV) have recently been found that also harbor apoptin-like proteins. Although the cancer cell specific activity of these apoptins appears to be well conserved, the precise functions and mechanisms of action are yet to be fully elucidated. Strategies for both delivering apoptin to treat tumors and disseminating the protein inside the tumor body are now being developed, and have shown promise in preclinical animal studies.
Collapse
Affiliation(s)
- Oscar M Rollano Peñaloza
- Department Clinical & Experimental Medicine, Division of Cell Biology, and Integrative Regenerative Medical Center, Linköping University, Linköping, Sweden; Instituto de Biologia Molecular y Biotecnologia, La Paz, Bolivia
| | | | - Joerg Stetefeld
- Department of Chemistry, University of Manitoba, Winnipeg, Canada
| | - Karolina Ossysek
- Department of Cell Biochemistry, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mariusz Madej
- Department of Cell Biochemistry, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Joanna Bereta
- Department of Cell Biochemistry, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mateusz Sobczak
- Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Shahla Shojaei
- Department of Biochemistry, Recombinant Protein Laboratory, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Ghavami
- Department of Human Anatomy & Cell Science, College of Medicine, Faculty of Health Sciences, and Manitoba Institute of Child Health, University of Manitoba, Winnipeg, Canada; Health Policy Research Centre, Shiraz University of Medical Science, Shiraz, Iran
| | - Marek J Łos
- Department Clinical & Experimental Medicine, Division of Cell Biology, and Integrative Regenerative Medical Center, Linköping University, Linköping, Sweden; Department of Pathology, Pomeranian Medical University, Szczecin, Poland.
| |
Collapse
|
142
|
Wang H, Wang X, Zhang S, Wang P, Zhang K, Liu Q. Sinoporphyrin sodium, a novel sensitizer, triggers mitochondrial-dependent apoptosis in ECA-109 cells via production of reactive oxygen species. Int J Nanomedicine 2014; 9:3077-90. [PMID: 25028547 PMCID: PMC4077608 DOI: 10.2147/ijn.s59302] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Sonodynamic therapy (SDT) is a promising method that uses ultrasound to activate certain chemical sensitizers for the treatment of cancer. The purpose of this study was to investigate the sonoactivity of a novel sensitizer, sinoporphyrin sodium (DVDMS), and its sonotoxicity in an esophageal cancer (ECA-109) cell line. Methods The fluorescence intensity of DVDMS, hematoporphyrin, protoporphyrin IX, and Photofrin II was detected by fluorescence microscopy and flow cytometry. Generation of singlet oxygen was measured using a 1, 3-diphenylisobenzofuran experiment. A 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide assay was used to examine cell viability. Production of reactive oxygen species (ROS) and destabilization of the mitochondrial membrane potential were assessed by flow cytometry. Apoptosis was analyzed using Annexin-PE/7-amino-actinomycin D staining. Confocal microscopy was performed to assess mitochondrial damage and identify release of cytochrome C after treatment. Western blots were used to determine expression of oxidative stress-related and apoptosis-associated protein. Ultrastructural changes in the cell were studied by scanning electron microscopy. Results DVDMS showed higher autofluorescence intensity and singlet oxygen production efficiency compared with other photosensitizers in both cancerous and normal cells. Compared with hematoporphyrin, DVDMS-mediated SDT was more cytotoxic in ECA-109 cells. Abundant intracellular ROS was found in the SDT groups, and the cytotoxicity induced by SDT was effectively remitted by ROS scavengers. DVDMS located mainly to the mitochondria of ECA-109 cells, which were seriously damaged after exposure to SDT. Release of cytochrome C, an increased rate of apoptosis, and activated apoptosis protein were detected in the SDT group. In addition, relatively severe cell damage was observed on scanning electron microscopy after treatment with DVDMS and SDT. Conclusion These results suggest that DVDMS could be activated by ultrasound, and that DVDMS mediates SDT-induced mitochondrial-dependent apoptosis in ECA-109 cells via production of ROS.
Collapse
Affiliation(s)
- Haiping Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, People's Republic of China
| | - Xiaobing Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, People's Republic of China
| | - Shaoliang Zhang
- Qinglong High-Tech Co, Ltd, Yichun, Jiangxi, People's Republic of China
| | - Pan Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, People's Republic of China
| | - Kun Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, People's Republic of China
| | - Quanhong Liu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
143
|
Dai Y, Zhang W, Zhou X, Shi J. Inhibition of c-Jun N-terminal kinase ameliorates early brain injury after subarachnoid hemorrhage through inhibition of a Nur77 dependent apoptosis pathway. Neurochem Res 2014; 39:1603-11. [PMID: 24928238 DOI: 10.1007/s11064-014-1355-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 05/26/2014] [Accepted: 06/03/2014] [Indexed: 12/21/2022]
Abstract
Nur77 is a potent pro-apoptotic member of the orphan nuclear receptor superfamily. Our previous study revealed Nur77-mediated apoptotic also involved in early brain injury (EBI) after experimental subarachnoid hemorrhage (SAH). Previous researches show that c-Jun N-terminal kinase (JNK) positively regulates Nur77 nuclear export and apoptosis by phosphorylating Nur77. To determine whether activation of JNK is directly associated with Nur77 dependent apoptosis pathway. We hypothesized that SP600125, a chemical inhibitor of JNK, may effectively ameliorate EBI by inhibiting Nur77 phosphorylation and its transcriptional activity. Hence, in this study was designed to explore the neuroprotective effects of SP600125 in EBI after SAH. Adult male SD rats were randomly assigned to four groups: control; SAH + DMSO; SAH + SP10 and SAH + SP30, a dose of 10 and 30 mg/kg SP600125 was directly administered intraperitoneally 30 min before and 2 h after SAH induction. SP600125 markedly decreased expressions of p-JNK, p-Nur77, Bcl-2, cyto C, caspase-3 and inhibited apoptosis. Improvement of neurological deficit, alleviation of brain edema and amelioration of EBI were obtained after treatment of SP600125. Transferase-mediated dUTP nick end labeling-positive cells were reduced markedly in brain cortex by SP600125. Our studies indicate JNK plays important roles in Nur77 activation. These findings strongly support the hypothesis that SP600125 treatment can ameliorate EBI after experimentally induced SAH by inhibiting a Nur77-dependent apoptotic pathway.
Collapse
Affiliation(s)
- Yuxiang Dai
- Department of Neurosurgery, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | | | | | | |
Collapse
|
144
|
Abstract
Hypospadias is a congenital hypoplasia of the penis, with displacement of the urethral opening along the ventral surface, and has been reported to be one of the most common congenital anomalies, occurring in approximately 1:250 to 1:300 live births. As hypospadias is reported to be an easily diagnosed malformation at the crossroads of genetics and environment, it is important to study the genetic component in order to elucidate its etiology. In this study, the gene expression profiles both in human hypospadias tissues and normal penile tissues were studied by Human Gene Expression Array. Twenty-four genes were found to be upregulated. Among these, ATF3 and CYR61 have been reported previously. Other genes that have not been previously reported were also found to be upregulated: BTG2, CD69, CD9, DUSP1, EGR1, EIF4A1, FOS, FOSB, HBEGF, HNRNPUL1, IER2, JUN, JUNB, KLF2, NR4A1, NR4A2, PTGS2, RGS1, RTN4, SLC25A25, SOCS3 and ZFP36 (p <0.05). Further studies including genome-wide association studies (GWAS) with expression studies in a large patient group will help us for identifiying the candidate gene(s) in the etiology of hypospadias.
Collapse
|
145
|
Kim YC, Song SB, Lee SK, Park SM, Kim YS. The Nuclear Orphan Receptor NR4A1 is Involved in the Apoptotic Pathway Induced by LPS and Simvastatin in RAW 264.7 Macrophages. Immune Netw 2014; 14:116-22. [PMID: 24851101 PMCID: PMC4022779 DOI: 10.4110/in.2014.14.2.116] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 03/26/2014] [Accepted: 03/28/2014] [Indexed: 01/16/2023] Open
Abstract
Macrophage death plays a role in several physiological and inflammatory pathologies such as sepsis and arthritis. In our previous work, we showed that simvastatin triggers cell death in LPS-activated RAW 264.7 mouse macrophage cells through both caspase-dependent and independent apoptotic pathways. Here, we show that the nuclear orphan receptor NR4A1 is involved in a caspase-independent apoptotic process induced by LPS and simvastatin. Simvastatin-induced NR4A1 expression in RAW 264.7 macrophages and ectopic expression of a dominant-negative mutant form of NR4A1 effectively suppressed both DNA fragmentation and the disruption of mitochondrial membrane potential (MMP) during LPS- and simvastatin-induced apoptosis. Furthermore, apoptosis was accompanied by Bcl-2-associated X protein (Bax) translocation to the mitochondria. Our findings suggest that NR4A1 expression and mitochondrial translocation of Bax are related to simvastatin-induced apoptosis in LPS-activated RAW 264.7 macrophages.
Collapse
Affiliation(s)
- Yong Chan Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea
| | - Seok Bean Song
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea
| | - Sang Kyu Lee
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea
| | - Sang Min Park
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea
| | - Young Sang Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea
| |
Collapse
|
146
|
Xu A, Liu J, Liu P, Jia M, Wang H, Tao L. Mitochondrial translocation of Nur77 induced by ROS contributed to cardiomyocyte apoptosis in metabolic syndrome. Biochem Biophys Res Commun 2014; 446:1184-9. [PMID: 24680679 DOI: 10.1016/j.bbrc.2014.03.089] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 03/18/2014] [Indexed: 10/25/2022]
Abstract
Metabolic syndrome is a major risk factor for cardiovascular diseases, and increased cardiomyocyte apoptosis which contributes to cardiac dysfunction after myocardial ischemia/reperfusion (MI/R) injury. Nur77, a nuclear orphan receptor, is involved in such various cellular events as apoptosis, proliferation, and glucose and lipid metabolism in several cell types. Apoptosis is positively correlated with mitochondrial translocation of Nur77 in the cancer cells. However, the roles of Nur77 on cardiac myocytes in patients with metabolic syndrome remain unclear. The objective of this study was to determine whether Nur77 may contribute to cardiac apoptosis in patients with metabolic syndrome after I/R injury, and, if so, to identify the underlying molecular mechanisms responsible. We used leptin-deficient (ob/ob) mice to make metabolic syndrome models. In this report, we observed that, accompanied by the substantial decline in apoptosis inducer Nur77, MI/R induced cardiac dysfunction was manifested as cardiomyopathy and increased ROS. Using the neonatal rat cardiac myocytes cultured in a high-glucose and high-fat medium, we found that excessive H2O2 led to the significant alteration in mitochondrial membrane potential and translocation of Nur77 from the nucleus to the mitochondria. However, inhibition of the relocation of Nur77 to mitochondria via Cyclosporin A reversed the changes in membrane potential mediated by H2O2 and reduced myocardial cell injury. Therefore, these data provide a potential underlying mechanism for cardiac dysfunction in metabolic syndrome and the suppression of Nur77 translocation may provide an effective approach to reduce cardiac injury in the process.
Collapse
Affiliation(s)
- Aibin Xu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China; Institute of Cardiovascular Disease, General Hospital of Beijing Command, PLA, Beijing, People's Republic of China
| | - Jingyi Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China; Institute of Cardiovascular Disease, General Hospital of Beijing Command, PLA, Beijing, People's Republic of China
| | - Peilin Liu
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Min Jia
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Han Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China.
| |
Collapse
|
147
|
Gao W, Fu Y, Yu C, Wang S, Zhang Y, Zong C, Xu T, Liu Y, Li X, Wang X. Elevation of NR4A3 expression and its possible role in modulating insulin expression in the pancreatic beta cell. PLoS One 2014; 9:e91462. [PMID: 24638142 PMCID: PMC3956668 DOI: 10.1371/journal.pone.0091462] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/11/2014] [Indexed: 11/19/2022] Open
Abstract
Background NR4A3/NOR-1 is a member of the NR4A orphan nuclear receptor subfamily, which contains early response genes that sense and respond to a variety of stimuli in the cellular environment. The role of NR4A3 in insulin expression in pancreatic beta cells remains unknown. Methods Dynamic changes in NR4A3 were examined in a pancreatic beta-cell line, MIN6, treated with thapsigargin (TG), palmitate (PA), tunicamycin (TM), and dithiothreitol (DTT), chemicals that produce cell stress and even apoptosis. We exploited virus infection techniques to induce expression of NR4A3 or three deletion mutants, and determined expression of insulin and insulin regulatory genes in MIN6 cells. Results TG and PA, two endoplasmic reticulum (ER) stress inducers, were able to induce unfolded protein response (UPR) activation and elevation of NR4A3 expression in MIN6 cells, whereas TM and DTT, two other ER stress inducers, were able to induce UPR activation but not NR4A3 elevation. MIN6 cells over-expressing NR4A3 protein after adenoviral infection exhibited reduced transcription of the insulin genes Ins1 and Ins2, and reduced insulin protein secretion, which were negatively correlated with NR4A3 expression levels. Functional analysis of different deletion mutants of NR4A3 showed that deleting the activation domain AF1 or the DNA-binding domain abolished the down-regulation of insulin transcription by NR4A3 in MIN6 cells, indicating that this down-regulative role was closely related to the NR4A3 trans-activation activity. Over-expression of NR4A3 in MIN6 cells resulted in reduced mRNA transcription of the insulin positive-regulation genes, Pdx1 and NeuroD1. Conclusion Some ER stress inducers, such as TG or PA, are able to elevate NR4A3 expression in MIN6 cells, while others, such as TM or DTT, are not. Over-expression of NR4A3 in MIN6 cells results in down-regulation of insulin gene transcription and insulin secretion. NR4A3 reduces insulin gene expression by modulating the expression of Pdx1 and NeuroD1.
Collapse
Affiliation(s)
- Weina Gao
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
- School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, China
| | - Yuchang Fu
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Cong Yu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Shunke Wang
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Yuchao Zhang
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Chen Zong
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Tongfu Xu
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
| | - Yong Liu
- The Institute for Nutritional Sciences, Chinese Academy of Science, Shanghai, China
| | - Xia Li
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
- * E-mail: (XL); (XW)
| | - Xiangdong Wang
- Department of Cell Biology, Shandong University School of Medicine, Jinan, China
- Key Laboratory of Protein Sciences for Chronic Degenerative Diseases in Universities of Shandong (Shandong University), Jinan, China
- * E-mail: (XL); (XW)
| |
Collapse
|
148
|
Dai Y, Sun Q, Zhang X, Hu Y, Zhou M, Shi J. Cyclosporin A ameliorates early brain injury after subarachnoid hemorrhage through inhibition of a Nur77 dependent apoptosis pathway. Brain Res 2014; 1556:67-76. [DOI: 10.1016/j.brainres.2014.01.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Revised: 01/25/2014] [Accepted: 01/30/2014] [Indexed: 12/25/2022]
|
149
|
Safe S, Jin UH, Hedrick E, Reeder A, Lee SO. Minireview: role of orphan nuclear receptors in cancer and potential as drug targets. Mol Endocrinol 2014; 28:157-172. [PMID: 24295738 PMCID: PMC3896638 DOI: 10.1210/me.2013-1291] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 11/21/2013] [Indexed: 01/03/2023] Open
Abstract
The nuclear orphan receptors for which endogenous ligands have not been identified include nuclear receptor (NR)0B1 (adrenal hypoplasia congenita critical region on chromosome X gene), NR0B2 (small heterodimer partner), NR1D1/2 (Rev-Erbα/β), NR2C1 (testicular receptor 2), NR2C2 (testicular receptor 4), NR2E1 (tailless), NR2E3 (photoreceptor-specific NR [PNR]), NR2F1 chicken ovalbumin upstream promoter transcription factor 1 (COUP-TFI), NR2F2 (COUP-TFII), NR2F6 (v-erbA-related protein), NR4A1 (Nur77), NR4A2 (Nurr1), NR4A3 (Nor1), and NR6A1 (GCNF). These receptors play essential roles in development, cellular homeostasis, and disease including cancer where over- or underexpression of some receptors has prognostic significance for patient survival. Results of receptor knockdown or overexpression in vivo and in cancer cell lines demonstrate that orphan receptors exhibit tumor-specific pro-oncogenic or tumor suppressor-like activity. For example, COUP-TFII expression is both a positive (ovarian) and negative (prostate and breast) prognostic factor for cancer patients; in contrast, the prognostic activity of adrenal hypoplasia congenita critical region on chromosome X gene for the same tumors is the inverse of COUP-TFII. Functional studies show that Nur77 is tumor suppressor like in acute leukemia, whereas silencing Nur77 in pancreatic, colon, lung, lymphoma, melanoma, cervical, ovarian, gastric, and some breast cancer cell lines induces one or more of several responses including growth inhibition and decreased survival, migration, and invasion. Although endogenous ligands for the orphan receptors have not been identified, there is increasing evidence that different structural classes of compounds activate, inactivate, and directly bind several orphan receptors. Thus, the screening and development of selective orphan receptor modulators will have important clinical applications as novel mechanism-based agents for treating cancer patients overexpressing one or more orphan receptors and also for combined drug therapies.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology (S.S., E.H., A.R.), Texas A&M University, College Station, Texas 77808; and Institute of Biosciences and Technology (S.S., U.-H.J., S.-O.L.), Texas A&M Health Science Center, Houston, Texas 77030
| | | | | | | | | |
Collapse
|
150
|
The aryl hydrocarbon receptor mediates raloxifene-induced apoptosis in estrogen receptor-negative hepatoma and breast cancer cells. Cell Death Dis 2014; 5:e1038. [PMID: 24481452 PMCID: PMC4040680 DOI: 10.1038/cddis.2013.549] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 11/04/2013] [Accepted: 11/05/2013] [Indexed: 12/21/2022]
Abstract
Identification of new molecular targets for the treatment of breast cancer is an important clinical goal, especially for triple-negative breast cancer, which is refractory to existing targeted treatments. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor known primarily as the mediator of dioxin toxicity. However, the AhR can also inhibit cellular proliferation in a ligand-dependent manner and act as a tumor suppressor in mice, and thus may be a potential anticancer target. To investigate the AhR as an anticancer target, we conducted a small molecule screen to discover novel AhR ligands with anticancer properties. We identified raloxifene, a selective estrogen receptor (ER) modulator currently used in the clinic for prevention of ER-positive breast cancer and osteoporosis in post-menopausal women, as an AhR activator. Raloxifene directly bound the AhR and induced apoptosis in ER-negative mouse and human hepatoma cells in an AhR-dependent manner, indicating that the AhR is a molecular target of raloxifene and mediates raloxifene-induced apoptosis in the absence of ER. Raloxifene selectively induced apoptosis of triple-negative MDA-MB-231 breast cancer cells compared with non-transformed mammary epithelial cells via the AhR. Combined with recent data showing that raloxifene inhibits triple-negative breast cancer xenografts in vivo (Int J Oncol. 43(3):785-92, 2013), our results support the possibility of repurposing of raloxifene as an AhR-targeted therapeutic for triple-negative breast cancer patients. To this end, we also evaluated the role of AhR expression on survival of patients diagnosed with breast cancer. We found that higher expression of the AhR is significantly associated with increased overall survival and distant metastasis-free survival in both hormone-dependent (ER-positive) and hormone-independent (ER and progesterone receptor (PR)-negative) breast cancers. Together, our data strongly support the possibility of using the AhR as a molecular target for the treatment of hormone-independent breast cancers.
Collapse
|