1
|
Rocha SM, Gustafson DL, Safe S, Tjalkens RB. Comparative safety, pharmacokinetics, and off-target assessment of 1,1-bis(3'-indolyl)-1-( p-chlorophenyl) methane in mouse and dog: implications for therapeutic development. Toxicol Res (Camb) 2024; 13:tfae059. [PMID: 38655145 PMCID: PMC11033559 DOI: 10.1093/toxres/tfae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/26/2024] Open
Abstract
The modified phytochemical derivative, 1,1-bis(3'-indolyl)-1-(p-chlorophenyl) methane (C-DIM12), has been identified as a potential therapeutic platform based on its capacity to improve disease outcomes in models of neurodegeneration and cancer. However, comprehensive safety studies investigating pathology and off-target binding have not been conducted. To address this, we administered C-DIM12 orogastrically to outbred male CD-1 mice for 7 days (50 mg/kg/day, 200 mg/kg/day, and 300 mg/kg/day) and investigated changes in hematology, clinical chemistry, and whole-body tissue pathology. We also delivered a single dose of C-DIM12 (1 mg/kg, 5 mg/kg, 25 mg/kg, 100 mg/kg, 300 mg/kg, 1,000 mg/kg) orogastrically to male and female beagle dogs and investigated hematology and clinical chemistry, as well as plasma pharmacokinetics over 48-h. Consecutive in-vitro off-target binding through inhibition was performed with 10 μM C-DIM12 against 68 targets in tandem with predictive off-target structural binding capacity. These data show that the highest dose C-DIM12 administered in each species caused modest liver pathology in mouse and dog, whereas lower doses were unremarkable. Off-target screening and predictive modeling of C-DIM12 show inhibition of serine/threonine kinases, calcium signaling, G-protein coupled receptors, extracellular matrix degradation, and vascular and transcriptional regulation pathways. Collectively, these data demonstrate that low doses of C-DIM12 do not induce pathology and are capable of modulating targets relevant to neurodegeneration and cancer.
Collapse
Affiliation(s)
- Savannah M Rocha
- Department of Environmental and Radiological Health Sciences, Colorado State University, 1680 Campus Delivery Fort Collins, CO 80523, USA
| | - Daniel L Gustafson
- Department of Clinical Sciences, Colorado State University, 1678 Campus Delivery Fort Collins, CO 80523, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M School of Veterinary, Medicine & Biomedical Sciences, 4466 TAMU College Station, TX 77843-4466, USA
| | - Ronald B Tjalkens
- Department of Environmental and Radiological Health Sciences, Colorado State University, 1680 Campus Delivery Fort Collins, CO 80523, USA
| |
Collapse
|
2
|
Chen JF, Wang J, Chai J, Jin W, Ren QL, Ma Q, Lu QX, Sun JJ, Mo DL, Zhang JQ, Xing BS. Transcriptome profiling of longissimus dorsi during different prenatal stages to identify genes involved in intramuscular fat deposition in lean and obese pig breeds. Mol Biol Rep 2024; 51:386. [PMID: 38441676 PMCID: PMC10914898 DOI: 10.1007/s11033-023-09088-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 11/29/2023] [Indexed: 03/07/2024]
Abstract
BACKGROUND There was significant difference in muscle development between fat-type and lean-type pig breeds. METHODS AND RESULTS In current study, transcriptome analysis and bioinformatics analysis were used to compare the difference in longissimus dorsi (LD) muscle at three time-points (38 days post coitus (dpc), 58 dpc, and 78 dpc ) between Huainan (HN) and Large white (LW) pig breeds. A total of 24500 transcripts were obtained in 18 samples, and 2319, 2799, and 3713 differently expressed genes (DEGs) were identified between these two breeds at 38 dpc, 58 dpc, and 78 dpc, respectively. And the number and foldchange of DEGs were increased, the alternative splice also increased. The cluster analysis of DEGs indicated the embryonic development progress of LD muscle between these two breeds was different. There were 539 shared DEGs between HN and LW at three stages, and the top-shared DEGs were associated with muscle development and lipid deposition, such as KLF4, NR4A1, HSP70, ZBTB16 and so on. CONCLUSIONS The results showed DEGs between Huainan (HN) and Large white (LW) pig breeds, and contributed to the understanding the muscle development difference between HN and LW, and provided basic materials for improvement of meat quality.
Collapse
Affiliation(s)
- Jun Feng Chen
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Huayuan Road No.116, Zhengzhou, 450002, Henan, China
| | - Jing Wang
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Huayuan Road No.116, Zhengzhou, 450002, Henan, China
| | - Jin Chai
- Agricultural Ministry Key Laboratory of Swine Breeding and Genetics & Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wei Jin
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Huayuan Road No.116, Zhengzhou, 450002, Henan, China
| | - Qiao Ling Ren
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Huayuan Road No.116, Zhengzhou, 450002, Henan, China
| | - Qiang Ma
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Huayuan Road No.116, Zhengzhou, 450002, Henan, China
| | - Qing Xia Lu
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Huayuan Road No.116, Zhengzhou, 450002, Henan, China
| | - Jia Jie Sun
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - De Lin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jia Qing Zhang
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Huayuan Road No.116, Zhengzhou, 450002, Henan, China
| | - Bao Song Xing
- Henan Key Laboratory of Farm Animal Breeding and Nutritional Regulation, Institute of Animal Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Huayuan Road No.116, Zhengzhou, 450002, Henan, China.
| |
Collapse
|
3
|
Zhang L, Martin G, Mohankumar K, Wright GA, Mariyam F, Safe S. Piperlongumine is a ligand for the orphan nuclear receptor 4A1 (NR4A1). Front Pharmacol 2023; 14:1223153. [PMID: 37808182 PMCID: PMC10551445 DOI: 10.3389/fphar.2023.1223153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/04/2023] [Indexed: 10/10/2023] Open
Abstract
Piperlongumine and derivatives are being developed as anticancer agents which act primarily as inducers of reactive oxygen species (ROS) in cancer cell lines. Many of the anticancer activities of piperlongumine resemble those observed for bis-indole derived compounds that bind the orphan nuclear receptor 4A1 (NR4A1) and act as inverse receptor agonists to inhibit NR4A1-regulated pro-oncogenic pathways and genes. In this study we show that like other NR4A1 inverse agonists piperlongumine inhibited RKO, SW480 and HCT116 colon cancer cell growth migration and invasion and induced apoptosis. Piperlongumine also downregulated the pro-reductant isocitrate dehydrogenase 1 (IDH1) and thioredoxin domain-containing 5 (TXNDC5) gene products resulting in the induction of ROS as previously observed for other inverse NR4A1 agonists. ROS also induced sestrin2 and this resulted in activation of AMPK phosphorylation and inhibition of mTOR pathway signaling. It has previously been reported that these pathways/genes are also regulated by inverse NR4A1 agonists or by knockdown of NR4A1. We also observed that piperlongumine directly bound NR4A1, inhibited NR4A1-dependent transactivation and interactions of the NR4A1/Sp1 complex bound to the GC-rich promoter of the NR4A1-regulated G9a gene.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, United States
| | - Greg Martin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, United States
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, United States
| | - Gus A. Wright
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Fuada Mariyam
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, United States
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
4
|
Zhang L, Mohankumar K, Martin G, Mariyam F, Park Y, Han SJ, Safe S. Flavonoids Quercetin and Kaempferol Are NR4A1 Antagonists and Suppress Endometriosis in Female Mice. Endocrinology 2023; 164:bqad133. [PMID: 37652054 PMCID: PMC10502789 DOI: 10.1210/endocr/bqad133] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023]
Abstract
Nuclear receptor 4A1 (NR4A1) plays an important role in endometriosis progression; levels of NR4A1 in endometriotic lesions are higher than in normal endometrium, and substituted bis-indole analogs (NR4A1) antagonists suppress endometriosis progression in mice with endometriosis. In addition, the flavonoids kaempferol and quercetin are natural products that directly bind NR4A1 and significantly repress the intrinsic NR4A1-dependent transcriptional activity in human endometriotic epithelial and stromal cells and Ishikawa endometrial cancer cells. NR4A1 knockdown and inhibition of NR4A1 by kaempferol and quercetin suppressed proliferation of human endometriotic epithelial cells and Ishikawa cells by inhibiting epidermal growth factor receptor/c-Myc/survivin-mediated growth-promoting and survival pathways, The mammalian target of rapamycin (mTOR) signaling and αSMA/CTGF/COL1A1/FN-mediated fibrosis signaling but increasing Thioredoxin domain Containing 5/SESN2-mediated oxidative/estrogen receptors stress signaling. In human endometriotic stromal cells, NR4A1 knockdown and inhibition of NR4A1 by kaempferol and quercetin primarily inhibited mTOR signaling by suppressing proliferation of human endometrial stromal cells. In addition, kaempferol and quercetin treatment also effectively suppressed the growth of endometriotic lesions in mice with endometriosis compared with the vehicle without any body weight changes. Therefore, kaempferol and quercetin are NR4A1 antagonists with potential as nutritional therapy for endometriosis.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Gregory Martin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Fuada Mariyam
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Yuri Park
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sang Jun Han
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
5
|
Wang H, Zhang M, Fang F, Xu C, Liu J, Gao L, Zhao C, Wang Z, Zhong Y, Wang X. The nuclear receptor subfamily 4 group A1 in human disease. Biochem Cell Biol 2023; 101:148-159. [PMID: 36861809 DOI: 10.1139/bcb-2022-0331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Nuclear receptor 4A1 (NR4A1), a member of the NR4A subfamily, acts as a gene regulator in a wide range of signaling pathways and responses to human diseases. Here, we provide a brief overview of the current functions of NR4A1 in human diseases and the factors involved in its function. A deeper understanding of these mechanisms can potentially improve drug development and disease therapy.
Collapse
Affiliation(s)
- Hongshuang Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Mengjuan Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Fang Fang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Chang Xu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Jiazhi Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Lanjun Gao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Chenchen Zhao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050091, China
| | - Zheng Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China.,Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yan Zhong
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China.,Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xiangting Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns Research, Shijiazhuang 050091, China
| |
Collapse
|
6
|
Deng S, Chen B, Huo J, Liu X. Therapeutic potential of NR4A1 in cancer: Focus on metabolism. Front Oncol 2022; 12:972984. [PMID: 36052242 PMCID: PMC9424640 DOI: 10.3389/fonc.2022.972984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Metabolic reprogramming is a vital hallmark of cancer, and it provides the necessary energy and biological materials to support the continuous proliferation and survival of tumor cells. NR4A1 is belonging to nuclear subfamily 4 (NR4A) receptors. NR4A1 plays diverse roles in many tumors, including melanoma, colorectal cancer, breast cancer, and hepatocellular cancer, to regulate cell growth, apoptosis, metastasis. Recent reports shown that NR4A1 exhibits unique metabolic regulating effects in cancers. This receptor was first found to mediate glycolysis via key enzymes glucose transporters (GLUTs), hexokinase 2 (HK2), fructose phosphate kinase (PFK), and pyruvate kinase (PK). Then its functions extended to fatty acid synthesis by modulating CD36, fatty acid-binding proteins (FABPs), sterol regulatory element-binding protein 1 (SREBP1), glutamine by Myc, mammalian target of rapamycin (mTOR), and hypoxia-inducible factors alpha (HIF-1α), respectively. In addition, NR4A1 is involving in amino acid metabolism and tumor immunity by metabolic processes. More and more NR4A1 ligands are found to participate in tumor metabolic reprogramming, suggesting that regulating NR4A1 by novel ligands is a promising approach to alter metabolism signaling pathways in cancer therapy. Basic on this, this review highlighted the diverse metabolic roles of NR4A1 in cancers, which provides vital references for the clinical application.
Collapse
Affiliation(s)
- Shan Deng
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bo Chen
- Materials Science and Devices Institute, Suzhou University of Science and Technology, Suzhou, China
| | - Jiege Huo
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Xin Liu, ; Jiege Huo,
| | - Xin Liu
- Third School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Orthopedics, Nanjing Lishui Hospital of Traditional Chinese Medicine, Nanjing, China
- *Correspondence: Xin Liu, ; Jiege Huo,
| |
Collapse
|
7
|
Xu Y, Tian J, Kang Q, Yuan H, Liu C, Li Z, Liu J, Li M. Knockout of Nur77 Leads to Amino Acid, Lipid, and Glucose Metabolism Disorders in Zebrafish. Front Endocrinol (Lausanne) 2022; 13:864631. [PMID: 35547009 PMCID: PMC9084189 DOI: 10.3389/fendo.2022.864631] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/14/2022] [Indexed: 12/25/2022] Open
Abstract
Orphan nuclear receptor Nur77 has been reported to be implicated in a diverse range of metabolic processes, including carbohydrate metabolism and lipid metabolism. However, the detailed mechanism of Nur77 in the regulation of metabolic pathway still needs to be further investigated. In this study, we created a global nur77 knockout zebrafish model by CRISPR/Cas9 technique, and then performed whole-organism RNA sequencing analysis in wildtype and nur77-deficient zebrafish to dissect the genetic changes in metabolic-related pathways. We found that many genes involved in amino acid, lipid, and carbohydrate metabolism changed by more than twofold. Furthermore, we revealed that nur77-/- mutant displayed increased total cholesterol (TC) and triglyceride (TG), alteration in total amino acids, as well as elevated glucose. We also demonstrated that the elevated glucose was not due to the change of glucose uptake but was likely caused by the disorder of glycolysis/gluconeogenesis and the impaired β-cell function, including downregulated insb expression, reduced β-cell mass, and suppressed insulin secretion. Importantly, we also verified that targeted expression of Nur77 in the β cells is sufficient to rescue the β-cell defects in global nur77-/- larvae zebrafish. These results provide new information about the global metabolic network that Nur77 signaling regulates, as well as the role of Nur77 in β-cell function.
Collapse
Affiliation(s)
- Yang Xu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Juanjuan Tian
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Qi Kang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Hang Yuan
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Chengdong Liu
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao, China
| | - Zhehui Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Jie Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
- *Correspondence: Mingyu Li, ; Jie Liu,
| | - Mingyu Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
- *Correspondence: Mingyu Li, ; Jie Liu,
| |
Collapse
|
8
|
Safe S, Shrestha R, Mohankumar K. Orphan nuclear receptor 4A1 (NR4A1) and novel ligands. Essays Biochem 2021; 65:877-886. [PMID: 34096590 PMCID: PMC11410023 DOI: 10.1042/ebc20200164] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/20/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022]
Abstract
The nuclear receptor (NR) superfamily of transcription factors encodes expression of 48 human genes that are important for maintaining cellular homeostasis and in pathophysiology, and this has been observed for all sub-families including orphan receptors for which endogenous ligands have not yet been identified. The orphan NR4A1 (Nur77 and TR3) and other members of this sub-family (NR4A2 and NR4A3) are immediate early genes induced by diverse stressors, and these receptors play an important role in the immune function and are up-regulated in some inflammatory diseases including solid tumors. Although endogenous ligands for NR4A have not been identified, several different classes of compounds have been characterized as NR4A1 ligands that bind the receptor. These compounds include cytosporone B and structurally related analogs, bis-indole derived (CDIM) compounds, the triterpenoid celastrol and a number of other chemicals including polyunsaturated fatty acids. NR4A1 ligands bind different regions/surfaces of NR4A1 and exhibit selective NR4A1 modulator (SNR4AM) activities that are dependent on ligand structure and cell/tissue context. NR4A1 ligands exhibit pharmacologic activities in studies on cancer, endometriosis metabolic and inflammatory diseases and are promising agents with clinical potential for treating multiple diseases.
Collapse
Affiliation(s)
- Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, U.S.A
| | - Rupesh Shrestha
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, U.S.A
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, U.S.A
| |
Collapse
|
9
|
Venu VKP, Saifeddine M, Mihara K, Faiza M, Gorobets E, Flewelling AJ, Derksen DJ, Hirota SA, Marei I, Al-Majid D, Motahhary M, Ding H, Triggle CR, Hollenberg MD. Metformin Prevents Hyperglycemia-Associated, Oxidative Stress-Induced Vascular Endothelial Dysfunction: Essential Role for the Orphan Nuclear Receptor Human Nuclear Receptor 4A1 (Nur77). Mol Pharmacol 2021; 100:428-455. [PMID: 34452975 DOI: 10.1124/molpharm.120.000148] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 08/17/2021] [Indexed: 01/22/2023] Open
Abstract
Vascular pathology is increased in diabetes because of reactive-oxygen-species (ROS)-induced endothelial cell damage. We found that in vitro and in a streptozotocin diabetes model in vivo, metformin at diabetes-therapeutic concentrations (1-50 µM) protects tissue-intact and cultured vascular endothelial cells from hyperglycemia/ROS-induced dysfunction typified by reduced agonist-stimulated endothelium-dependent, nitric oxide-mediated vasorelaxation in response to muscarinic or proteinase-activated-receptor 2 agonists. Metformin not only attenuated hyperglycemia-induced ROS production in aorta-derived endothelial cell cultures but also prevented hyperglycemia-induced endothelial mitochondrial dysfunction (reduced oxygen consumption rate). These endothelium-protective effects of metformin were absent in orphan-nuclear-receptor Nr4a1-null murine aorta tissues in accord with our observing a direct metformin-Nr4a1 interaction. Using in silico modeling of metformin-NR4A1 interactions, Nr4a1-mutagenesis, and a transfected human embryonic kidney 293T cell functional assay for metformin-activated Nr4a1, we identified two Nr4a1 prolines, P505/P549 (mouse sequences corresponding to human P501/P546), as key residues for enabling metformin to affect mitochondrial function. Our data indicate a critical role for Nr4a1 in metformin's endothelial-protective effects observed at micromolar concentrations, which activate AMPKinase but do not affect mitochondrial complex-I or complex-III oxygen consumption rates, as does 0.5 mM metformin. Thus, therapeutic metformin concentrations requiring the expression of Nr4a1 protect the vasculature from hyperglycemia-induced dysfunction in addition to metformin's action to enhance insulin action in patients with diabetes. SIGNIFICANCE STATEMENT: Metformin improves diabetic vasodilator function, having cardioprotective effects beyond glycemic control, but its mechanism to do so is unknown. We found that metformin at therapeutic concentrations (1-50µM) prevents hyperglycemia-induced endothelial dysfunction by attenuating reactive oxygen species-induced damage, whereas high metformin (>250 µM) impairs vascular function. However, metformin's action requires the expression of the orphan nuclear receptor NR4A1/Nur77. Our data reveal a novel mechanism whereby metformin preserves diabetic vascular endothelial function, with implications for developing new metformin-related therapeutic agents.
Collapse
Affiliation(s)
- Vivek Krishna Pulakazhi Venu
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Mahmoud Saifeddine
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Koichiro Mihara
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Muniba Faiza
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Evgueni Gorobets
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Andrew J Flewelling
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Darren J Derksen
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Simon A Hirota
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Isra Marei
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Dana Al-Majid
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Majid Motahhary
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Hong Ding
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Chris R Triggle
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| | - Morley D Hollenberg
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology (V.K.P.V, M.S., K.M., M.M., S.A.H., M.D.H.), and Department of Medicine (M.D.H.), University of Calgary Cumming School of Medicine, Calgary AB, Canada; Alberta Children's Hospital Research Institute and Department of Chemistry, University of Calgary AB, Canada (E.G., A.J.F., D.D.); Departments of Pharmacology and Medical Education, Weill Cornell Medicine in Qatar, Al-Rayyan, Doha, Qatar (I. M., D. A-M., H.D., C.R.T.) and Bioinformatics (M.F.), Jamia Millia Islamia (Central University), Jaima Nagar, Okhla New Delhi, India
| |
Collapse
|
10
|
Mohammed I, Hollenberg MD, Ding H, Triggle CR. A Critical Review of the Evidence That Metformin Is a Putative Anti-Aging Drug That Enhances Healthspan and Extends Lifespan. Front Endocrinol (Lausanne) 2021; 12:718942. [PMID: 34421827 PMCID: PMC8374068 DOI: 10.3389/fendo.2021.718942] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/15/2021] [Indexed: 12/11/2022] Open
Abstract
The numerous beneficial health outcomes associated with the use of metformin to treat patients with type 2 diabetes (T2DM), together with data from pre-clinical studies in animals including the nematode, C. elegans, and mice have prompted investigations into whether metformin has therapeutic utility as an anti-aging drug that may also extend lifespan. Indeed, clinical trials, including the MILES (Metformin In Longevity Study) and TAME (Targeting Aging with Metformin), have been designed to assess the potential benefits of metformin as an anti-aging drug. Preliminary analysis of results from MILES indicate that metformin may induce anti-aging transcriptional changes; however it remains controversial as to whether metformin is protective in those subjects free of disease. Furthermore, despite clinical use for over 60 years as an anti-diabetic drug, the cellular mechanisms by which metformin exerts either its actions remain unclear. In this review, we have critically evaluated the literature that has investigated the effects of metformin on aging, healthspan and lifespan in humans as well as other species. In preparing this review, particular attention has been placed on the strength and reproducibility of data and quality of the study protocols with respect to the pharmacokinetic and pharmacodynamic properties of metformin. We conclude that despite data in support of anti-aging benefits, the evidence that metformin increases lifespan remains controversial. However, via its ability to reduce early mortality associated with various diseases, including diabetes, cardiovascular disease, cognitive decline and cancer, metformin can improve healthspan thereby extending the period of life spent in good health. Based on the available evidence we conclude that the beneficial effects of metformin on aging and healthspan are primarily indirect via its effects on cellular metabolism and result from its anti-hyperglycemic action, enhancing insulin sensitivity, reduction of oxidative stress and protective effects on the endothelium and vascular function.
Collapse
Affiliation(s)
- Ibrahim Mohammed
- Department of Medical Education, Weill Cornell Medicine-Qatar, Al-Rayyan, Qatar
- *Correspondence: Chris R. Triggle, ; Ibrahim Mohammed,
| | - Morley D. Hollenberg
- Inflammation Research Network and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
- Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Hong Ding
- Department of Medical Education, Weill Cornell Medicine-Qatar, Al-Rayyan, Qatar
- Departments of Medical Education and Pharmacology, Weill Cornell Medicine-Qatar, Al-Rayyan, Qatar
| | - Chris R. Triggle
- Department of Medical Education, Weill Cornell Medicine-Qatar, Al-Rayyan, Qatar
- Departments of Medical Education and Pharmacology, Weill Cornell Medicine-Qatar, Al-Rayyan, Qatar
- *Correspondence: Chris R. Triggle, ; Ibrahim Mohammed,
| |
Collapse
|
11
|
Shrestha R, Mohankumar K, Jin UH, Martin G, Safe S. The Histone Methyltransferase Gene G9A Is Regulated by Nuclear Receptor 4A1 in Alveolar Rhabdomyosarcoma Cells. Mol Cancer Ther 2020; 20:612-622. [PMID: 33277444 DOI: 10.1158/1535-7163.mct-20-0474] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/09/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022]
Abstract
The histone methyltransferase G9A (EHMT2) gene catalyzes methylation of histone 3 lysine 9 (H3K9), and this gene silencing activity contributes to the tumor promoter-like activity of G9A in several tumor types including alveolar rhabdomyosarcoma (ARMS). Previous studies show the orphan nuclear receptor 4A1 (NR4A1, Nur77) is overexpressed in rhabdomyosarcoma and exhibits pro-oncogenic activity. In this study, we show that knockdown of NR4A1 in ARMS cells decreased expression of G9A mRNA and protein. Moreover, treatment of ARMS cells with several bis-indole-derived NR4A1 ligands (antagonists) including 1,1-bis(3'-indolyl)-1-(4-hydroxyphenyl)methane (CDIM8), 3,5-dimethyl (3,5-(CH3)2), and 3-bromo-5-methoxy (3-Br-5-OCH3) analogs also decreased G9A expression. Furthermore, NR4A1 antagonists also decreased G9A expression in breast, lung, liver, and endometrial cancer cells confirming that G9A is an NR4A1-regulated gene in ARMS and other cancer cell lines. Mechanistic studies showed that the NR4A1/Sp1 complex interacted with the GC-rich 511 region of the G9A promoter to regulate G9A gene expression. Moreover, knockdown of NR4A1 or treatment with NR4A1 receptor antagonists decreased overall H3K9me2, H3K9me2 associated with the PTEN promoter, and PTEN-regulated phospho-Akt. In vivo studies showed that the NR4A1 antagonist (3-Br-5-OCH3) inhibited tumor growth in athymic nude mice bearing Rh30 ARMS cells and confirmed that G9A was an NR4A1-regulated gene that can be targeted by NR4A1 receptor antagonists.
Collapse
Affiliation(s)
- Rupesh Shrestha
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Gregory Martin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Stephen Safe
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas. .,Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| |
Collapse
|
12
|
Mohankumar K, Li X, Sung N, Cho YJ, Han SJ, Safe S. Bis-Indole-Derived Nuclear Receptor 4A1 (NR4A1, Nur77) Ligands as Inhibitors of Endometriosis. Endocrinology 2020; 161:5758064. [PMID: 32099996 PMCID: PMC7105386 DOI: 10.1210/endocr/bqaa027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023]
Abstract
Endometriosis is an inflammatory disease that primarily affects women during their reproductive years, and since current hormonal therapies are of concern, new hormone-independent treatment regimens are needed. The orphan nuclear receptor 4A1 (NR4A1, Nur77) is expressed in patient-derived (stromal) endometriotic cells and also epithelial cell lines, and we observed that knockdown of NR4A1 in patient-derived ectopic endometrium-isolated ovarian endometrioma (ESECT)-7 and ESECT-40 cells decreased cell proliferation and induced apoptosis. Moreover, the treatment of these cells with bis-indole derived NR4A1 ligands 1,1-bis(3'-indolyl)-1-(p-hydroxyphenyl)methane (DIM-C-pPhOH) and its buttressed 3-chloro-5-methoxy analog (DIM-C-pPhOH-3-Cl-5-OCH3) inhibited cell growth and induced apoptosis and related genes. The compounds exhibit NR4A1 antagonist activities in both functional and transactivation assays whereas these effects were not observed in normal endometrial cells. We also observed that NR4A1 knockdown and treatment with NR4A1 antagonists decreased fibrosis, α-smooth muscle actin, and related pro-fibrotic genes in ESECT-7 and ESECT-40 cells, and similar results were observed in epithelial-derived endometriotic cell lines. Moreover, in an endometriosis mouse model with auto-transplantation and also in severe combined immune deficiency mice transplanted with human endometriotic cells treatment with 25 mg/kg/day DIM-C-pPhOH-3-Cl-5-OCH3 significantly inhibited growth and expansion of endometriotic lesions. Thus, bis-indole-derived NR4A1 ligands represent a novel class of drugs as nonhormonal therapy for endometriosis.
Collapse
Affiliation(s)
- Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX US
| | - Xi Li
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX US
| | - Nuri Sung
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX US
| | - Yeon Jean Cho
- Department of Obstetrics and Gynecology, Dong-A University, College of Medicine, Busan, Republic of Korea
| | - Sang Jun Han
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX US
- Correspondence: Stephen Safe, Department of Veterinary Physiology & Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466. E-mail: ; or Sang Jun Han, Department of molecular and Cell Biology, Baylor college of Medicine, Houston, TX 77030. E-mail:
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX US
- Correspondence: Stephen Safe, Department of Veterinary Physiology & Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466. E-mail: ; or Sang Jun Han, Department of molecular and Cell Biology, Baylor college of Medicine, Houston, TX 77030. E-mail:
| |
Collapse
|
13
|
Ming Y, Yin Y, Sun Z. Interaction of Nuclear Receptor Subfamily 4 Group A Member 1 (Nr4a1) and Liver Linase B1 (LKB1) Mitigates Type 2 Diabetes Mellitus by Activating Monophosphate-Activated Protein Kinase (AMPK)/Sirtuin 1 (SIRT1) Axis and Inhibiting Nuclear Factor-kappa B (NF-κB) Activation. Med Sci Monit 2020; 26:e920278. [PMID: 31939452 PMCID: PMC6982402 DOI: 10.12659/msm.920278] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Nuclear receptor subfamily 4 group A member 1 (Nr4a1) has been increasingly investigated in association with type 2 diabetes mellitus (T2DM). This study aimed to explore its efficacy with liver kinase B1 (LKB1) and potential signaling pathways in T2DM. MATERIAL AND METHODS A T2DM model in rats was established by high-fat diet and injection of 30 mg/kg streptozotocin. The ectopic expression of Nr4a1 or in combination with LKB1 was performed in T2DM rats to probe their effects on T2DM. Then, the weight and indicators of blood lipid and blood glucose in normal rats and T2DM rats were measured. The volume change of adipocytes and the size of lipid droplets in white adipose tissue (WAT) were observed by hematoxylin-eosin staining and oil red O staining, respectively. We also measured levels of Nr4a1, LKB1, and adenosine monophosphate-activated protein kinase (AMPK)/sirtuin 1 (SIRT1)/Nuclear factor-kappa B (NF-kappaB) axis-related proteins. RESULTS In T2DM rats, Nr4a1 was highly expressed, and body weight, blood lipid and blood glucose were increased, and the volume of adipocytes and the size of lipid droplets in WAT were increased, which were all reversed by low expression of Nr4a1. After treatment with Nr4a1 and LKB1 together, T2DM rats showed decreased levels of blood lipid, blood glucose, and reduced volume of adipocytes and lipid droplet size in WAT, with activated AMPK/SIRT1 signaling pathway and inhibited NF-kappaB. CONCLUSIONS Our results highlight that interaction of Nr4a1 and LKB1 can mitigate T2DM by activating the AMPK/SIRT1 signaling pathway and inhibiting NF-kappaB activation. This may offer new insight for T2DM treatment.
Collapse
Affiliation(s)
- Yi Ming
- Department of Endocrinology, Weifang People's Hospital, Weifang, Shandong, China (mainland)
| | - Youmin Yin
- Department of Endocrinology, Weifang People's Hospital, Weifang, Shandong, China (mainland)
| | - Zhaoli Sun
- Department of Endocrinology, Weifang People's Hospital, Weifang, Shandong, China (mainland)
| |
Collapse
|
14
|
Karki K, Wright GA, Mohankumar K, Jin UH, Zhang XH, Safe S. A Bis-Indole-Derived NR4A1 Antagonist Induces PD-L1 Degradation and Enhances Antitumor Immunity. Cancer Res 2020; 80:1011-1023. [PMID: 31911554 DOI: 10.1158/0008-5472.can-19-2314] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/20/2019] [Accepted: 01/03/2020] [Indexed: 12/31/2022]
Abstract
PD-L1 is expressed in tumor cells and its interaction with PD-1 plays an important role in evading immune surveillance; this can be overcome using PD-L1 or PD-1 immunotherapy antibodies. This study reports a novel approach for targeting PD-L1. In human breast cancer cell lines and 4T1 mouse mammary tumor cells, PD-L1 expression was regulated by the nuclear receptor NR4A1/Sp1 complex bound to the proximal germinal center (GC)-rich region of the PD-L1 gene promoter. Treatment of breast cancer cells with bis-indole-derived NR4A1 antagonists including 1,1-bis(3'-indolyl)-1-(3-chloro-4-hydroxy-5-methoxyphenyl)methane (Cl-OCH3) decreased expression of PD-L1 mRNA, promoter-dependent luciferase activity, and protein. In in vivo studies using a syngeneic mouse model bearing orthotopically injected 4T1 cells, Cl-OCH3 decreased tumor growth and weight and inhibited lung metastasis. Cl-OCH3 also decreased expression of CD3+/CD4+/CD25+/FoxP3+ regulatory T cells and increased the Teff/Treg ratio. Therefore, the potent anticancer activities of NR4A1 antagonists are also accompanied by enhanced antitumor immunity in PD-L1-expressing triple-negative breast cancer and thus represent a novel class of drugs that mimic immunotherapy. SIGNIFICANCE: These findings show that the orphan nuclear receptor NR4A1 controls PD-L1 expression and identify a chemical probe capable of disrupting this regulatory axis.
Collapse
Affiliation(s)
- Keshav Karki
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Gus A Wright
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Xing-Han Zhang
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas.
| |
Collapse
|
15
|
Function of Nr4a Orphan Nuclear Receptors in Proliferation, Apoptosis and Fuel Utilization Across Tissues. Cells 2019; 8:cells8111373. [PMID: 31683815 PMCID: PMC6912296 DOI: 10.3390/cells8111373] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/24/2019] [Accepted: 10/30/2019] [Indexed: 12/21/2022] Open
Abstract
The Nr4a family of nuclear hormone receptors is composed of three members-Nr4a1/Nur77, Nr4a2/Nurr1 and Nr4a3/Nor1. While currently defined as ligandless, these transcription factors have been shown to regulate varied processes across a host of tissues. Of particular interest, the Nr4a family impinge, in a tissue dependent fashion, on cellular proliferation, apoptosis and fuel utilization. The regulation of these processes occurs through both nuclear and non-genomic pathways. The purpose of this review is to provide a balanced perspective of the tissue specific and Nr4a family member specific, effects on cellular proliferation, apoptosis and fuel utilization.
Collapse
|
16
|
Hedrick E, Li X, Cheng Y, Lacey A, Mohankumar K, Zarei M, Safe S. Potent inhibition of breast cancer by bis-indole-derived nuclear receptor 4A1 (NR4A1) antagonists. Breast Cancer Res Treat 2019; 177:29-40. [PMID: 31119568 PMCID: PMC6681651 DOI: 10.1007/s10549-019-05279-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/13/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND Nuclear receptor 4A1 (NR4A1) is overexpressed in mammary tumors, and the methylene-substituted bis-indole derivative 1,1-bis(3'-indolyl)-1-(p-hydroxyphenyl)methane (DIM-C-pPhOH) acts as an NR4A1 antagonist (inverse agonist) and inhibits NR4A1-regulated pro-oncogenic pathways/genes in breast and other cancer cells. METHODS Buttressed analogs of DIM-C-pPhOH were synthesized by condensation of the substituted p-hydroxybenzaldehydes with indole. Breast cancer cell growth, survival, and migration assays were carried out by cell counting, Annexin V staining, and Boyden chamber assays, respectively. Changes in RNA and protein expression were determined by RT-PCR and western blots, respectively. Analysis of RNAseq results was carried out using Ingenuity Pathway Analysis, and in vivo potencies of NR4A1 antagonists were determined in athymic nude mice bearing MDA-MB-231 cells in an orthotopic model. RESULTS Ingenuity Pathway analysis of common genes modulated by NR4A1 knockdown or treatment with DIM-C-pPhOH showed that changes in gene expression were consistent with the observed decreased functional responses, namely inhibition of growth and migration and increased apoptosis. DIM-C-pPhOH is rapidly metabolized and the effects and potencies of buttressed analogs of DIM-C-pPhOH which contain one or two substituents ortho to the hydroxyl groups were investigated using NR4A1-regulated gene/gene products as endpoints. The buttressed analogs were more potent than DIM-C-pPhOH in both in vitro assays and as inhibitors of mammary tumor growth. Moreover, using 1,1-bis(3'-indolyl)-1-(3-chloro-4-hydroxy-5-methoxyphenyl)methane (DIM-C-pPhOh-3-Cl-5-OCH3) significant tumor growth inhibition was observed at doses as low as 2 mg/kg/d which was at least an order of magnitude more potent than DIM-C-pPhOH. CONCLUSIONS These buttressed analogs represent a more potent set of second generation NR4A1 antagonists as inhibitors of breast cancer.
Collapse
Affiliation(s)
- Erik Hedrick
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
- Cleveland Clinic, Lerner Research Institute, Cleveland, OH, 44195, USA
| | - Xi Li
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
| | - Yating Cheng
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
- MD Anderson Cancer Center, Houston, TX, 77030, USA
| | | | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
| | - Mahsa Zarei
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
17
|
Mohankumar K, Li X, Sridharan S, Karki K, Safe S. Nuclear receptor 4A1 (NR4A1) antagonists induce ROS-dependent inhibition of mTOR signaling in endometrial cancer. Gynecol Oncol 2019; 154:218-227. [PMID: 31053403 DOI: 10.1016/j.ygyno.2019.04.678] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/01/2019] [Accepted: 04/21/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVES NR4A1 is overexpressed in many solid tumors, and the objectives of this study were to investigate the expression and functional role of this receptor in endometrial cancer cells and demonstrate that NR4A1 antagonist inhibit mTOR. METHODS Ishikawa and Hec-1B endometrial cells were used as models to investigate the parallel effects of NR4A1 knockdown by RNA interference (siNR4A1) and treatment with bis-indole-derived NR4A1 ligands (antagonists) on cell growth and survival by determining cell numbers and effects on Annexin V staining. Western blot analysis of whole cell lysates was used to determine effects of these treatments on expression of growth promoting, survival and apoptotic genes and mTOR signaling. Effects of NR4A1 antagonists on tumor growth were determined in athymic nude mice bearing Hec-1B cells as xenografts. RESULTS siNR4A1 or treatment with bis-indole-derived NR4A1 antagonists inhibited growth of endometrial cancer cells in vitro and endometrial tumors in vivo and this was accompanied by decreased expression of growth promoting and survival genes and mTOR inhibition. CONCLUSIONS NR4A1 exhibited pro-oncogenic activity in endometrial cells due, in part, to regulation of cell growth, survival and mTOR signaling, and all of these pathways and their associated gene products were inhibited after treatment with bis-indole-derived NR4A1 antagonists. Moreover, these compounds also blocked endometrial tumor growth in vivo demonstrating that NR4A1 is a potential novel drug target for treatment of endometrial cancer.
Collapse
Affiliation(s)
- Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas AM University, College Station, TX 77843, USA
| | - Xi Li
- Department of Veterinary Physiology and Pharmacology, Texas AM University, College Station, TX 77843, USA
| | - Subhashree Sridharan
- Department of Medical Physiology, College of Medicine, Texas AM University, College Station, TX 77843, USA
| | - Keshav Karki
- Department of Veterinary Physiology and Pharmacology, Texas AM University, College Station, TX 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas AM University, College Station, TX 77843, USA.
| |
Collapse
|