1
|
Qian J, Guo Y, Khan B, Shi J, Hou Y. GW501516 facilitated tumor immune escape by inhibiting phagocytosis. Eur J Pharmacol 2025; 995:177418. [PMID: 39993702 DOI: 10.1016/j.ejphar.2025.177418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025]
Abstract
The CD47/SIRPα innate immune checkpoint plays a critical role in regulating tumor immune escape. GW501516, a peroxisome proliferator-activated receptor delta (PPARδ) agonist, is known to promote cancer cell metabolism, proliferation, and inflammation; however, its regulatory mechanism in colon tumor immune escape remains unclear. In this study, qPCR analysis revealed that GW501516 treatment upregulated CD47 gene expression in colon cancer cells. Additionally, GW501516 increased membrane-associated CD47 protein levels in these cells. Mechanistically, luciferase reporter assays demonstrated that GW501516 enhanced CD47 gene transcription activity in colon cancer cells. Co-culture experiments with macrophages further showed that GW501516 treatment suppressed macrophage phagocytic capacity. Crucially, PPARδ knockout abolished GW501516-induced CD47 expression, indicating PPARδ dependency. In vivo implanted tumor models demonstrated that GW501516 facilitated tumor immune escape, whereas PPARδ loss reversed this effect. Collectively, these findings suggest that GW501516 activates PPARδ to promote colon tumor immune escape via CD47 upregulation.
Collapse
Affiliation(s)
- Jing Qian
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China, 212013
| | - Yilei Guo
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China, 212013
| | - Bibimaryam Khan
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China, 212013
| | - Juanjuan Shi
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China, 212013
| | - Yongzhong Hou
- School of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China, 212013.
| |
Collapse
|
2
|
Wang C, Lv T, Jin B, Li Y, Fan Z. Regulatory role of PPAR in colorectal cancer. Cell Death Discov 2025; 11:28. [PMID: 39875357 PMCID: PMC11775197 DOI: 10.1038/s41420-025-02313-2] [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: 07/13/2024] [Revised: 12/11/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most common tumors in the digestive system, and the majority of patients are found to be in advanced stages, which is a burden to human health all over the world. Moreover, in recent years, CRC has been progressively becoming younger, with an increasing incidence mainly among patients <50 years old. Despite the increase in awareness of CRC and the continuous improvement of medical treatment nowadays, the challenge of CRC still needs to be conquered. By now, the pathogenesis of CRC is complex and not fully understood. With the deepening of research, it has been revealed that PPARs, as a transcription factor, are inextricably linked to CRC. This article outlines the mechanisms by which PPARs are involved in CRC development. An in-depth understanding of the pathways related to PPARs may provide new ways of developing effective therapies for CRC with PPARs as potential targets.
Collapse
Affiliation(s)
- Cong Wang
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
- Department of General Surgery, The Third People's Hospital of Dalian, Faculty of Medicine, Dalian University of Technology, Dalian, China
| | - Tingcong Lv
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Binghui Jin
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Yang Li
- Department of Breast Surgery, Cancer Hospital of China Medical University, Shenyang, China.
- Department of Breast Surgery, Liaoning Cancer Hospital & Institute, Shenyang, China.
| | - Zhe Fan
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China.
- Department of General Surgery, The Third People's Hospital of Dalian, Faculty of Medicine, Dalian University of Technology, Dalian, China.
| |
Collapse
|
3
|
Chong S, Woolnough CA, Koyyalamudi SR, Perera NJ. Reversible Gynecomastia and Hypogonadism Due to Usage of Commercial Performance-Enhancing Supplement Use. JCEM CASE REPORTS 2024; 2:luae148. [PMID: 39145153 PMCID: PMC11321837 DOI: 10.1210/jcemcr/luae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Indexed: 08/16/2024]
Abstract
Commercially available performance-enhancing supplements can contain banned performance-enhancing drugs (PEDs) and undisclosed steroid hormones that can induce hormonal abnormalities with associated clinical signs. We present a case of a 40-year-old male who developed bilateral gynecomastia and biochemical hypogonadotropic hypogonadism with a corresponding 6-month history of consuming commercially available performance-enhancing supplements for gym workouts. These performance-enhancing supplements were found to contain amounts of RAD-140, a selective androgen receptor modulator, MK-677, a GH secretagogue and cardarine, all of which are banned PEDs. In vitro analysis also detected undisclosed hormones testosterone, estradiol, and GH in all 3 supplements, with further steroid analysis using liquid chromatography mass spectrometry identifying an unidentified compound coeluting close to the testosterone peak. Cessation of these supplements led to full resolution of symptoms including normalization of hypogonadotropic hypogonadism. This case highlights the need for clinicians to consider commercially available performance-enhancing supplements as potential sources of PEDs and exogenous steroid hormones that can have adverse clinical consequences.
Collapse
Affiliation(s)
- Serena Chong
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia
| | - Catherine A Woolnough
- Department of Chemical Pathology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia
| | - Sundar R Koyyalamudi
- Institute of Endocrinology and Diabetes, The Children's Hospital, Westmead, New South Wales 2145, Australia
| | - Nimalie J Perera
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia
- Department of Chemical Pathology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia
- Sydney Medical School and Charles Perkins Centre, University of Sydney, Sydney, New South Wales 2050, Australia
| |
Collapse
|
4
|
Manickasamy MK, Jayaprakash S, Girisa S, Kumar A, Lam HY, Okina E, Eng H, Alqahtani MS, Abbas M, Sethi G, Kumar AP, Kunnumakkara AB. Delineating the role of nuclear receptors in colorectal cancer, a focused review. Discov Oncol 2024; 15:41. [PMID: 38372868 PMCID: PMC10876515 DOI: 10.1007/s12672-023-00808-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/20/2023] [Indexed: 02/20/2024] Open
Abstract
Colorectal cancer (CRC) stands as one of the most prevalent form of cancer globally, causing a significant number of deaths, surpassing 0.9 million in the year 2020. According to GLOBOCAN 2020, CRC ranks third in incidence and second in mortality in both males and females. Despite extensive studies over the years, there is still a need to establish novel therapeutic targets to enhance the patients' survival rate in CRC. Nuclear receptors (NRs) are ligand-activated transcription factors (TFs) that regulate numerous essential biological processes such as differentiation, development, physiology, reproduction, and cellular metabolism. Dysregulation and anomalous expression of different NRs has led to multiple alterations, such as impaired signaling cascades, mutations, and epigenetic changes, leading to various diseases, including cancer. It has been observed that differential expression of various NRs might lead to the initiation and progression of CRC, and are correlated with poor survival outcomes in CRC patients. Despite numerous studies on the mechanism and role of NRs in this cancer, it remains of significant scientific interest primarily due to the diverse functions that various NRs exhibit in regulating key hallmarks of this cancer. Thus, modulating the expression of NRs with their agonists and antagonists, based on their expression levels, holds an immense prospect in the diagnosis, prognosis, and therapeutical modalities of CRC. In this review, we primarily focus on the role and mechanism of NRs in the pathogenesis of CRC and emphasized the significance of targeting these NRs using a variety of agents, which may represent a novel and effective strategy for the prevention and treatment of this cancer.
Collapse
Affiliation(s)
- Mukesh Kumar Manickasamy
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Sujitha Jayaprakash
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Huiyan Eng
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, 61421, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, 61421, Abha, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore.
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
5
|
Li Y, Pan Y, Zhao X, Wu S, Li F, Wang Y, Liu B, Zhang Y, Gao X, Wang Y, Zhou H. Peroxisome proliferator-activated receptors: A key link between lipid metabolism and cancer progression. Clin Nutr 2024; 43:332-345. [PMID: 38142478 DOI: 10.1016/j.clnu.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023]
Abstract
Lipids represent the essential components of membranes, serve as fuels for high-energy processes, and play crucial roles in signaling and cellular function. One of the key hallmarks of cancer is the reprogramming of metabolic pathways, especially abnormal lipid metabolism. Alterations in lipid uptake, lipid desaturation, de novo lipogenesis, lipid droplets, and fatty acid oxidation in cancer cells all contribute to cell survival in a changing microenvironment by regulating feedforward oncogenic signals, key oncogenic functions, oxidative and other stresses, immune responses, or intercellular communication. Peroxisome proliferator-activated receptors (PPARs) are transcription factors activated by fatty acids and act as core lipid sensors involved in the regulation of lipid homeostasis and cell fate. In addition to regulating whole-body energy homeostasis in physiological states, PPARs play a key role in lipid metabolism in cancer, which is receiving increasing research attention, especially the fundamental molecular mechanisms and cancer therapies targeting PPARs. In this review, we discuss how cancer cells alter metabolic patterns and regulate lipid metabolism to promote their own survival and progression through PPARs. Finally, we discuss potential therapeutic strategies for targeting PPARs in cancer based on recent studies from the last five years.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yujie Pan
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Xin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
6
|
Sun G, Li J, Liu X, Liu Y, Wen X, Sun H, Xu QL. Organophosphorus-Catalyzed "Dual-Substrate Deoxygenation" Strategy for C-S Bond Formation from Sulfonyl Chlorides and Alcohols/Acids. J Org Chem 2023. [PMID: 37296496 DOI: 10.1021/acs.joc.3c00532] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
A green method to construct C-S bonds using sulfonyl chlorides and alcohols/acids via a PIII/PV═O catalytic system is reported. The organophosphorus-catalyzed umpolung reaction promotes us to propose the "dual-substrate deoxygenation" strategy. Herein, we adopt the "dual-substrate deoxygenation" strategy, which achieves the deoxygenation of sulfonyl chlorides and alcohols/acids to synthesize thioethers/thioesters driven by PIII/PV═O redox cycling. The catalytic method represents an operationally simple approach using stable phosphine oxide as a precatalyst and shows broad functional group tolerance. The potential application of this protocol is demonstrated by the late-stage diversification of drug analogues.
Collapse
Affiliation(s)
- Gang Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Jing Li
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Xin Liu
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Yiting Liu
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Xiaoan Wen
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Qing-Long Xu
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| |
Collapse
|
7
|
UZUN M, ILHAN YS, BOZDAG A, YILMAZ M, ARTAS G, KULOGLU T. Asprosin, irisin, and meteorin-like protein immunoreactivity in different stages of colorectal adenocarcinoma. Pathol Res Pract 2023; 245:154432. [PMID: 37019019 DOI: 10.1016/j.prp.2023.154432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/15/2023] [Accepted: 03/28/2023] [Indexed: 04/04/2023]
Abstract
OBJECTIVE In this study, we aimed to investigate the immunoreactivity of asprosin, irisin, and meteorin-like protein (METRNL) in different stages of colorectal adenocarcinoma, which is the most common malignancy of the gastrointestinal tract. MATERIALS AND METHODS Overall, 60 patients with colorectal adenocarcinoma, including 20 well (Group 1), moderately (Group 2), and poorly differentiated (Group 3) cases, respectively, and 20 with normal colonic mucosa, were examined using light microscopy for immunohistochemical staining of asprosin, METRNL, and irisin. RESULTS Compared with the control group, a significant increase in irisin and asprosin immunoreactivity was found in the grade 1 and 2 colorectal adenocarcinoma groups. Moreover, compared with the grade 1 and 2 groups, this immunoreactivity was significantly decreased in the grade 3 colorectal adenocarcinoma group. Although there was no significant difference in METRNL immunoreactivity between the grade 1 and control groups, a statistically significant increase in this immunoreactivity was found in the grade 2 group. In contrast, METRNL immunoreactivity was significantly decreased in the grade 3 group compared with the grade 2 group. CONCLUSION We found that in early-stage colorectal adenocarcinoma there was an increase in the immunoreactivity of asprosin and irisin, but in the advanced stage there was a decrease in immunoreactivity. Although METRNL immunoreactivity did not change in the control and grade 1 groups, it was found to increase significantly in the grade 2 group and decrease in the grade 3 group.
Collapse
|
8
|
Wagner N, Wagner KD. Peroxisome Proliferator-Activated Receptors and the Hallmarks of Cancer. Cells 2022; 11:cells11152432. [PMID: 35954274 PMCID: PMC9368267 DOI: 10.3390/cells11152432] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) function as nuclear transcription factors upon the binding of physiological or pharmacological ligands and heterodimerization with retinoic X receptors. Physiological ligands include fatty acids and fatty-acid-derived compounds with low specificity for the different PPAR subtypes (alpha, beta/delta, and gamma). For each of the PPAR subtypes, specific pharmacological agonists and antagonists, as well as pan-agonists, are available. In agreement with their natural ligands, PPARs are mainly focused on as targets for the treatment of metabolic syndrome and its associated complications. Nevertheless, many publications are available that implicate PPARs in malignancies. In several instances, they are controversial for very similar models. Thus, to better predict the potential use of PPAR modulators for personalized medicine in therapies against malignancies, it seems necessary and timely to review the three PPARs in relation to the didactic concept of cancer hallmark capabilities. We previously described the functions of PPAR beta/delta with respect to the cancer hallmarks and reviewed the implications of all PPARs in angiogenesis. Thus, the current review updates our knowledge on PPAR beta and the hallmarks of cancer and extends the concept to PPAR alpha and PPAR gamma.
Collapse
Affiliation(s)
- Nicole Wagner
- Correspondence: (N.W.); (K.-D.W.); Tel.: +33-489-153-713 (K.-D.W.)
| | | |
Collapse
|
9
|
PPARβ/δ Augments IL-1β-Induced COX-2 Expression and PGE2 Biosynthesis in Human Mesangial Cells via the Activation of SIRT1. Metabolites 2022; 12:metabo12070595. [PMID: 35888719 PMCID: PMC9320509 DOI: 10.3390/metabo12070595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/11/2022] [Accepted: 06/22/2022] [Indexed: 12/10/2022] Open
Abstract
Peroxisome proliferator-activated receptor β/δ (PPARβ/δ), a ligand-activated nuclear receptor, regulates lipid and glucose metabolism and inflammation. PPARβ/δ can exert an anti-inflammatory effect by suppressing proinflammatory cytokine production. Cyclooxygenase-2 (COX-2)-triggered inflammation plays a crucial role in the development of many inflammatory diseases, including glomerulonephritis. However, the effect of PPARβ/δ on the expression of COX-2 in the kidney has not been fully elucidated. The present study showed that PPARβ/δ was functionally expressed in human mesangial cells (hMCs), where its expression was increased by interleukin-1β (IL-1β) treatment concomitant with enhanced COX-2 expression and prostaglandin E2 (PGE2) biosynthesis. The treatment of hMCs with GW0742, a selective agonist of PPARβ/δ, or the overexpression of PPARβ/δ via an adenovirus-mediated approach significantly increased COX-2 expression and PGE2 production. PPARβ/δ could further augment the IL-1β-induced COX-2 expression and PGE2 production in hMCs. Moreover, both PPARβ/δ activation and overexpression markedly increased sirtuin 1 (SIRT1) expression. The inhibition or knockdown of SIRT1 significantly attenuated the effects of PPARβ/δ on the IL-1β-induced expression of COX-2 and PGE2 biosynthesis. Taken together, PPARβ/δ could augment the IL-1β-induced COX-2 expression and PGE2 production in hMCs via the SIRT1 pathway. Given the critical role of COX-2 in glomerulonephritis, PPARβ/δ may represent a novel target for the treatment of renal inflammatory diseases.
Collapse
|
10
|
Han W, Wang N, Kong R, Bao W, Lu J. Ligand-activated PPARδ expression promotes hepatocellular carcinoma progression by regulating the PI3K-AKT signaling pathway. J Transl Med 2022; 20:86. [PMID: 35151320 PMCID: PMC8840031 DOI: 10.1186/s12967-022-03288-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/31/2022] [Indexed: 11/22/2022] Open
Abstract
Background Peroxisome proliferator-activated receptor-beta/delta (PPARδ) was considered as the key regulator involved in the evolution of various tumors. Given that PPARδ potential role in hepatocellular carcinoma (HCC) is still obscure, we comprehensively assessed its expression pattern, prognosis, functions and correlation with tumor microenvironment in HCC using public database data and in vitro studies. Methods Transcriptional data and clinical data in the TCGA and GEO database were analyzed in R software. Quantitative real-time polymerase chain reaction (qRT-PCR), western blotting and immunohistochemistry were used to detect the expression level of related RNA and proteins. The malignant biological characteristics were explored by cell counting Kit-8 (CCK8), 5-Ethynyl-2ʹ-deoxyuridine (EdU) assay and wound healing assay. Results Our results illustrated that PPARδ expression was significantly higher in HCC tissues and HCC cell lines. Elevated expression of PPARδ suggested poor clinical staging and prognosis in HCC. Ligand-activated PPARδ expression promoted the proliferation and invasion of HCC cells via PDK1/AKT/GSK3β signaling pathway. The expression of PPARδ was closely related to the HCC tumor microenvironment. Conclusions PPARδ plays an important part in HCC progression, penetrating investigation of the related regulatory mechanism may shed light upon further biological and pharmacological value.
Collapse
|
11
|
Chen M, Lin W, Ye R, Yi J, Zhao Z. PPARβ/δ Agonist Alleviates Diabetic Osteoporosis via Regulating M1/M2 Macrophage Polarization. Front Cell Dev Biol 2021; 9:753194. [PMID: 34901001 PMCID: PMC8661472 DOI: 10.3389/fcell.2021.753194] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/05/2021] [Indexed: 02/05/2023] Open
Abstract
Diabetic osteoporosis is a common complication in diabetic patients, leading to increased fracture risk and impaired bone healing. As a member of the peroxisome proliferator-activated receptor (PPAR) family, PPARβ/δ agonist is suggested as a therapeutic target for the treatment of metabolic syndrome, and has been reported to positively regulate bone turnover by improving osteogenesis. However, its regulatory role in diabetic osteoporosis has not been reported yet. Here, we explored the therapeutic effects and potential mechanisms of PPARβ/δ agonist to the osteoporotic phenotypes of diabetic mice. Our results indicated that the osteoporotic phenotypes could be significantly ameliorated in diabetic mice by the administration of PPARβ/δ agonists. In vitro experiments suggested that PPARβ/δ agonist treatment could alleviate the abnormal increase of osteoclast activity in diabetic mice by rectifying high glucose-mediated macrophage dysfunction instead of directly inhibiting osteoclast differentiation. Mechanistically, Angptl4 may act as a downstream target of PPARβ/δ to regulate macrophage polarization. In conclusion, our study demonstrates the potential of PPARβ/δ agonist as a therapeutic target for the treatment of osteoporosis and immune homeostasis disorder in diabetic patients.
Collapse
Affiliation(s)
- Miao Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weimin Lin
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianru Yi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Liu L, Gao H, Wen T, Gu T, Zhang S, Yuan Z. Tanshinone IIA attenuates AOM/DSS-induced colorectal tumorigenesis in mice via inhibition of intestinal inflammation. PHARMACEUTICAL BIOLOGY 2021; 59:89-96. [PMID: 33535870 PMCID: PMC8871617 DOI: 10.1080/13880209.2020.1865412] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
CONTEXT Tanshinone IIA is a natural extract derived from a Chinese medicinal herb with multiple bioactivities; however, whether and how tanshinone IIA protects against colorectal cancer (CRC) are uncertain. OBJECTIVE We investigated the potential beneficial effects of tanshinone IIA in a colitis-associated colorectal tumorigenesis mouse model and its underlying mechanisms. MATERIALS AND METHODS Male C57BL/6 mice were treated with azoxymethane (AOM) 10 mg/kg body weight and dextran sulphate sodium (2.5% DSS) to induce a colitis-associated cancer model. Tanshinone IIA (200 mg/kg body weight) was given to the mice intraperitoneally. After 12 weeks, all mice were sacrificed to measure tumour formation, intestinal permeability, neutrophil infiltration, and colonic inflammation. In addition, whether tanshinone IIA has inhibitory effects on neutrophil activation was determined through in vitro investigations. RESULTS We observed that tanshinone IIA significantly decreased tumour formation in AOM/DSS-treated mice compared to AOM/DSS-treated alone mice (0.266 ± 0.057 vs. 0.78 ± 0.153, p = 0.013). Tanshinone IIA also decreased intestinal permeability compared to that in AOM/DSS-treated alone mice (3.12 ± 0.369 vs. 5.06 ± 0.597, p = 0.034) and consequently reduced neutrophil infiltration of the colonic mucosa (53.25 ± 8.85 vs. 107.6 ± 13.09, p = 0.014) as well as intestinal inflammation in mice. Mechanistically, tanshinone IIA downregulated the NF-κB signalling pathway in the colonic tumours of AOM/DSS-treated mice. In vitro assays further validated that tanshinone IIA suppressed LPS-induced neutrophil activation. CONCLUSION These data suggest that tanshinone IIA alleviates colorectal tumorigenesis through inhibition of intestinal inflammation. Tanshinone IIA may have a therapeutic potential for CRC in clinical practice.
Collapse
Affiliation(s)
- Lijie Liu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Oncology, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Hanjing Gao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Radiation Oncology, Tianjin 4TH Centre Hospital, Tianjin, China
| | - Tao Wen
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Tao Gu
- Department of Oncology, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Shuang Zhang
- Department of Cardiology, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Zhiyong Yuan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- CONTACT Zhiyong Yuan Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy, West Huan-Hu Road, Hexi District, Tianjin300060, China
| |
Collapse
|
13
|
Chen K, Zhang B, Li J, Pan A, Cao L, Zhao X, Huang S, Chen L. TiaochangXiaoliu decoction inhibits azomethane (AOM)/dextran sulfate sodium (DSS)-induced colorectal cancer by regulating immune response. J Gastrointest Oncol 2021; 12:2223-2231. [PMID: 34790387 DOI: 10.21037/jgo-21-580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/29/2021] [Indexed: 11/06/2022] Open
Abstract
Background TiaochangXiaoliu decoction (TXD) has an anti-tumor effect in clinical practice. We further investigated the role of TXD in colorectal cancer (CRC). Methods Mouse models of CRC were induced by azomethane (AOM)/dextran sulfate sodium (DSS), with sixty male C57BL/6 mice randomly divided into six groups (10 mice/group): a control group, AOM/DSS group, TXD at low dose (L-dose) group, middle dose (M-dose) group, high dose (H-dose) group, and Celecoxib (Cel) group. The colorectum, serum, and plasma of mice in each group was collected following sacrifice to record the number of tumors. HE staining was utilized for observing pathological damage to colorectal tissues, ELISA used for detecting INF-γ, IL-2, and TNF-α expression in serum, and flow cytometry used for measuring the proportion of CD4+, CD8+, CD4+/CD8+, and NK cells in plasma. Results Compared with the control group, the AOM/DSS group showed tumor masses in the colorectum and different degrees of pathological damage in the intestine. AOM/DSS induction also resulted in an increase in INF-γ, IL-2, and TNF-α expression in serum, and a decrease in the percentages of CD4+, CD8+, CD4+/CD8+, and NK cells(P<0.05). In comparison with the AOM/DSS group, with the increase of TXD dose, the number of tumors decreased significantly, and intestinal structure and mucosal inflammatory cell infiltration also improved. Further, in comparison with the AOM/DSS group, all three doses of TXD and celecoxib caused an increase in the contents of CD4+, CD8+, CD4+/CD8+, and NK cells in plasma. In addition, in the M-dose, H-dose, and Cel groups, INF-γ, IL-2, and TNF-α expression showed a marked decrease, and the reduction in these two groups treated with TXD was dose-dependent. Conclusions TXD leads to a marked reduction in the number of tumors and inflammatory cell infiltration in CRC mice. This decoction significantly decreased the levels of INF-γ, IL-2, and TNF-α in serum, and increased the contents of CD4+, CD8+, CD4+/CD8+, and NK cell in the plasma of mice with AOM/DSS-induced CRC.
Collapse
Affiliation(s)
- Kefang Chen
- Department of Spleen and Stomach Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Beiping Zhang
- Department of Spleen and Stomach Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianjun Li
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Aizhen Pan
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Linhui Cao
- Department of Traditional Chinese Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiying Zhao
- Department of Spleen and Stomach Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Suiping Huang
- Department of Spleen and Stomach Diseases, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liudan Chen
- Department of Acupuncture and Moxibustion, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Ding J, Gou Q, Jia X, Liu Q, Jin J, Shi J, Hou Y. AMPK phosphorylates PPARδ to mediate its stabilization, inhibit glucose and glutamine uptake and colon tumor growth. J Biol Chem 2021; 297:100954. [PMID: 34270958 PMCID: PMC8397901 DOI: 10.1016/j.jbc.2021.100954] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/25/2021] [Accepted: 07/09/2021] [Indexed: 12/25/2022] Open
Abstract
Peroxisome proliferator-activated receptor δ (PPARδ) is a nuclear receptor transcription factor that plays an important role in the regulation of metabolism, inflammation, and cancer. In addition, the nutrient-sensing kinase 5'AMP-activated protein kinase (AMPK) is a critical regulator of cellular energy in coordination with PPARδ. However, the molecular mechanism of the AMPK/PPARδ pathway on cancer progression is still unclear. Here, we found that activated AMPK induced PPARδ-S50 phosphorylation in cancer cells, whereas the PPARδ/S50A (nonphosphorylation mimic) mutant reversed this event. Further analysis showed that the PPARδ/S50E (phosphorylation mimic) but not the PPARδ/S50A mutant increased PPARδ protein stability, which led to reduced p62/SQSTM1-mediated degradation of misfolded PPARδ. Furthermore, PPARδ-S50 phosphorylation decreased PPARδ transcription activity and alleviated PPARδ-mediated uptake of glucose and glutamine in cancer cells. Soft agar and xenograft tumor model analysis showed that the PPARδ/S50E mutant but not the PPARδ/S50A mutant inhibited colon cancer cell proliferation and tumor growth, which was associated with inhibition of Glut1 and SLC1A5 transporter protein expression. These findings reveal a new mechanism of AMPK-induced PPARδ-S50 phosphorylation, accumulation of misfolded PPARδ protein, and inhibition of PPARδ transcription activity contributing to the suppression of colon tumor formation.
Collapse
Affiliation(s)
- Jiajun Ding
- School of Life Sciences, Jiangsu University, Zhenjiang, PR China
| | - Qian Gou
- School of Medicine, Jiangsu University, Zhenjiang, PR China
| | - Xiao Jia
- School of Life Sciences, Jiangsu University, Zhenjiang, PR China
| | - Qian Liu
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, PR China
| | - Jianhua Jin
- Department of Oncology, Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, PR China
| | - Juanjuan Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, PR China.
| | - Yongzhong Hou
- School of Life Sciences, Jiangsu University, Zhenjiang, PR China.
| |
Collapse
|
15
|
Di Maio G, Alessio N, Demirsoy IH, Peluso G, Perrotta S, Monda M, Di Bernardo G. Evaluation of Browning Agents on the White Adipogenesis of Bone Marrow Mesenchymal Stromal Cells: A Contribution to Fighting Obesity. Cells 2021; 10:403. [PMID: 33669222 PMCID: PMC7919793 DOI: 10.3390/cells10020403] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/28/2021] [Accepted: 02/10/2021] [Indexed: 01/19/2023] Open
Abstract
Brown-like adipocytes can be induced in white fat depots by a different environmental or drug stimuli, known as "browning" or "beiging". These brite adipocytes express thermogenin UCP1 protein and show different metabolic advantages, such as the ability to acquire a thermogenic phenotype corresponding to standard brown adipocytes that counteracts obesity. In this research, we evaluated the effects of several browning agents during white adipocyte differentiation of bone marrow-derived mesenchymal stromal cells (MSCs). Our in vitro findings identified two compounds that may warrant further in vivo investigation as possible anti-obesity drugs. We found that rosiglitazone and sildenafil are the most promising drug candidates for a browning treatment of obesity. These drugs are already available on the market for treating diabetes and erectile dysfunction, respectively. Thus, their off-label use may be contemplated, but it must be emphasized that some severe side effects are associated with use of these drugs.
Collapse
Affiliation(s)
- Girolamo Di Maio
- Human Physiology and Unit of Dietetic and Sports Medicine Section, Department of Experimental Medicine, School of Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (G.D.M.); (M.M.)
| | - Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, School of Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (N.A.); (I.H.D.)
| | - Ibrahim Halil Demirsoy
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, School of Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (N.A.); (I.H.D.)
| | | | - Silverio Perrotta
- Dipartimento della Donna, del Bambino e di Chirurgia Generale e Specialistica, School of Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy;
| | - Marcellino Monda
- Human Physiology and Unit of Dietetic and Sports Medicine Section, Department of Experimental Medicine, School of Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (G.D.M.); (M.M.)
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, School of Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (N.A.); (I.H.D.)
| |
Collapse
|
16
|
Kadayat TM, Shrestha A, Jeon YH, An H, Kim J, Cho SJ, Chin J. Targeting Peroxisome Proliferator-Activated Receptor Delta (PPARδ): A Medicinal Chemistry Perspective. J Med Chem 2020; 63:10109-10134. [DOI: 10.1021/acs.jmedchem.9b01882] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Tara Man Kadayat
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Aarajana Shrestha
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Yong Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Republic of Korea
| | - Hongchan An
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jina Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Sung Jin Cho
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu 41404, Republic of Korea
| | - Jungwook Chin
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| |
Collapse
|
17
|
Xiao F, Wang Y, Shao T, Jin G. Acetonitrilated Unsymmetric BODIPYs having glycine fluorescence responsive quenching: Design, synthesis and spectroscopic properties. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 233:118211. [PMID: 32155579 DOI: 10.1016/j.saa.2020.118211] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/29/2020] [Accepted: 02/29/2020] [Indexed: 06/10/2023]
Abstract
A series of novel N≡C-CH2-B-F system BODIPY were designed and synthesized by introducing aldehyde and acetonitrile units which gave positive influence to spectroscopic and chemical properties of BODIPY derivatives. The effects of glycine (Gly) on the target products were studied via ultraviolet and visible spectrophotometry (UV-Vis) and photoluminescence (PL) under different conditions of the presence and absence of cations (K+, Ca2+, Zn2+). It was showed that glycine has an intense quenching effect on the compounds in both the presence and absence of ions with a dramatic color change from notable red to light orange owing to the addition of Gly. With regard to cells imaging investigation, the products showed the prominent fluorescence in cholangiocarcinoma cells. The luminescent effect of compounds 1 and 3 entering the cells was significantly stronger than that of compound 2. In addition, pertaining to anticancer properties, two human cancer cell lines (RBE, HCCC-9810) and one normal cell line (L-02) were evaluated for in vitro cytotoxicity. The target compounds, 1-3, exhibited moderate antitumor activity, of which compound 1 was found to be the most potent derivative with IC50 values of 119.31 ± 6.25, 114.73 ± 3.25, and 106.33 ± 5.22 against RBE, HCCC-9810, and L-02 cells, respectively, slightly weaker than the positive control 5-FU.
Collapse
Affiliation(s)
- Fuyan Xiao
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Yuling Wang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Tingyu Shao
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China
| | - Guofan Jin
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
18
|
PPARδ is a regulator of autophagy by its phosphorylation. Oncogene 2020; 39:4844-4853. [PMID: 32439863 DOI: 10.1038/s41388-020-1329-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
In response to nutrient deficiency, autophagy degrades cytoplasmic materials and organelles in lysosomes, which is nutrient recycling, whereas activation of EGFR mediates autophagy suppression in response to growth factors. It is unclear whether PPARδ could be the regulator of autophagy in response to active EGFR. Here we found that EGFR induced PPARδ phosphorylation at tyrosine-108 leading to increased binding of LC3 to PPARδ by its LIR (LC3 interacting region) motif, consequently, inhibited autophagic flux. Conversely, EGFR inhibitor treatment reversed this event. Furthermore, EGFR-mediated PPARδ phosphorylation at tyrosine-108 led to autophagy inhibition and tumor growth. These findings suggest that PPARδ serves as a regulator of autophagy by its phosphorylation.
Collapse
|
19
|
Wagner N, Wagner KD. PPAR Beta/Delta and the Hallmarks of Cancer. Cells 2020; 9:cells9051133. [PMID: 32375405 PMCID: PMC7291220 DOI: 10.3390/cells9051133] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear hormone receptor family. Three different isoforms, PPAR alpha, PPAR beta/delta and PPAR gamma have been identified. They all form heterodimers with retinoic X receptors to activate or repress downstream target genes dependent on the presence/absence of ligands and coactivators or corepressors. PPARs differ in their tissue expression profile, ligands and specific agonists and antagonists. PPARs attract attention as potential therapeutic targets for a variety of diseases. PPAR alpha and gamma agonists are in clinical use for the treatment of dyslipidemias and diabetes. For both receptors, several clinical trials as potential therapeutic targets for cancer are ongoing. In contrast, PPAR beta/delta has been suggested as a therapeutic target for metabolic syndrome. However, potential risks in the settings of cancer are less clear. A variety of studies have investigated PPAR beta/delta expression or activation/inhibition in different cancer cell models in vitro, but the relevance for cancer growth in vivo is less well documented and controversial. In this review, we summarize critically the knowledge of PPAR beta/delta functions for the different hallmarks of cancer biological capabilities, which interplay to determine cancer growth.
Collapse
|
20
|
Jin G, Xiao F, Li Z, Qi X, Zhao L, Sun X. Design, Synthesis, and Dual Evaluation of Quinoline and Quinolinium Iodide Salt Derivatives as Potential Anticancer and Antibacterial Agents. ChemMedChem 2020; 15:600-609. [PMID: 32068948 DOI: 10.1002/cmdc.202000002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/06/2020] [Indexed: 12/20/2022]
Abstract
A series of novel quinoline and quinolinium iodide derivatives were designed and synthesized to discover potential anticancer and antibacterial agents. With regard to anticancer properties, in vitro cytotoxicities against three human cancer cell lines (A-549, HeLa and SGC-7901) were evaluated. The antibacterial properties against two strains, Escherichia coli (ATCC 29213) and Staphylococcus aureus (ATCC 8739), along with minimum inhibitory concentration (MIC) values were evaluated. The target alkyliodine substituted compounds exhibited significant antitumor and antibacterial activity, of which compound 8-((4-(benzyloxy)phenyl)amino)-7-(ethoxycarbonyl)-5-propyl-[1,3]dioxolo[4,5-g]quinolin-5-ium (12) was found to be the most potent derivative with IC50 values of 4.45±0.88, 4.74±0.42, 14.54±1.96, and 32.12±3.66 against A-549, HeLa, SGC-7901, and L-02 cells, respectively, stronger than the positive controls 5-FU and MTX. Furthermore, compound 12 had the most potent bacterial inhibitory activity. The MIC of this compound against both E. coli and S. aureus was 3.125 nmol ⋅ mL-1 , which was smaller than that against the reference agents amoxicillin and ciprofloxacin.
Collapse
Affiliation(s)
- Guofan Jin
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Fuyan Xiao
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Zhenwang Li
- College of Animal Science and Technique, Bayi Agriculture University, Daqing, 163319, Heilongjiang, China
| | - Xueyong Qi
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, China
| | - Lei Zhao
- Siping Institute for Food and Drug Control, Siping, 136000, China
| | - Xianyu Sun
- College of Animal Science and Technique, Bayi Agriculture University, Daqing, 163319, Heilongjiang, China
| |
Collapse
|
21
|
Xu X, Zhang C, Xia Y, Yu J. Over expression of METRN predicts poor clinical prognosis in colorectal cancer. Mol Genet Genomic Med 2019; 8:e1102. [PMID: 31859449 PMCID: PMC7057108 DOI: 10.1002/mgg3.1102] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/12/2019] [Accepted: 12/05/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The role of meteorin (METRN) in colorectal cancer has not been reported previously. We aimed to explore the relationship between METRN and colorectal cancer (CRC) prognosis. METHODS Data were retrieved from the Gene Expression Omnibus database. Gene expression values were log2 transformed and normalized by quantile normalization. Missing values were imputed with the R impute package. Differentially expressed genes were analyzed using the R limma package. METRN expression was compared between normal and CRC tissues and among different stages and subtypes of CRC. We assessed the relationship between METRN and KRAS/BRAF mutations in CRC. Five-year overall (OS), disease-free (DFS), and disease-specific survival (DSS) rates were determined by Kaplan-Meier analysis and analyzed by log-rank test. RESULTS METRN was expressed at a higher level in CRC (p = .0011) than in normal tissues, especially in advanced stages (p = .0343). METRN expression levels were higher in the MSI (dMMR) subtype (p < .001) and usually with BRAF mutations (p < .0001). METRN overexpression was associated with poor prognosis and low OS (p = .01014), DFS (p = .0146), and DSS (p < .0001) rates. CONCLUSION METRN overexpression is a predictive factor for poor prognosis in patients with CRC.
Collapse
Affiliation(s)
- Xin Xu
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chihao Zhang
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Xia
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiwei Yu
- Department of General Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
22
|
Gou Q, Dong C, Jin J, Liu Q, Lu W, Shi J, Hou Y. PPARα agonist alleviates tumor growth and chemo-resistance associated with the inhibition of glucose metabolic pathway. Eur J Pharmacol 2019; 863:172664. [PMID: 31539552 DOI: 10.1016/j.ejphar.2019.172664] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 12/17/2022]
Abstract
As a nuclear receptor, peroxisome-proliferator-activated receptor α (PPARα) plays a critical role in regulation of metabolism and cancer, while the effect of PPARα agonist on cancer cell glucose metabolism-linked tumor growth is still unclear. Here we found that PPARα agonist (Wy14,643) decreased Glut1 (Glucose transporter 1) gene and protein expressions of colorectal cancer cell lines in response to normoxia or hypoxia. Dual-luciferase analysis showed that Wy14,643 inhibited Glut1 transcription activity. Importantly, ChIP-qPCR analysis showed that Wy14,643 increased the binding of PPARα to Glut1 promoter region. Wy14,643 suppressed Glut1 transcription activity resulting in reduced influx of glucose in cancer cells in response to normoxia or hypoxia. Further analysis showed that Wy14,643-mediated inhibition of tumor growth and chemo-resistance was associated with inhibition of mTOR pathway. Taken together, PPARα agonist Wy14,643 suppressed Glut1 transcription activity, glucose uptake and mTOR pathway in colorectal cancer cells, which was involved in reduced tumor growth and chemo-resistance. These findings provided a novel therapy strategy for cancer progression.
Collapse
Affiliation(s)
- Qian Gou
- Department of Oncology, The Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, Jiangsu Province. 213162, People's Republic of China; Institute of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212000, People's Republic of China
| | - Chen Dong
- Institute of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212000, People's Republic of China
| | - Jianhua Jin
- Department of Oncology, The Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, Jiangsu Province. 213162, People's Republic of China
| | - Qian Liu
- Department of Oncology, The Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, Jiangsu Province. 213162, People's Republic of China
| | - Wenbin Lu
- Department of Oncology, The Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, Jiangsu Province. 213162, People's Republic of China.
| | - Juanjuan Shi
- Institute of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212000, People's Republic of China.
| | - Yongzhong Hou
- Department of Oncology, The Affiliated Wujin People's Hospital, Jiangsu University, Changzhou, Jiangsu Province. 213162, People's Republic of China; Institute of Life Science, Jiangsu University, Zhenjiang, Jiangsu Province, 212000, People's Republic of China.
| |
Collapse
|
23
|
Ding J, Gou Q, Jin J, Shi J, Liu Q, Hou Y. Metformin inhibits PPARδ agonist-mediated tumor growth by reducing Glut1 and SLC1A5 expressions of cancer cells. Eur J Pharmacol 2019; 857:172425. [DOI: 10.1016/j.ejphar.2019.172425] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 05/14/2019] [Accepted: 05/27/2019] [Indexed: 12/22/2022]
|