1
|
Kounatidis D, Tentolouris N, Vallianou NG, Mourouzis I, Karampela I, Stratigou T, Rebelos E, Kouveletsou M, Stamatopoulos V, Tsaroucha E, Dalamaga M. The Pleiotropic Effects of Lipid-Modifying Interventions: Exploring Traditional and Emerging Hypolipidemic Therapies. Metabolites 2024; 14:388. [PMID: 39057711 PMCID: PMC11278853 DOI: 10.3390/metabo14070388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerotic cardiovascular disease poses a significant global health issue, with dyslipidemia standing out as a major risk factor. In recent decades, lipid-lowering therapies have evolved significantly, with statins emerging as the cornerstone treatment. These interventions play a crucial role in both primary and secondary prevention by effectively reducing cardiovascular risk through lipid profile enhancements. Beyond their primary lipid-lowering effects, extensive research indicates that these therapies exhibit pleiotropic actions, offering additional health benefits. These include anti-inflammatory properties, improvements in vascular health and glucose metabolism, and potential implications in cancer management. While statins and ezetimibe have been extensively studied, newer lipid-lowering agents also demonstrate similar pleiotropic effects, even in the absence of direct cardiovascular benefits. This narrative review explores the diverse pleiotropic properties of lipid-modifying therapies, emphasizing their non-lipid effects that contribute to reducing cardiovascular burden and exploring emerging benefits for non-cardiovascular conditions. Mechanistic insights into these actions are discussed alongside their potential therapeutic implications.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece;
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Theodora Stratigou
- Department of Endocrinology and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Eleni Rebelos
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Marina Kouveletsou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | | | - Eleni Tsaroucha
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
2
|
Jin J, Liang X, Bi W, Liu R, Zhang S, He Y, Xie Q, Liu S, Xiao JC, Zhang P. Identification of a Difluorinated Alkoxy Sulfonyl Chloride as a Novel Antitumor Agent for Hepatocellular Carcinoma through Activating Fumarate Hydratase Activity. Pharmaceuticals (Basel) 2023; 16:1705. [PMID: 38139831 PMCID: PMC10748328 DOI: 10.3390/ph16121705] [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: 10/31/2023] [Revised: 11/18/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Fenofibrate is known as a lipid-lowering drug. Although previous studies have reported that fenofibrate exhibits potential antitumor activities, IC50 values of fenofibrate could be as high as 200 μM. Therefore, we investigated the antitumor activities of six synthesized fenofibrate derivatives. We discovered that one compound, SIOC-XJC-SF02, showed significant antiproliferative activity on human hepatocellular carcinoma (HCC) HCCLM3 cells and HepG2 cells (the IC50 values were 4.011 μM and 10.908 μM, respectively). We also found this compound could inhibit the migration of human HCC cells. Transmission electron microscope and flow cytometry assays demonstrated that this compound could induce apoptosis of human HCC cells. The potential binding sites of this compound acting on human HCC cells were identified by mass spectrometry-cellular thermal shift assay (MS-CETSA). Molecular docking, Western blot, and enzyme activity assay-validated binding sites in human HCC cells. The results showed that fumarate hydratase may be a potential binding site of this compound, exerting antitumor effects. A xenograft model in nude mice demonstrated the anti-liver cancer activity and the mechanism of action of this compound. These findings indicated that the antitumor effect of this compound may act via activating fumarate hydratase, and this compound may be a promising antitumor candidate for further investigation.
Collapse
Affiliation(s)
- Jin Jin
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xujun Liang
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Wu Bi
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ruijie Liu
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Sai Zhang
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yi He
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qingming Xie
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shilei Liu
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ji-Chang Xiao
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Pengfei Zhang
- NHC Key Laboratory of Cancer Proteomics, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China; (J.J.); (W.B.); (R.L.); (Q.X.); (S.L.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
3
|
Cizkova K, Tauber Z. Fibrates Affect Levels of Phosphorylated p38 in Intestinal Cells in a Differentiation-Dependent Manner. Int J Mol Sci 2023; 24:ijms24097695. [PMID: 37175404 PMCID: PMC10178720 DOI: 10.3390/ijms24097695] [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: 03/31/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Fibrates are widely used hypolipidaemic agents that act as ligands of the peroxisome proliferator-activated receptor α (PPARα). p38 is a protein kinase that is mainly activated by environmental and genotoxic stress. We investigated the effect of the PPARα activators fenofibrate and WY-14643 and the PPARα inhibitor GW6471 on the levels of activated p38 (p-p38) in the colorectal cancer cell lines HT-29 and Caco2 in relation to their differentiation status. Fibrates increased p-p38 in undifferentiated HT-29 cells, whereas in other cases p-p38 expression was decreased. HT-29 cells showed p-p38 predominantly in the cytoplasm, whereas Caco2 cells showed higher nuclear positivity. The effect of fibrates may depend on the differentiation status of the cell, as differentiated HT-29 and undifferentiated Caco2 cells share similar characteristics in terms of villin, CYP2J2, and soluble epoxide hydrolase (sEH) expression. In human colorectal carcinoma, higher levels of p-p38 were detected in the cytoplasm, whereas in normal colonic surface epithelium, p-p38 showed nuclear positivity. The decrease in p-p38 positivity was associated with a decrease in sEH, consistent with in vitro results. In conclusion, fibrates affect the level of p-p38, but its exact role in the process of carcinogenesis remains unclear and further research is needed in this area.
Collapse
Affiliation(s)
- Katerina Cizkova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Zdenek Tauber
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| |
Collapse
|
4
|
PPARs and the Kynurenine Pathway in Melanoma-Potential Biological Interactions. Int J Mol Sci 2023; 24:ijms24043114. [PMID: 36834531 PMCID: PMC9960262 DOI: 10.3390/ijms24043114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors involved in various physiological and pathological processes within the skin. PPARs regulate several processes in one of the most aggressive skin cancers, melanoma, including proliferation, cell cycle, metabolic homeostasis, cell death, and metastasis. In this review, we focused not only on the biological activity of PPAR isoforms in melanoma initiation, progression, and metastasis but also on potential biological interactions between the PPAR signaling and the kynurenine pathways. The kynurenine pathway is a major pathway of tryptophan metabolism leading to nicotinamide adenine dinucleotide (NAD+) production. Importantly, various tryptophan metabolites exert biological activity toward cancer cells, including melanoma. Previous studies confirmed the functional relationship between PPAR and the kynurenine pathway in skeletal muscles. Despite the fact this interaction has not been reported in melanoma to date, some bioinformatics data and biological activity of PPAR ligands and tryptophan metabolites may suggest a potential involvement of these metabolic and signaling pathways in melanoma initiation, progression, and metastasis. Importantly, the possible relationship between the PPAR signaling pathway and the kynurenine pathway may relate not only to the direct biological effect on melanoma cells but also to the tumor microenvironment and the immune system.
Collapse
|
5
|
Miglitol, an Oral Antidiabetic Drug, Downregulates Melanogenesis in B16F10 Melanoma Cells through the PKA, MAPK, and GSK3β/β-Catenin Signaling Pathways. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010115. [PMID: 36615308 PMCID: PMC9822252 DOI: 10.3390/molecules28010115] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022]
Abstract
Hyperpigmentation is a common condition that causes darker spots or patches on the skin, which often look brown, black, gray, red, or pink. This results in unresolved psychological impact due to high anxiety, depression, and somatoform disorder. We aimed to repurpose an antidiabetic drug, miglitol, as an effective compound against hyperpigmentation when applied as a cosmeceutical agent. The present study investigated the antimelanogenic effects of miglitol and the trehalase inhibitor validamycin A. Miglitol in isolation exhibited no cytotoxicity and significantly reduced the melanin production and intracellular tyrosinase activity in B16F10 melanoma cells. The Western blotting results showed that miglitol reduces the expression of melanogenic regulatory factors, including tyrosinase, tyrosinase-related protein (TRP)-1, TRP-2, and microphthalmia-associated transcription factor (MITF). Mechanistically, miglitol appears to suppress melanin synthesis through cAMP-dependent protein kinase (PKA)-dependent downregulation of MITF, a master transcription factor in melanogenesis. The antimelanogenic effects of miglitol was mediated by downregulation of the p38 signaling pathway and upregulation of extracellular signal-regulated kinase (ERK). Moreover, miglitol decreases P-GSK3β and β-catenin levels compared to those in the untreated group. However, miglitol activated P-β-catenin expression compared to that in the untreated group. Finally, we tested the potential of miglitol in topical application through primary human skin irritation tests on the normal skin (upper back) of 33 volunteers. In these assays, miglitol (125 and 250 μM) did not induce any adverse reactions. Taken together, these findings suggest that the regulation of melanogenesis by miglitol may be mediated by the PKA, MAPK, and GSK3β/β-Catenin signaling pathways and that miglitol might provide new insights into drug repurposing for the treatment of hyperpigmentation symptoms.
Collapse
|
6
|
3,4,5-Trimethoxycinnamate thymol ester inhibits melanogenesis in normal human melanocytes and 3D human epidermal equivalents via the PGC-1α-independent PPARγ partial agonism. J Dermatol Sci 2022; 106:12-20. [DOI: 10.1016/j.jdermsci.2022.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 02/10/2022] [Accepted: 02/27/2022] [Indexed: 11/23/2022]
|
7
|
Wang J, Luo L, Ding Q, Wu Z, Peng Y, Li J, Wang X, Li W, Liu G, Zhang B, Tang Y. Development of a Multi-Target Strategy for the Treatment of Vitiligo via Machine Learning and Network Analysis Methods. Front Pharmacol 2021; 12:754175. [PMID: 34603063 PMCID: PMC8479195 DOI: 10.3389/fphar.2021.754175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/03/2021] [Indexed: 01/14/2023] Open
Abstract
Vitiligo is a complex disorder characterized by the loss of pigment in the skin. The current therapeutic strategies are limited. The identification of novel drug targets and candidates is highly challenging for vitiligo. Here we proposed a systematic framework to discover potential therapeutic targets, and further explore the underlying mechanism of kaempferide, one of major ingredients from Vernonia anthelmintica (L.) willd, for vitiligo. By collecting transcriptome and protein-protein interactome data, the combination of random forest (RF) and greedy articulation points removal (GAPR) methods was used to discover potential therapeutic targets for vitiligo. The results showed that the RF model performed well with AUC (area under the receiver operating characteristic curve) = 0.926, and led to prioritization of 722 important transcriptomic features. Then, network analysis revealed that 44 articulation proteins in vitiligo network were considered as potential therapeutic targets by the GAPR method. Finally, through integrating the above results and proteomic profiling of kaempferide, the multi-target strategy for vitiligo was dissected, including 1) the suppression of the p38 MAPK signaling pathway by inhibiting CDK1 and PBK, and 2) the modulation of cellular redox homeostasis, especially the TXN and GSH antioxidant systems, for the purpose of melanogenesis. Meanwhile, this strategy may offer a novel perspective to discover drug candidates for vitiligo. Thus, the framework would be a useful tool to discover potential therapeutic strategies and drug candidates for complex diseases.
Collapse
Affiliation(s)
- Jiye Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Lin Luo
- Key Laboratory of Xinjiang Phytomedicine Resources of Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| | - Qiong Ding
- Key Laboratory of Xinjiang Phytomedicine Resources of Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| | - Zengrui Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yayuan Peng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jie Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiaoqin Wang
- Key Laboratory of Xinjiang Phytomedicine Resources of Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China.,Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Bo Zhang
- Key Laboratory of Xinjiang Phytomedicine Resources of Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China.,Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
8
|
Tomas-Hernandez S, Blanco J, Garcia-Vallvé S, Pujadas G, Ojeda-Montes MJ, Gimeno A, Arola L, Minghetti L, Beltrán-Debón R, Mulero M. Anti-Inflammatory and Immunomodulatory Effects of the Grifola frondosa Natural Compound o-Orsellinaldehyde on LPS-Challenged Murine Primary Glial Cells. Roles of NF-κβ and MAPK. Pharmaceutics 2021; 13:806. [PMID: 34071571 PMCID: PMC8229786 DOI: 10.3390/pharmaceutics13060806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/18/2021] [Accepted: 05/25/2021] [Indexed: 12/17/2022] Open
Abstract
In response to foreign or endogenous stimuli, both microglia and astrocytes adopt an activated phenotype that promotes the release of pro-inflammatory mediators. This inflammatory mechanism, known as neuroinflammation, is essential in the defense against foreign invasion and in normal tissue repair; nevertheless, when constantly activated, this process can become detrimental through the release of neurotoxic factors that amplify underlying disease. In consequence, this study presents the anti-inflammatory and immunomodulatory properties of o-orsellinaldehyde, a natural compound found by an in silico approach in the Grifola frondosa mushroom, in astrocytes and microglia cells. For this purpose, primary microglia and astrocytes were isolated from mice brain and cultured in vitro. Subsequently, cells were exposed to LPS in the absence or presence of increasing concentrations of this natural compound. Specifically, the results shown that o-orsellinaldehyde strongly inhibits the LPS-induced inflammatory response in astrocytes and microglia by decreasing nitrite formation and downregulating iNOS and HO-1 expression. Furthermore, in microglia cells o-orsellinaldehyde inhibits NF-κB activation; and potently counteracts LPS-mediated p38 kinase and JNK phosphorylation (MAPK). In this regard, o-orsellinaldehyde treatment also induces a significant cell immunomodulation by repolarizing microglia toward the M2 anti-inflammatory phenotype. Altogether, these results could partially explain the reported beneficial effects of G. frondosa extracts on inflammatory conditions.
Collapse
Affiliation(s)
- Sarah Tomas-Hernandez
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology, Campus Sescelades, Universitat Rovira i Virgili (URV), 43007 Tarragona, Catalonia, Spain; (S.T.-H.); (S.G.-V.); (G.P.); (M.J.O.-M.); (A.G.)
| | - Jordi Blanco
- Physiology Unit, Laboratory of Toxicology and Environmental Health, Research in Neurobehavior and Health (NEUROLAB), School of Medicine, IISPV, Universitat Rovira i Virgili (URV), 43202 Tarragona, Catalonia, Spain;
| | - Santiago Garcia-Vallvé
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology, Campus Sescelades, Universitat Rovira i Virgili (URV), 43007 Tarragona, Catalonia, Spain; (S.T.-H.); (S.G.-V.); (G.P.); (M.J.O.-M.); (A.G.)
| | - Gerard Pujadas
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology, Campus Sescelades, Universitat Rovira i Virgili (URV), 43007 Tarragona, Catalonia, Spain; (S.T.-H.); (S.G.-V.); (G.P.); (M.J.O.-M.); (A.G.)
| | - María José Ojeda-Montes
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology, Campus Sescelades, Universitat Rovira i Virgili (URV), 43007 Tarragona, Catalonia, Spain; (S.T.-H.); (S.G.-V.); (G.P.); (M.J.O.-M.); (A.G.)
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Aleix Gimeno
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology, Campus Sescelades, Universitat Rovira i Virgili (URV), 43007 Tarragona, Catalonia, Spain; (S.T.-H.); (S.G.-V.); (G.P.); (M.J.O.-M.); (A.G.)
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Baldiri Reixac 10-12, 08020 Barcelona, Catalonia, Spain
| | - Lluís Arola
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Campus Sescelades, Universitat Rovira i Virgili (URV), 43007 Tarragona, Catalonia, Spain;
| | - Luisa Minghetti
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Raúl Beltrán-Debón
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43007 Tarragona, Catalonia, Spain;
| | - Miquel Mulero
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Campus Sescelades, Universitat Rovira i Virgili (URV), 43007 Tarragona, Catalonia, Spain;
| |
Collapse
|
9
|
Hydroxychloroquine Potentiates Apoptosis Induced by PPAR α Antagonist in 786-O Clear Cell Renal Cell Carcinoma Cells Associated with Inhibiting Autophagy. PPAR Res 2021; 2021:6631605. [PMID: 33959154 PMCID: PMC8075691 DOI: 10.1155/2021/6631605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/14/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the major pathological pattern of renal cell carcinoma. The ccRCC cells exhibit a certain degree of inherent drug resistance due to some genetic mutations. In recent years, peroxisome proliferator-activated receptor-α (PPARα) antagonists have been reported as a targeted therapeutic drug capable of inducing apoptosis and cell cycle arrest in the ccRCC cell line. Autophagy, which can be induced by stress in eukaryotic cells, plays a complex role in the proliferation, survival, and death of tumor cells. In our study, we found that the expression of PPARα was low in highly differentiated ccRCC tissues and 786-O cell line but high in poorly differentiated ccRCC tissues. The level of PPARα expression in ccRCC tissues is correlated to the grade of differentiation, but not to the sex or age of ccRCC patients. The findings also revealed that the PPARα antagonist GW6471 can lower cell viability and induce autophagy in the 786-O ccRCC cell line. This autophagy can be inhibited by hydroxychloroquine. When treated with a combination of hydroxychloroquine and GW6471, the viability of the 786-O cells was decreased further when compared to the treatment with GW6471 or hydroxychloroquine alone, and apoptosis was promoted. Meanwhile, when human kidney 2 cells were cotreated with hydroxychloroquine and GW6471, cell viability was only slightly influenced. Hence, our finding indicates that the combination of GW6471 and hydroxychloroquine may constitute a novel and potentially effective treatment for ccRCC. Furthermore, this approach is likely to be safe owing to its minimal effects on normal renal tissues.
Collapse
|
10
|
Jung HJ, Kim SM, Kim DH, Bang E, Kang D, Lee S, Chun P, Moon HR, Chung HY. 2,4-Dihydroxyphenyl-benzo[d]thiazole (MHY553), a synthetic PPARα agonist, decreases age-associated inflammatory responses through PPARα activation and RS scavenging in the skin. Exp Gerontol 2020; 143:111153. [PMID: 33189833 DOI: 10.1016/j.exger.2020.111153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/16/2022]
Abstract
We previously reported that 2,4-dihydroxyphenyl-benzo[d]thiazole (MHY553) is a PPARα agonist, which has been shown to inhibit tyrosinase activity in murine melanocyte and alleviate hepatic steatosis in aged rats. This study investigated the effects of MHY553 on the age-related occurrence of inflammatory responses via the molecular modulation of the nuclear factor-κB (NF-κB) signaling pathway in the skin of aged rats and skin fibroblast cells. Moreover, we investigated the antioxidant effect of MHY553 via in vitro assays of reactive oxygen species (ROS) and peroxynitrite (ONOO-) scavenging activities. We also scrutinized the ability of MHY553 as a PPARα activator in aged rat skin and H2O2-induced Hs27 fibroblast cells. In vivo experiments were performed in young, aged, and MHY553-fed aged rats (3 mg or 5 mg∙kg -1∙day -1 for 4 weeks). MHY553 dose-dependently scavenged ROS and ONOO-. Furthermore, we found that MHY553 suppressed the NF-κB transcription factor and downregulated mitogen-activated protein kinase (MAPK)/activator protein-1 (AP-1) signaling. MHY553 also inhibited the expression of pro-inflammatory cytokines including COX-2, iNOS, IL-1β, and IL-6. Our findings indicate the MHY553 scavenges ROS/reactive nitrogen species and inhibits inflammatory cytokines through PPARα activation in the skin. Thus, these results suggest that MHY553 may be of therapeutic interest for protecting skin from oxidative stress-induced damage and intrinsic aging.
Collapse
Affiliation(s)
- Hee Jin Jung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Seong Min Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Jinju, Gyeongsang 52828, Republic of Korea
| | - Dae Hyun Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - EunJin Bang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Dongwan Kang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Sanggwon Lee
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Pusoon Chun
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae 47392, Republic of Korea
| | - Hyung Ryong Moon
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
11
|
Shehata WA, Maraee A, Mehesin M, Tayel N, Azmy R. Genetic polymorphism of liver X receptor gene in vitiligo: Does it have an association? J Cosmet Dermatol 2020; 20:1906-1914. [PMID: 33031595 DOI: 10.1111/jocd.13772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND Vitiligo is an acquired depigmentation of the skin and the mucous membranes, exhibited as white macules and patches due to selective loss of melanocytes. Etiological theories of vitiligo include genetic, immunological, neurohormonal, cytotoxic, biochemical, oxidative stress, and newer theories of melanocytorrhagy and diminished melanocytes survival. It has been revealed that liver X receptor alpha gene is expressed in skin tissue such as sebaceous glands, hair follicle, keratinocytes, and fibroblasts and is linked to various skin disorders as acne vulgaris and psoriasis. AIM OF THE STUDY To evaluate the association between liver X receptor-α gene polymorphism (rs11039155 and rs2279238) and vitiligo and whether they are related to disease activity and severity or not. SUBJECTS AND METHODS 50 vitiligo patients and 20 age- and sex-matched apparently healthy controls were enrolled. All the included subjects were genotyped using polymerase chain reaction-restriction fragment length polymorphism analysis technique for (-6G/A) and (+1257C/T) SNPs. RESULTS Significant statistical difference between cases and controls regarding genotype and allele frequencies for -6G/A polymorphism with predominance of AA genotype (OR: 5.1, 95% CI: 1.6-15.9) and A allele (OR: 5.3, 95% CI: 1.6-15.9) in cases and also for +1257C/T polymorphism with predominance of TT genotype OR: 9.2 (95% CI: 1.4-82.9) and T allele OR: 3.4 (95% CI: 1.4-8.1) in vitiligo cases. No significant relationship between -6G/A genotypes nor +1257C/T genotypes and disease activity and severity. CONCLUSION The study showed significant association between Liver X receptor gene polymorphisms (-6G/A, +1257 C/T) and development of vitiligo in Egyptian patients. However, it failed to show any relation with disease activity nor severity.
Collapse
Affiliation(s)
- Wafaa A Shehata
- Dermatology, Andrology and STDs Department, Menoufia University, Shebin El-Kom, Egypt
| | - Alaa Maraee
- Dermatology, Andrology and STDs Department, Menoufia University, Shebin El-Kom, Egypt
| | - Marwa Mehesin
- General Practitioner in Health Sector, Shebin El-Kom, Egypt
| | - Nermin Tayel
- Lecturer of Molecular Diagnostics & Therapeutics, Molecular Diagnostics & Therapeutics Department, Genetic Engineering and Biotechnology Research Institute, Sadat City University, Sadat, Egypt
| | - Rania Azmy
- Medical Biochemistry and Molecular Biology Department, Menoufia University, Shebin El-Kom, Egypt
| |
Collapse
|
12
|
Targeting steroid receptor RNA activator (SRA), a long non-coding RNA, enhances melanogenesis through activation of TRP1 and inhibition of p38 phosphorylation. PLoS One 2020; 15:e0237577. [PMID: 32790741 PMCID: PMC7425936 DOI: 10.1371/journal.pone.0237577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 07/29/2020] [Indexed: 12/20/2022] Open
Abstract
Abnormal skin melanin homeostasis results in refractory pigmentary diseases. Melanogenesis is influenced by gene regulation, ultraviolet radiation, and host epigenetic responses. Steroid receptor RNA activator (SRA), a long noncoding RNA, is known to regulate steroidogenesis and tumorigenesis. However, how SRA contributes to melanogenesis remains unknown. Using RNA interference against SRA in B16 and A375 melanoma cells, we observed increased pigmentation and increased expression of TRP1 and TRP2 at transcriptional and translational levels only in B16 cells. The constitutive phosphorylation of p38 in B16-shCtrl cells was inhibited in cells with knocked down SRAi. Moreover, the melanin content of control B16 cells was increased by SB202190, a p38 inhibitor. Furthermore, reduced p38 phosphorylation, enhanced TRP1 expression, and hypermelanosis were observed in A375 cells with RNA interference. These results indicate that SRA-p38-TRP1 axis has a regulatory role in melanin homeostasis and that SRA might be a potential therapeutic target for treating pigmentary diseases.
Collapse
|
13
|
Zhao Z, Ma S, Dong X. Anti-melanogenesis Efficacy of Ginkgolide B is Favored by Attenuating Oxidative Stress and Melanin Synthesis in B16f1 Melanoma Cell Model. INT J PHARMACOL 2019. [DOI: 10.3923/ijp.2019.560.566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
14
|
Schütz R, Rawlings A, Wandeler E, Jackson E, Trevisan S, Monneuse J, Bendik I, Massironi M, Imfeld D. Bio-derived hydroxystearic acid ameliorates skin age spots and conspicuous pores. Int J Cosmet Sci 2019; 41:240-256. [PMID: 30955220 PMCID: PMC6852045 DOI: 10.1111/ics.12529] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/03/2019] [Indexed: 12/21/2022]
Abstract
INTRODUCTION We report on the preparation and efficacy of 10-hydroxystearic acid (HSA) that improves facial age spots and conspicuous pores. METHODS The hydration of oleic acid into HSA was catalyzed by the oleate hydratase from Escherichia coli. Following treatment with HSA, collagen type I and type III was assessed in primary human dermal fibroblasts together with collagen type III, p53 protein levels and sunburn cells (SBC) after UVB irradiation (1 J cm-2 ) by immunohistochemistry on human ex vivo skin. UVB-induced expression of matrix metalloprotease-1 (MMP-1) was determined from full thickness skin by RT-qPCR. Modification of the fibroblast secretome by HSA was studied by mass-spectrometry-based proteomics. In a full-face, double blind, vehicle-controlled trial HSA was assessed for its effects on conspicuous facial pore size and degree of pigmentation of age spots in Caucasian women over an 8-week period. RESULTS HSA was obtained in enantiomeric pure, high yield (≥80%). Collagen type I and type III levels were dose-dependently increased (96% and 244%; P < 0.01) in vitro and collagen type III in ex vivo skin by +57% (P < 0.01) by HSA. HSA also inhibited UVB-induced MMP-1 gene expression (83%; P < 0.01) and mitigated SBC induction (-34% vs. vehicle control) and reduced significantly UV-induced p53 up-regulation (-46% vs. vehicle control; P < 0.01) in irradiated skin. HSA modified the fibroblast secretome with significant increases in proteins associated with the WNT pathway that could reduce melanogenesis and proteins that could modify dermal fibroblast activity and keratinocyte differentiation to account for the alleviation of conspicuous pores. Docking studies in silico and EC50 determination in reporter gene assays (EC50 5.5 × 10-6 M) identified HSA as a peroxisomal proliferator activated receptor-α (PPARα) agonist. Clinically, HSA showed a statistically significant decrease of surface and volume of skin pores (P < 0.05) after 8 weeks of application and age spots became significantly less pigmented than the surrounding skin (contrast, P < 0.05) after 4 weeks. CONCLUSION HSA acts as a PPARα agonist to reduce the signs of age spots and conspicuous pores by significantly modulating the expression of p53, SBC, MMP-1 and collagen together with major changes in secreted proteins that modify keratinocyte, melanocyte and fibroblast cell behavior.
Collapse
Affiliation(s)
- R. Schütz
- DSM Nutritional Products Ltd.KaiseraugstSwitzerland
| | | | - E. Wandeler
- DSM Nutritional Products Ltd.KaiseraugstSwitzerland
| | - E. Jackson
- DSM Nutritional Products Ltd.KaiseraugstSwitzerland
| | | | | | - I. Bendik
- DSM Nutritional Products Ltd.KaiseraugstSwitzerland
| | | | - D. Imfeld
- DSM Nutritional Products Ltd.KaiseraugstSwitzerland
| |
Collapse
|
15
|
Anti-Oxidant and Anti-Melanogenic Properties of Essential Oil from Peel of Pomelo cv. Guan Xi. Molecules 2019; 24:molecules24020242. [PMID: 30634693 PMCID: PMC6359654 DOI: 10.3390/molecules24020242] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 11/17/2022] Open
Abstract
Here, we investigated the anti-oxidant and anti-melanogenic effects of pomelo peel essential oil (PPEO) from pomelo cv. Guan Xi. The volatile chemical composition of PPEO was analyzed with gas chromatography–mass spectrometry (GC/MS). The most abundant component of PPEO was limonene (55.92%), followed by β-myrcene (31.17%), and β-pinene (3.16%). PPEO showed strong anti-oxidant activities against 2,2-diphenyl-2-picryhydrazyl (DPPH), 2,2′-azinobis-(3-ethylbenzthiazoline-6-sulphonate (ABTS) and superoxide anion free radicals. Based on the B16 melanoma cell system, the effects of PPEO on the viability and morphology of B16 cells and the production of melanin were evaluated. The results revealed that PPEO at concentrations below 50 μg/mL could decrease the melanin content without affecting cell viability and morphology. Intracellular tyrosinase (TYR) activity and Western blot analysis showed that PPEO could down-regulate the expression level of TYR in B16 cells and dose-dependently inhibit TYR activity (by a maximum of 64.54%). In conclusion, PPEO has good anti-oxidant and anti-melanogenic activity, and thus can be widely used as a natural antioxidant in the food, pharmaceutical, and cosmetic industries.
Collapse
|
16
|
Lian X, Wang G, Zhou H, Zheng Z, Fu Y, Cai L. Anticancer Properties of Fenofibrate: A Repurposing Use. J Cancer 2018; 9:1527-1537. [PMID: 29760790 PMCID: PMC5950581 DOI: 10.7150/jca.24488] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/25/2018] [Indexed: 12/22/2022] Open
Abstract
Cancer is a leading cause of death throughout the world, and cancer therapy remains a big medical challenge in terms of both its therapeutic efficacy and safety. Therefore, to find out a safe anticancer drug has been long goal for oncologist and medical scientists. Among clinically used medicines with no or little toxicity, fenofibrate is a drug of the fibrate class that plays an important role in lowering the levels of serum cholesterol and triglycerides while elevating the levels of high-density lipoproteins. Recently, several studies have implied that fenofibrate may exert anticancer effects via a variety of pathways involved in apoptosis, cell-cycle arrest, invasion, and migration. Given the great potential that fenofibrate may have anticancer effects, this review was to investigate all published works which directly or indirectly support the anticancer activity of fenofibrate. These studies provide evidence that fenofibrate exerted antitumor effects in several human cancer cell lines, such as breast, liver, glioma, prostate, pancreas, and lung cancer cell lines. Among these studies some have further confirmed the possibility and efficacy of fenofibrate anticancer in xenograft mouse models. In the last part of this review, we also discuss the potential mechanisms of action of fenofibrate based on the available information. Overall, we may repurpose fenofibrate as an anticancer drug in cancer treatment, which urgently need further and comprehensively investigated.
Collapse
Affiliation(s)
- Xin Lian
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
| | - Gang Wang
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China
| | - Honglan Zhou
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China
| | - Zongyu Zheng
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
| | - Yaowen Fu
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China
| | - Lu Cai
- Department of Urology, the First Hospital of Jilin University; 71 Xinmin Street, Changchun 130021, China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA.,Departments of Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
17
|
Furue K, Mitoma C, Tsuji G, Furue M. Protective role of peroxisome proliferator-activated receptor α agonists in skin barrier and inflammation. Immunobiology 2017; 223:327-330. [PMID: 29111315 DOI: 10.1016/j.imbio.2017.10.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 10/24/2017] [Indexed: 01/10/2023]
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is one of the three isoforms of PPARs, which are ligand-activated nuclear transcription factors. PPARα is highly expressed in liver and its agonists are widely used to treat hyperlipidemia. Epidermal keratinocytes express all three isoforms (α, β/δ, and γ) of PPARs and PPARα is particularly important for regulating the epidermal barrier and inflammation. Agonistic ligation of PPARα protects the epidermal barrier function and inhibits the inflammatory response in dermatitis. In this review, we summarize recent topics on the role of PPARα in skin biology and discuss the potential use of topical PPARα agonists for treating atopic dermatitis and other eczemas.
Collapse
Affiliation(s)
- Kazuhisa Furue
- Department of Dermatology, Kyushu University, Fukuoka, Japan
| | - Chikage Mitoma
- Department of Dermatology, Kyushu University, Fukuoka, Japan; Research and Clinical Center for Yusho and Dioxin, Kyushu University, Fukuoka, Japan
| | - Gaku Tsuji
- Department of Dermatology, Kyushu University, Fukuoka, Japan
| | - Masutaka Furue
- Department of Dermatology, Kyushu University, Fukuoka, Japan; Research and Clinical Center for Yusho and Dioxin, Kyushu University, Fukuoka, Japan; Division of Skin Surface Sensing, Department of Dermatology, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
18
|
Anticancer activity of salicin and fenofibrate. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:1061-1071. [PMID: 28733879 DOI: 10.1007/s00210-017-1407-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 07/12/2017] [Indexed: 12/12/2022]
|
19
|
Zhou J, Ren T, Li Y, Cheng A, Xie W, Xu L, Peng L, Lin J, Lian L, Diao Y, Jin X, Yang L. Oleoylethanolamide inhibits α-melanocyte stimulating hormone-stimulated melanogenesis via ERK, Akt and CREB signaling pathways in B16 melanoma cells. Oncotarget 2017; 8:56868-56879. [PMID: 28915638 PMCID: PMC5593609 DOI: 10.18632/oncotarget.18097] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/11/2017] [Indexed: 12/22/2022] Open
Abstract
The present study aimed to examine the potential inhibitory activity of oleoylethanolamide (OEA) on α-melanocyte stimulating hormone (α-MSH)-stimulated melanogenesis and the molecular mechanism(s) involved in the process in B16 mouse melanoma cells. Our data demonstrated that OEA markedly inhibited melanin synthesis and tyrosinase activity in α-MSH-stimulated B16 cells. In addition, the expression of melanogenesis-related proteins, such as melanocortin-1 receptor (MC1R), microphthalmia-associated transcription factor (MITF), tyrosinase-related protein-1 (TRP-1) and tyrosinase, was suppressed in a concentration-dependent manner by OEA. In addition, OEA may suppress melanogenesis through a peroxisome proliferator-activated receptor α (PPARα)-independent pathway. Moreover, OEA activated ERK, Akt, p38 pathways and inhibits CREB pathway in α-MSH-stimulated B16 cells. The specific ERK inhibitor PD98059 partly blocked OEA-inhibited melanin synthesis and tyrosinase activity and partly abrogated the OEA-suppressed expression of melanogenic proteins. Furthermore, OEA presented remarkable inhibition on the body pigmentation in the zebrafish model system. Our findings demonstrated that OEA is an effective inhibitor of hyperpigmentation through activation of ERK, Akt and p38 pathways, inhibition of the CREB pathway, and subsequent down-regulation of MITF, TRP-1 and tyrosinase production.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Tong Ren
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, China
| | - Ying Li
- Department of Pharmacy, Xiamen Medical College, Xiamen, China
| | - Anran Cheng
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Wanyi Xie
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Lanxi Xu
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Lu Peng
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Jinbin Lin
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Lianxiang Lian
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Yong Diao
- School of Biomedical Sciences, Huaqiao University, Quanzhou, China
| | - Xin Jin
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| | - Lichao Yang
- Xiamen Key Laboratory of Chiral Drugs, Medical College, Xiamen University, Xiamen, China
| |
Collapse
|
20
|
Peroxisome proliferator-activated receptor α (PPARα) contributes to control of melanogenesis in B16 F10 melanoma cells. Arch Dermatol Res 2017; 309:141-157. [PMID: 28084540 DOI: 10.1007/s00403-016-1711-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 12/20/2016] [Accepted: 12/23/2016] [Indexed: 01/10/2023]
Abstract
Recent studies revealed the cooperation between peroxisome proliferator-activated receptor gamma (PPARγ) and α-MSH signaling, which results in enhanced melanogenesis in melanocytes and melanoma cells. However, the agonists of PPARα, such as fenofibrate, exert depigmenting effect. Therefore, we aimed to check how the PPARα expression level affects the antimelanogenic activity of fenofibrate and whether PPARα modulates melanogenesis independently of its agonist. To answer these questions, we used three B16 F10-derived cell lines, which varied in the PPARα expression level and were developed by stable transfection with plasmids driving shRNA-based PPARα silencing or overexpression of PPARα-emerald GFP fusion protein. Melanin contents were assessed with electron paramagnetic resonance spectroscopy along with color component image analysis-a novel approach to pigment content characteristics in melanoma cells. B16 F10 wt and Ctrl shRNA lines showed intermediate pigmentation, whereas the pigmentation of the B16 F10-derived cell lines was inversely correlated with the PPARα expression level. We observed that cells overexpressing PPARα were almost amelanotic and cells with reduced PPARα protein level were heavily melanized. Furthermore, fenofibrate down-regulated the melanogenic apparatus (MITF, tyrosinase, and tyrosinase-related proteins) in the cells with the regular PPARα expression level resulting in their visibly lower total melanin content in all the cell lines. From these observations, we conclude that fenofibrate works as a strong depigmenting agent, which acts independently of PPARα, but in an additive fashion. Our results also indicate that alterations in PGC-1a acetylation and expression level might contribute to the regulation of melanogenesis by PPARα and fenofibrate.
Collapse
|
21
|
Bezafibrate Attenuates Pressure Overload-Induced Cardiac Hypertrophy and Fibrosis. PPAR Res 2017; 2017:5789714. [PMID: 28127304 PMCID: PMC5239981 DOI: 10.1155/2017/5789714] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/30/2016] [Accepted: 11/21/2016] [Indexed: 11/24/2022] Open
Abstract
Background. Peroxisome proliferator-activated receptor-α (PPAR-α) is closely associated with the development of cardiac hypertrophy. Previous studies have indicated that bezafibrate (BZA), a PPAR-α agonist, could attenuate insulin resistance and obesity. This study was designed to determine whether BZA could protect against pressure overload-induced cardiac hypertrophy. Methods. Mice were orally given BZA (100 mg/kg) for 7 weeks beginning 1 week after aortic banding (AB) surgery. Cardiac hypertrophy was assessed based on echocardiographic, histological, and molecular aspects. Moreover, neonatal rat ventricular cardiomyocytes (NRVMs) were used to investigate the effects of BZA on the cardiomyocyte hypertrophic response in vitro. Results. Our study demonstrated that BZA could alleviate cardiac hypertrophy and fibrosis in mice subjected to AB surgery. BZA treatment also reduced the phosphorylation of protein kinase B (AKT)/glycogen synthase kinase-3β (GSK3β) and mitogen-activated protein kinases (MAPKs). BZA suppressed phenylephrine- (PE-) induced hypertrophy of cardiomyocyte in vitro. The protective effects of BZA were abolished by the treatment of the PPAR-α antagonist in vitro. Conclusions. BZA could attenuate pressure overload-induced cardiac hypertrophy and fibrosis.
Collapse
|
22
|
Wright SK, Wuertz BR, Harris G, Abu Ghazallah R, Miller WA, Gaffney PM, Ondrey FG. Functional activation of PPARγ in human upper aerodigestive cancer cell lines. Mol Carcinog 2016; 56:149-162. [PMID: 26999671 DOI: 10.1002/mc.22479] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 02/21/2016] [Accepted: 03/01/2016] [Indexed: 11/07/2022]
Abstract
Upper aerodigestive cancer is an aggressive malignancy with relatively stagnant long-term survival rates over 20 yr. Recent studies have demonstrated that exploitation of PPARγ pathways may be a novel therapy for cancer and its prevention. We tested whether PPARγ is expressed and inducible in aerodigestive carcinoma cells and whether it is present in human upper aerodigestive tumors. Human oral cancer CA-9-22 and NA cell lines were treated with the PPAR activators eicosatetraynoic acid (ETYA), 15-deoxy-δ- 12,14-prostaglandin J2 (PG-J2), and the thiazolidinedione, ciglitazone, and evaluated for their ability to functionally activate PPARγ luciferase reporter gene constructs. Cellular proliferation and clonogenic potential after PPARγ ligand treatment were also evaluated. Aerodigestive cancer specimens and normal tissues were evaluated for PPARγ expression on gene expression profiling and immunoblotting. Functional activation of PPARγ reporter gene constructs and increases in PPARγ protein were confirmed in the nuclear compartment after PPARγ ligand treatment. Significant decreases in cell proliferation and clonogenic potential resulted from treatment. Lipid accumulation was induced by PPARγ activator treatment. 75% of tumor specimens and 100% of normal control tissues expressed PPARγ RNA, and PPARγ protein was confirmed in 66% of tumor specimens analyzed by immunoblotting. We conclude PPARγ can be functionally activated in upper aerodigestive cancer and that its activation downregulates several features of the neoplastic phenotype. PPARγ expression in human upper aerodigestive tract tumors and normal cells potentially legitimizes it as a novel intervention target in this disease. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Simon K Wright
- Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota
| | - Beverly R Wuertz
- Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota
| | - George Harris
- Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota
| | - Raed Abu Ghazallah
- Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota
| | - Wendy A Miller
- Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota
| | - Patrick M Gaffney
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Frank G Ondrey
- Department of Otolaryngology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
23
|
Ren H, Wang G, Chen L, Jiang J, Liu L, Li N, Zhao J, Sun X, Zhou P. Genome-wide analysis of long non-coding RNAs at early stage of skin pigmentation in goats (Capra hircus). BMC Genomics 2016; 17:67. [PMID: 26785828 PMCID: PMC4719336 DOI: 10.1186/s12864-016-2365-3] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 01/05/2016] [Indexed: 01/10/2023] Open
Abstract
Background Long noncoding RNAs (lncRNAs) play roles in almost all biological processes; however, their function and profile in skin development and pigmentation is less understood. In addition, because lncRNAs are species-specific, their function in goats has not been established. Result We systematically identified lncRNAs in 100-day-old fetal skin by deep RNA-sequencing using the Youzhou dark goat (dark skin) and Yudong white goat (white skin) as a model of skin pigmentation. A total of 841,895,634 clean reads were obtained from six libraries (samples). We identified 1336 specific lncRNAs in fetal skin that belonged to three subtypes, including 999 intergenic lncRNAs (lincRNAs), 218 anti-sense lncRNAs, and 119 intronic lncRNAs. Our results demonstrated significant differences in gene architecture and expression among the three lncRNA subtypes, particularly in terms of density and position bias of transpose elements near the transcription start site. We also investigated the impact of lncRNAs on its target genes in cis and trans, indicating that these lncRNAs have a strict tissue specificity and functional conservation during skin development and pigmentation. Conclusion The present study provides a resource for lncRNA studies in diseases involved in pigmentation and skin development. It expands our knowledge about lncRNA biology as well as contributes to the annotation of the goat genome. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2365-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hangxing Ren
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China. .,Chongqing Engineering Research Center for Goats, Chongqing, 402460, China.
| | - Gaofu Wang
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China. .,Chongqing Engineering Research Center for Goats, Chongqing, 402460, China.
| | - Lei Chen
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China.
| | - Jing Jiang
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China. .,Chongqing Engineering Research Center for Goats, Chongqing, 402460, China.
| | - Liangjia Liu
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China. .,Chongqing Engineering Research Center for Goats, Chongqing, 402460, China.
| | - Nianfu Li
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China. .,Youyang Animal Husbandry Bureau, Chongqing, 409800, China.
| | - Jinhong Zhao
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China. .,Chongqing Engineering Research Center for Goats, Chongqing, 402460, China.
| | - Xiaoyan Sun
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China. .,Chongqing Engineering Research Center for Goats, Chongqing, 402460, China.
| | - Peng Zhou
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China. .,Chongqing Engineering Research Center for Goats, Chongqing, 402460, China.
| |
Collapse
|
24
|
Yang CH, Huang YC, Tsai ML, Cheng CY, Liu LL, Yen YW, Chen WL. Inhibition of melanogenesis by β-caryophyllene from lime mint essential oil in mouse B16 melanoma cells. Int J Cosmet Sci 2015; 37:550-4. [PMID: 25819153 DOI: 10.1111/ics.12224] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 03/07/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Volatile essential oils of mint species are used for cosmetics and in skin care products. In this study, we evaluated the main chemical components of the lime mint and the anti-melanogenic properties of its main components. METHODS The essential oil was analysed by gas chromatography-mass spectrometry (GC/MS). The anti-melanogenic effects of mint essential oil and β-caryophyllene were investigated in B16F10 murine melanoma cells. RESULTS The main components of lime mint essential oil were found to be D-limonene (41.10%), D-carvone (8.58%), δ-selinene (6.73%) and β-caryophyllene (6.24%). The lime mint essential oil reduced melanin production in a dose-dependent manner in murine B16F10 cells. β-Caryophyllene, one of the main compounds in lime mint essential oil, could reduce melanogenesis by down-regulating the expression of MITF, TRP-1, TRP-2 and tyrosinase, resulting in a decrease in melanin content decrease. CONCLUSION These results reveal that lime mint essential oil and β-caryophyllene are considered to be valuable as potential skin-whitening agents.
Collapse
Affiliation(s)
- C-H Yang
- Department of Cosmetic Science, Providence University, Taichung, 43301, Taiwan.,Department of Applied Chemistry, Providence University, Taichung, 43301, Taiwan
| | - Y-C Huang
- Department of Cosmetic Science, Providence University, Taichung, 43301, Taiwan
| | - M-L Tsai
- Department of Cosmetic Science, Chia Nan University of Pharmacy & Science, Tainan, 71710, Taiwan
| | - C-Y Cheng
- Department of Cosmetic Science, Providence University, Taichung, 43301, Taiwan.,Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, 40227, Taiwan
| | - L-L Liu
- Department of Cosmetic Science, Providence University, Taichung, 43301, Taiwan
| | - Y-W Yen
- Department of Cosmetic Science, Chia Nan University of Pharmacy & Science, Tainan, 71710, Taiwan
| | - W-L Chen
- Department of Cosmetic Science, Providence University, Taichung, 43301, Taiwan.,Department of Applied Chemistry, Providence University, Taichung, 43301, Taiwan
| |
Collapse
|
25
|
Hair dyes resorcinol and lawsone reduce production of melanin in melanoma cells by tyrosinase activity inhibition and decreasing tyrosinase and microphthalmia-associated transcription factor (MITF) expression. Int J Mol Sci 2015; 16:1495-508. [PMID: 25584612 PMCID: PMC4307316 DOI: 10.3390/ijms16011495] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/04/2015] [Indexed: 12/20/2022] Open
Abstract
Hair coloring products are one of the most important cosmetics for modern people; there are three major types of hair dyes, including the temporary, semi-permanent and permanent hair dyes. The selected hair dyes (such as ammonium persulfate, sodium persulfate, resorcinol and lawsone) are the important components for hair coloring products. Therefore, we analyzed the effects of these compounds on melanogenesis in B16-F10 melanoma cells. The results proved that hair dyes resorcinol and lawsone can reduce the production of melanin. The results also confirmed that resorcinol and lawsone inhibit mushroom and cellular tyrosinase activities in vitro. Resorcinol and lawsone can also downregulate the protein levels of tyrosinase and microphthalmia-associated transcription factor (MITF) in B16-F10 cells. Thus, we suggest that frequent use of hair dyes may have the risk of reducing natural melanin production in hair follicles. Moreover, resorcinol and lawsone may also be used as hypopigmenting agents to food, agricultural and cosmetic industry in the future.
Collapse
|
26
|
Nan YM, Wang RQ, Fu N. Peroxisome proliferator-activated receptor α, a potential therapeutic target for alcoholic liver disease. World J Gastroenterol 2014; 20:8055-8060. [PMID: 25009377 PMCID: PMC4081676 DOI: 10.3748/wjg.v20.i25.8055] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/02/2014] [Accepted: 03/13/2014] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver injury represents a progressive process with a range of consequences including hepatic steatosis, steatohepatitis, liver fibrosis, cirrhosis, and hepatocellular carcinoma. Targeting key molecular regulators involved in the development of alcoholic liver injury may be of great value in the prevention of liver injury. Peroxisome proliferator-activated receptor α (PPARα) plays a pivotal role in modulation of hepatic lipid metabolism, oxidative stress, inflammatory response and fibrogenesis. As such, PPARα may be a potential therapeutic target for the treatment of alcoholic liver disease.
Collapse
|
27
|
Chiang HM, Chien YC, Wu CH, Kuo YH, Wu WC, Pan YY, Su YH, Wen KC. Hydroalcoholic extract of Rhodiola rosea L. (Crassulaceae) and its hydrolysate inhibit melanogenesis in B16F0 cells by regulating the CREB/MITF/tyrosinase pathway. Food Chem Toxicol 2013; 65:129-39. [PMID: 24380755 DOI: 10.1016/j.fct.2013.12.032] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 12/17/2013] [Accepted: 12/19/2013] [Indexed: 11/28/2022]
Abstract
We investigated the effects of an aqueous alcohol extract of Rhodiola rosea (R. rosea) and its hydrolysate on melanin synthesis and the mechanisms mediating the activity. The ratio of tyrosol to salidroside was 2.3 in hydroalcoholic extract, and 51.0 in hydrolysate. We found that R. rosea extract and its hydrolysate inhibited melanin synthesis and tyrosinase activity in mouse melanoma cells (B16F0 cells). R. rosea extract also inhibited gene and protein expression of melanocortin 1 receptor (MC1R) and inhibited c-AMP response element binding protein (CREB) phosphorylation, suppressed the activation of AKT and glycogen synthase kinase-3 beta (GSK3β), and inhibited the expression of microphthalmia-associated transcription factor (MITF) and tyrosinase-related protein 1 (TRP-1). R. rosea hydrolysate inhibited the phosphorylation of CREB, the activation of AKT and GSK3β, and the expression of MITF and tyrosinase. Our results suggest that R. rosea extract is a novel tyrosinase inhibitor and that it exerts its effects by regulating the CREB/MITF/tyrosinase pathway in B16F0. Further in vivo studies are needed to determine the effectiveness of R. rosea extract as a skin whitening agent.
Collapse
Affiliation(s)
- Hsiu-Mei Chiang
- Department of Cosmeceutics, China Medical University, Taichung 404, Taiwan.
| | - Yin-Chih Chien
- Department of Cosmeceutics, China Medical University, Taichung 404, Taiwan; Graduate Institute of Pharmacy, China Medical University, Taichung, Taiwan
| | - Chieh-Hsi Wu
- Graduate Institute of Pharmacy, China Medical University, Taichung, Taiwan
| | - Yueh-Hsiung Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 404, Taiwan; Department of Biotechnology, Asia University, Taichung 413, Taiwan
| | - Wan-Chen Wu
- Department of Cosmeceutics, China Medical University, Taichung 404, Taiwan
| | - Yu-Yun Pan
- Department of Cosmeceutics, China Medical University, Taichung 404, Taiwan
| | - Yu-Han Su
- Department of Cosmeceutics, China Medical University, Taichung 404, Taiwan
| | - Kuo-Ching Wen
- Department of Cosmeceutics, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|