1
|
Al Subait A, Alghamdi RH, Ali R, Alsharidah A, Huwaizi S, Alkhodier RA, Almogren AS, Alzomia BA, Alaskar A, Boudjelal M. Discovery of PPAR Alpha Lipid Pathway Modulators That Do Not Bind Directly to the Receptor as Potential Anti-Cancer Compounds. Int J Mol Sci 2025; 26:736. [PMID: 39859448 PMCID: PMC11766124 DOI: 10.3390/ijms26020736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are considered good drug targets for breast cancer because of their involvement in fatty acid metabolism that induces cell proliferation. In this study, we used the KAIMRC1 breast cancer cell line. We showed that the PPARE-Luciferase reporter gets highly activated without adding any exogenous ligand when PPAR alpha is co-transfected, and the antagonist GW6471 can inhibit the activity. Using this reporter system, we screened 240 compounds representing kinase inhibitors, epigenetic modulators, and stem cell differentiators and identified compounds that inhibit the PPARα-activated PPARE-Luciferase reporter in the KAIMRC1 cell. We selected 11 compounds (five epigenetic modulators, two stem cell differentiators, and four kinase inhibitors) that inhibited the reporter by at least 40% compared to the controls (DMSO-treated cells). We tested them in a dose-dependent manner and measured the KAIMRC1 cell viability after 48 h. All 11 compounds induced the cell killing at different IC50 values. We selected two compounds, PHA665752 and NSC3852, to dissect how they kill KAIMRC1 cells compared to the antagonist GW6741. First, molecular docking and a TR-FRET PPARα binding assay showed that compared to GW6471, these two compounds could not bind to PPARα. This means they inhibit the PPARα pathway independently rather than binding to the receptor. We further confirmed that PHA665752 and NSC3852 induce cell killing depending on the level of PPARα expression, and as such, their potency for killing the SW620 colon cancer cell line that expresses the lowest level of PPARα was less potent than for the KAIMRC1 and MDA-MB-231 cell lines. Further, using an apoptosis array and fatty acid gene expression panel, we found that both compounds regulate the PPARα pathway by controlling the genes involved in the fatty acid oxidation process. Our findings suggest that these two compounds have opposite effects involving fatty acid oxidation in the KAIMRC1 breast cancer cell line. Although we do not fully understand their mechanism of action, our data provide new insights into the potential role of these compounds in targeting breast cancer cells.
Collapse
Affiliation(s)
- Arwa Al Subait
- Medical Research Core Facility and Platforms (MRCFP)-Drug Discovery Platform, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGHA), Riyadh 11481, Saudi Arabia; (A.A.S.)
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGHA), Riyadh 11481, Saudi Arabia
| | - Raghad H. Alghamdi
- King Abdulaziz and His Companions Foundation for Giftedness and Creativity (MAWHIBA), Riyadh 11481, Saudi Arabia;
| | - Rizwan Ali
- Medical Research Core Facility and Platforms (MRCFP)-Drug Discovery Platform, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGHA), Riyadh 11481, Saudi Arabia; (A.A.S.)
| | - Amani Alsharidah
- College of Science, King Saud University, Riyadh 11459, Saudi Arabia;
| | - Sarah Huwaizi
- Medical Research Core Facility and Platforms (MRCFP)-Drug Discovery Platform, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGHA), Riyadh 11481, Saudi Arabia; (A.A.S.)
| | - Reem A. Alkhodier
- Department of Pharmaceutical Sciences, College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGHA), Riyadh 11481, Saudi Arabia
| | - Aljawharah Saud Almogren
- Medical Research Core Facility and Platforms (MRCFP)-Drug Discovery Platform, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGHA), Riyadh 11481, Saudi Arabia; (A.A.S.)
| | - Barrak A. Alzomia
- Medical Research Core Facility and Platforms (MRCFP)-Drug Discovery Platform, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGHA), Riyadh 11481, Saudi Arabia; (A.A.S.)
| | - Ahmad Alaskar
- Medical Research Core Facility and Platforms (MRCFP)-Drug Discovery Platform, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGHA), Riyadh 11481, Saudi Arabia; (A.A.S.)
| | - Mohamed Boudjelal
- Medical Research Core Facility and Platforms (MRCFP)-Drug Discovery Platform, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs (MNGHA), Riyadh 11481, Saudi Arabia; (A.A.S.)
| |
Collapse
|
2
|
Ammazzalorso A, Tacconelli S, Contursi A, Hofling U, Cerchia C, Di Berardino S, De Michele A, Amoroso R, Lavecchia A, Patrignani P. A sulfonimide derivative of bezafibrate as a dual inhibitor of cyclooxygenase-2 and PPARα. Front Pharmacol 2024; 15:1488722. [PMID: 39660001 PMCID: PMC11628281 DOI: 10.3389/fphar.2024.1488722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024] Open
Abstract
Background PPARα and cyclooxygenase (COX)-2 are overexpressed in certain types of cancer. Thus, developing a dual inhibitor that targets both could be more effective as an anticancer agent than single inhibitors. We have previously shown that an analog of the bezafibrate named AA520 is a PPARα antagonist. Herein, we report the identification of AA520 as a potent COX-2 inhibitor using in silico approaches. In addition, we performed a thorough pharmacological characterization of AA520 towards COX-1 and COX-2 in different in vitro models. Methods AA520 was characterized for inhibiting platelet COX-1 and monocyte COX-2 activity in human whole blood (HWB) and for effects on lipidomics of eicosanoids using LC-MS/MS. The kinetics of the interaction of AA520 with COX-2 was assessed in the human colon cancer cell line, HCA-7, expressing only COX-2, by testing the COX-2 activity after extensive washing of the cells. The impact of AA520 on cancer cell viability, metabolic activity, and cytotoxicity was tested using the MTT reagent. Results In HWB, AA520 inhibited in a concentration-dependent fashion LPS-stimulated leukocyte prostaglandin (PG) E2 generation with an IC50 of 0.10 (95% CI: 0.05-0.263) μM while platelet COX-1 was not affected up to 300 μM. AA520 did not affect LPS-induced monocyte COX-2 expression, and other eicosanoids generated by enzymatic and nonenzymatic pathways. AA520 inhibited COX-2-dependent PGE2 generation in the colon cancer cell line HCA7. Comparison of the inhibition of COX-2 and its reversibility by AA520, indomethacin (a time-dependent inhibitor), acetylsalicylic acid (ASA) (an irreversible inhibitor), and ibuprofen (a reversible inhibitor) showed that the compound is acting by forming a tightly bound COX-2 interaction. This was confirmed by docking and molecular dynamics studies. Moreover, AA520 (1 μM) significantly reduced MTT in HCA7 cells. Conclusion We have identified a highly selective COX-2 inhibitor with a unique scaffold. This inhibitor retains PPARα antagonism at the same concentration range. It has the potential to be effective in treating certain types of cancer, such as hepatocellular carcinoma (HCC) and renal cell carcinoma (RCC), where COX-2 and PPARα are overexpressed.
Collapse
Affiliation(s)
| | - Stefania Tacconelli
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University, Chieti, Italy
| | - Annalisa Contursi
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University, Chieti, Italy
| | - Ulrika Hofling
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University, Chieti, Italy
| | - Carmen Cerchia
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Naples, Italy
| | - Sara Di Berardino
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University, Chieti, Italy
| | - Alessandra De Michele
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University, Chieti, Italy
| | - Rosa Amoroso
- Department of Pharmacy, “G. d’Annunzio” University, Chieti, Italy
| | - Antonio Lavecchia
- Department of Pharmaceutical and Toxicological Chemistry, University of Naples “Federico II”, Naples, Italy
| | - Paola Patrignani
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), and Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University, Chieti, Italy
| |
Collapse
|
3
|
Moreno-Rodríguez N, Laghezza A, Cerchia C, Sokolova DV, Spirina TS, De Filippis B, Romanelli V, Recio R, Fernández I, Loiodice F, Pokrovsky VS, Ammazzalorso A, Lavecchia A. Synthesis and in vitro cytotoxicity of benzoxazole-based PPARα/γ antagonists in colorectal cancer cell lines. Arch Pharm (Weinheim) 2024; 357:e2400086. [PMID: 38807029 DOI: 10.1002/ardp.202400086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/30/2024]
Abstract
A series of benzoxazole-based amides and sulfonamides were synthesized and evaluated for their human peroxisome proliferator-activated receptor (PPAR)α and PPARγ activity. All tested compounds showed a dual antagonist profile on both PPAR subtypes; based on transactivation results, seven compounds were selected to test their in vitro antiproliferative activity in a panel of eight cancer cell lines with different expression rates of PPARα and PPARγ. 3f was identified as the most cytotoxic compound, with higher potency in the colorectal cancer cell lines HT-29 and HCT116. Compound 3f induced a concentration-dependent activation of caspases and cell-cycle arrest in both colorectal cancer models. Docking experiments were also performed to shed light on the putative binding mode of this novel class of dual PPARα/γ antagonists.
Collapse
Affiliation(s)
- Nazaret Moreno-Rodríguez
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Antonio Laghezza
- Department of Pharmacy-Drug Science, University of Bari "Aldo Moro", Bari, Italy
| | - Carmen Cerchia
- "Drug Discovery" Laboratory, Department of Pharmacy, University of Napoli "Federico II", Napoli, Italy
| | - Darina V Sokolova
- Research, Institute of Experimental Therapy and Diagnostics of Tumor, NN Blokhin National Medical Center of Oncology, Moscow, Russia
- Department of Biochemistry, Patrice Lumumba Peoples' Friendship University, Moscow, Russia
| | - Tatiana S Spirina
- Research, Institute of Experimental Therapy and Diagnostics of Tumor, NN Blokhin National Medical Center of Oncology, Moscow, Russia
- Department of Biochemistry, Patrice Lumumba Peoples' Friendship University, Moscow, Russia
| | - Barbara De Filippis
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Virgilio Romanelli
- "Drug Discovery" Laboratory, Department of Pharmacy, University of Napoli "Federico II", Napoli, Italy
| | - Rocío Recio
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Inmaculada Fernández
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Fulvio Loiodice
- Department of Pharmacy-Drug Science, University of Bari "Aldo Moro", Bari, Italy
| | - Vadim S Pokrovsky
- Research, Institute of Experimental Therapy and Diagnostics of Tumor, NN Blokhin National Medical Center of Oncology, Moscow, Russia
- Department of Biochemistry, Patrice Lumumba Peoples' Friendship University, Moscow, Russia
| | | | - Antonio Lavecchia
- "Drug Discovery" Laboratory, Department of Pharmacy, University of Napoli "Federico II", Napoli, Italy
| |
Collapse
|
4
|
Xiang X, Li F, Zhou S, Zeng Y, Deng X, Zhang H, Li J, Liu H, Rao J, Gao L, Zhang C, Wen Q, Gao L, Zhang X. Significance of PPARA as a Treatment Target for Chronic Lymphocytic Leukemia. PPAR Res 2023; 2023:8456833. [PMID: 37404899 PMCID: PMC10317583 DOI: 10.1155/2023/8456833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/28/2023] [Accepted: 05/24/2023] [Indexed: 07/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor alpha (PPARA) has been suggested as a therapeutic target for chronic lymphocytic leukemia (CLL). However, the underlying molecular mechanism remains largely unclear. In this study, we analyzed DNA next-generation sequencing (NGS) data and clinical information from 86 CLL patients to identify gene markers related to treatment-free survival (TFS) length. We then constructed a genetic network that includes CLL promoters, treatment targets, and TFS-related marker genes. To assess the significance of PPARA within the network, we utilized degree centrality (DC) and pathway enrichment score (EScore). Clinical and NGS data revealed 10 TFS length-related gene markers, including RPS15, FOXO1, FBXW7, KMT2A, NOTCH1, GNA12, EGR2, GNA13, KDM6A, and ATM. Through literature data mining, 83 genes were identified as CLL upstream promoters and treatment targets. Among them, PPARA exhibited a stronger connection to CLL and TFS-related gene markers, as evidenced by its ranking at No. 13 based on DC, compared to most of the other promoters (>84%). Additionally, PPARA co-functions with 70 out of 92 in-network genes in various functional pathways/gene groups related to CLL pathology, such as regulation of cell adhesion, inflammation, reactive oxygen species, and cell differentiation. Based on our findings, PPARA is considered one of the critical genes within a large genetic network that influences the prognosis and TFS of CLL through multiple pathogenic pathways.
Collapse
Affiliation(s)
- Xixi Xiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Fu Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Sha Zhou
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Yunjing Zeng
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Xiaojuan Deng
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Hongyang Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Jiali Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Hongyun Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Jun Rao
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Lei Gao
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Cheng Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Qin Wen
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Li Gao
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| | - Xi Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Key Subject of Chongqing, Chongqing 400037, China
| |
Collapse
|
5
|
Recinella L, De Filippis B, Libero ML, Ammazzalorso A, Chiavaroli A, Orlando G, Ferrante C, Giampietro L, Veschi S, Cama A, Mannino F, Gasparo I, Bitto A, Amoroso R, Brunetti L, Leone S. Anti-Inflammatory, Antioxidant, and WAT/BAT-Conversion Stimulation Induced by Novel PPAR Ligands: Results from Ex Vivo and In Vitro Studies. Pharmaceuticals (Basel) 2023; 16:346. [PMID: 36986448 PMCID: PMC10056895 DOI: 10.3390/ph16030346] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Activation of peroxisome proliferator-activated receptors (PPARs) not only regulates multiple metabolic pathways, but mediates various biological effects related to inflammation and oxidative stress. We investigated the effects of four new PPAR ligands containing a fibrate scaffold-the PPAR agonists (1a (αEC50 1.0 μM) and 1b (γEC50 0.012 μM)) and antagonists (2a (αIC50 6.5 μM) and 2b (αIC50 0.98 μM, with a weak antagonist activity on γ isoform))-on proinflammatory and oxidative stress biomarkers. The PPAR ligands 1a-b and 2a-b (0.1-10 μM) were tested on isolated liver specimens treated with lipopolysaccharide (LPS), and the levels of lactate dehydrogenase (LDH), prostaglandin (PG) E2, and 8-iso-PGF2α were measured. The effects of these compounds on the gene expression of the adipose tissue markers of browning, PPARα, and PPARγ, in white adipocytes, were evaluated as well. We found a significant reduction in LPS-induced LDH, PGE2, and 8-iso-PGF2α levels after 1a treatment. On the other hand, 1b decreased LPS-induced LDH activity. Compared to the control, 1a stimulated uncoupling protein 1 (UCP1), PR-(PRD1-BF1-RIZ1 homologous) domain containing 16 (PRDM16), deiodinase type II (DIO2), and PPARα and PPARγ gene expression, in 3T3-L1 cells. Similarly, 1b increased UCP1, DIO2, and PPARγ gene expression. 2a-b caused a reduction in the gene expression of UCP1, PRDM16, and DIO2 when tested at 10 μM. In addition, 2a-b significantly decreased PPARα gene expression. A significant reduction in PPARγ gene expression was also found after 2b treatment. The novel PPARα agonist 1a might be a promising lead compound and represents a valuable pharmacological tool for further assessment. The PPARγ agonist 1b could play a minor role in the regulation of inflammatory pathways.
Collapse
Affiliation(s)
- Lucia Recinella
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | | | | | | | | | - Giustino Orlando
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | - Claudio Ferrante
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | | | - Serena Veschi
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | - Alessandro Cama
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | - Federica Mannino
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Irene Gasparo
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Rosa Amoroso
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | - Luigi Brunetti
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| | - Sheila Leone
- Department of Pharmacy, G. d’Annunzio University, 66100 Chieti, Italy
| |
Collapse
|
6
|
PPAR Ligands Induce Antiviral Effects Targeting Perturbed Lipid Metabolism during SARS-CoV-2, HCV, and HCMV Infection. BIOLOGY 2022; 11:biology11010114. [PMID: 35053112 PMCID: PMC8772958 DOI: 10.3390/biology11010114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 12/24/2022]
Abstract
Simple Summary The current coronavirus disease 2019 pandemic turned the attention of researchers to developing novel strategies to counteract virus infections. Despite several antiviral drugs being commercially available, there is an urgent need to identify novel molecules efficacious against viral infections that act through different mechanisms of action. In this context, our attention is focused on novel compounds acting on nuclear receptors, whose activity could be beneficial in viral infections, including coronavirus, hepatitis C virus, and cytomegalovirus. Abstract The manipulation of host metabolisms by viral infections has been demonstrated by several studies, with a marked influence on the synthesis and utilization of glucose, nucleotides, fatty acids, and amino acids. The ability of virus to perturb the metabolic status of the infected organism is directly linked to the outcome of the viral infection. A great deal of research in recent years has been focusing on these metabolic aspects, pointing at modifications induced by virus, and suggesting novel strategies to counteract the perturbed host metabolism. In this review, our attention is turned on PPARs, nuclear receptors controlling multiple metabolic actions, and on the effects played by PPAR ligands during viral infections. The role of PPAR agonists and antagonists during SARS-CoV-2, HCV, and HCMV infections will be analyzed.
Collapse
|
7
|
Ammazzalorso A, Bruno I, Florio R, De Lellis L, Laghezza A, Cerchia C, De Filippis B, Fantacuzzi M, Giampietro L, Maccallini C, Tortorella P, Veschi S, Loiodice F, Lavecchia A, Cama A, Amoroso R. Sulfonimide and Amide Derivatives as Novel PPARα Antagonists: Synthesis, Antiproliferative Activity, and Docking Studies. ACS Med Chem Lett 2020; 11:624-632. [PMID: 32435362 PMCID: PMC7236056 DOI: 10.1021/acsmedchemlett.9b00666] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/03/2020] [Indexed: 12/30/2022] Open
Abstract
An agonist-antagonist switching strategy was performed to discover novel PPARα antagonists. Phenyldiazenyl derivatives of fibrates were developed, bearing sulfonimide or amide functional groups. A second series of compounds was synthesized, replacing the phenyldiazenyl moiety with amide or urea portions. Final compounds were screened by transactivation assay, showing good PPARα antagonism and selectivity at submicromolar concentrations. When tested in cancer cell models expressing PPARα, selected derivatives induced marked effects on cell viability. Notably, 3c, 3d, and 10e displayed remarkable antiproliferative effects in two paraganglioma cell lines, with CC50 lower than commercial PPARα antagonist GW6471 and a negligible toxicity on normal fibroblast cells. Docking studies were also performed to elucidate the binding mode of these compounds and to help interpretation of SAR data.
Collapse
Affiliation(s)
- Alessandra Ammazzalorso
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Isabella Bruno
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Rosalba Florio
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Laura De Lellis
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Antonio Laghezza
- Department of Pharmacy-Drug Science, University of Bari “Aldo Moro”, Via E. Orabona 4, 70126 Bari, Italy
| | - Carmen Cerchia
- Department of Pharmacy, “Drug Discovery” Laboratory, University of Napoli “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy
| | - Barbara De Filippis
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Marialuigia Fantacuzzi
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Letizia Giampietro
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Cristina Maccallini
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Paolo Tortorella
- Department of Pharmacy-Drug Science, University of Bari “Aldo Moro”, Via E. Orabona 4, 70126 Bari, Italy
| | - Serena Veschi
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Fulvio Loiodice
- Department of Pharmacy-Drug Science, University of Bari “Aldo Moro”, Via E. Orabona 4, 70126 Bari, Italy
| | - Antonio Lavecchia
- Department of Pharmacy, “Drug Discovery” Laboratory, University of Napoli “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy
| | - Alessandro Cama
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
- Center for Advanced Studies and Technology CAST, Via Luigi Polacchi 11, 66100 Chieti, Italy
| | - Rosa Amoroso
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
8
|
Liu Y, Chen C, Sun LY, Gao H, Zhen JB, Yang KW. meta-Substituted benzenesulfonamide: a potent scaffold for the development of metallo-β-lactamase ImiS inhibitors. RSC Med Chem 2020; 11:259-267. [PMID: 33479632 PMCID: PMC7412727 DOI: 10.1039/c9md00455f] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/26/2019] [Indexed: 01/25/2023] Open
Abstract
Metallo-β-lactamase (MβL) ImiS contributes to the emergence of carbapenem resistance. A potent scaffold, meta-substituted benzenesulfonamide, was constructed and assayed against MβLs. The twenty-one obtained molecules specifically inhibited ImiS (IC50 = 0.11-9.3 μM); 2g was found to be the best inhibitor (IC50 = 0.11 μM), and 1g and 2g exhibited partially mixed inhibition with K i of 8.0 and 0.55 μM. The analysis of the structure-activity relationship revealed that the meta-substitutes improved the inhibitory activity of the inhibitors. Isothermal titration calorimetry (ITC) assays showed that 2g reversibly inhibited ImiS. The benzenesulfonamides exhibited synergistic antibacterial effects against E. coli BL21 (DE3) cells with ImiS, resulting in a 2-4-fold reduction in the MIC of imipenem and meropenem. Also, mouse experiments showed that 2g had synergistic efficacy with meropenem and significantly reduced the bacterial load in the spleen and liver after a single intraperitoneal dose. Tracing the ImiS in living E. coli cells by RS at a super-resolution level (3D-SIM) showed that the target was initially associated on the surface of the cells, then there was a high density of uniform localization distributed in the cytosol of cells, and it finally accumulated in the formation of inclusion bodies at the cell poles. Docking studies suggested that the sulfonamide group acted as a zinc-binding group to coordinate with Zn(ii) and the residual amino acid within the CphA active center, tightly anchoring the inhibitor at the active site. This study provides a highly promising scaffold for the development of inhibitors of ImiS, even the B2 subclasses of MβLs.
Collapse
Affiliation(s)
- Ya Liu
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education , College of Chemistry and Materials Science , Northwest University , Xi'an 710127 , P. R. China .
| | - Cheng Chen
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education , College of Chemistry and Materials Science , Northwest University , Xi'an 710127 , P. R. China .
| | - Le-Yun Sun
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education , College of Chemistry and Materials Science , Northwest University , Xi'an 710127 , P. R. China .
| | - Han Gao
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education , College of Chemistry and Materials Science , Northwest University , Xi'an 710127 , P. R. China .
| | - Jian-Bin Zhen
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education , College of Chemistry and Materials Science , Northwest University , Xi'an 710127 , P. R. China .
| | - Ke-Wu Yang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education , College of Chemistry and Materials Science , Northwest University , Xi'an 710127 , P. R. China .
| |
Collapse
|
9
|
Synthesis of novel benzothiazole amides: Evaluation of PPAR activity and anti-proliferative effects in paraganglioma, pancreatic and colorectal cancer cell lines. Bioorg Med Chem Lett 2019; 29:2302-2306. [PMID: 31272790 DOI: 10.1016/j.bmcl.2019.06.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/26/2022]
Abstract
The reduced activation of PPARs has a positive impact on cancer cell growth and viability in multiple preclinical tumor models, suggesting a new therapeutic potential for PPAR antagonists. In the present study, the benzothiazole amides 2a-g were synthesized and their activities on PPARs were investigated. Transactivation assay showed a moderate activity of the novel compounds as PPARα antagonists. Notably, in cellular assays they exhibited cytotoxicity in pancreatic, colorectal and paraganglioma cancer cells overexpressing PPARα. In particular, compound 2b showed the most remarkable inhibition of viability (greater than 90%) in two paraganglioma cell lines, with IC50 values in the low micromolar range. In addition, 2b markedly impaired colony formation capacity in the same cells. Taken together, these results show a relevant anti-proliferative potential of compound 2b, which appears particularly effective in paraganglioma, a rare tumor poorly responsive to chemotherapy.
Collapse
|
10
|
Ammazzalorso A, Carradori S, Angeli A, Akdemir A, De Filippis B, Fantacuzzi M, Giampietro L, Maccallini C, Amoroso R, Supuran CT. Fibrate-based N-acylsulphonamides targeting carbonic anhydrases: synthesis, biochemical evaluation, and docking studies. J Enzyme Inhib Med Chem 2019; 34:1051-1061. [PMID: 31074307 PMCID: PMC6522927 DOI: 10.1080/14756366.2019.1611801] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
A large library of fibrate-based N-acylsulphonamides was designed, synthesised, and fully characterised in order to propose them as zinc binders for the inhibition of human carbonic anhydrase (hCA) enzymatic activity. Synthesised compounds were tested against four hCAs (I, II, IX, and XII) revealing a promising submicromolar inhibitory activity characterised by an isozyme selectivity pattern. Structural modifications explored within this scaffold are: presence of an aryl ring on the sulphonamide, p-substitution of this aryl ring, benzothiazole or benzophenone as core nuclei, and an n-propyl chain or a geminal dimethyl at Cα carbon. Biological results fitted well with molecular modelling analyses, revealing a putative direct interaction with the zinc ion in the active site of hCA I, II and IX. These findings supported the exploration of less investigated secondary sulphonamides as potential hCA inhibitors.
Collapse
Affiliation(s)
| | - Simone Carradori
- a Department of Pharmacy , "G. d'Annunzio" University of Chieti-Pescara , Chieti , Italy
| | - Andrea Angeli
- b Laboratorio di Chimica Bioinorganica , Università degli Studi di Firenze , Florence , Italy
| | - Atilla Akdemir
- c Department of Pharmacology, Faculty of Pharmacy, Computer-Aided Drug Discovery Laboratory , Bezmialem Vakif University , Istanbul , Turkey
| | - Barbara De Filippis
- a Department of Pharmacy , "G. d'Annunzio" University of Chieti-Pescara , Chieti , Italy
| | - Marialuigia Fantacuzzi
- a Department of Pharmacy , "G. d'Annunzio" University of Chieti-Pescara , Chieti , Italy
| | - Letizia Giampietro
- a Department of Pharmacy , "G. d'Annunzio" University of Chieti-Pescara , Chieti , Italy
| | - Cristina Maccallini
- a Department of Pharmacy , "G. d'Annunzio" University of Chieti-Pescara , Chieti , Italy
| | - Rosa Amoroso
- a Department of Pharmacy , "G. d'Annunzio" University of Chieti-Pescara , Chieti , Italy
| | - Claudiu T Supuran
- b Laboratorio di Chimica Bioinorganica , Università degli Studi di Firenze , Florence , Italy.,d Neurofarba Department , Section of Pharmaceutical and Nutriceutical Sciences, Università degli Studi di Firenze , Florence , Italy
| |
Collapse
|
11
|
Development of Fibrates as Important Scaffolds in Medicinal Chemistry. ChemMedChem 2019; 14:1051-1066. [DOI: 10.1002/cmdc.201900128] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Indexed: 12/13/2022]
|
12
|
Ammazzalorso A, Amoroso R. Inhibition of PPARγ by Natural Compounds as a Promising Strategy in Obesity and Diabetes. THE OPEN MEDICINAL CHEMISTRY JOURNAL 2019. [DOI: 10.2174/1874104501913010007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A wide group of natural compounds (flavonoids, stilbenes, neolignans and others) has been identified as Peroxisome Proliferator-Activated Receptor (PPAR) agonists, with a large variety of chemical structure and different activity versus the three PPAR subtypes. These receptors are transcription factors controlling metabolic pathways in the organism, involved in lipid and glucose metabolism, cell differentiation and energy homeostasis. Otherwise, very little is known about natural compounds able to inhibit PPARs. A number of studies demonstrate that PPARγ repression has a beneficial effect in reducing body weight and improving insulin sensitivity, suggesting a potential clinical role in obesity and type 2 diabetes. This review analyzes natural compounds able to repress PPAR activity and their potential use in metabolic disorders.
Collapse
|
13
|
Jia WQ, Jing Z, Liu X, Feng XY, Liu YY, Wang SQ, Xu WR, Liu JW, Cheng XC. Virtual identification of novel PPARα/γ dual agonists by scaffold hopping of saroglitazar. J Biomol Struct Dyn 2017; 36:3496-3512. [DOI: 10.1080/07391102.2017.1392363] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Wen-Qing Jia
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Zhi Jing
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xin Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xiao-Yan Feng
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ya-Ya Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Shu-Qing Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wei-Ren Xu
- Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Jian-Wen Liu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xian-Chao Cheng
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
14
|
Ammazzalorso A, De Lellis L, Florio R, Bruno I, De Filippis B, Fantacuzzi M, Giampietro L, Maccallini C, Perconti S, Verginelli F, Cama A, Amoroso R. Cytotoxic effect of a family of peroxisome proliferator-activated receptor antagonists in colorectal and pancreatic cancer cell lines. Chem Biol Drug Des 2017; 90:1029-1035. [PMID: 28544586 DOI: 10.1111/cbdd.13026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/22/2022]
Abstract
Recent studies report an interesting role of peroxisome proliferator-activated receptor (PPAR) antagonists in different tumor models, being these compounds able to perturb metabolism and viability in cancer cells. In this work, the identification of a novel PPAR antagonist, showing inhibitory activity on PPARα and a weaker antagonism on PPARγ, is described. The activity of this compound and of a series of chemical analogues was investigated in selected tumor cell lines, expressing both PPARα and PPARγ. Data obtained show a dose-dependent cytotoxic effect of the novel PPAR antagonist in colorectal and pancreatic cancer models.
Collapse
Affiliation(s)
- Alessandra Ammazzalorso
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Laura De Lellis
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy.,Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Rosalba Florio
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy.,Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Isabella Bruno
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Barbara De Filippis
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Marialuigia Fantacuzzi
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Letizia Giampietro
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Cristina Maccallini
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| | - Silvia Perconti
- Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Fabio Verginelli
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy.,Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Alessandro Cama
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy.,Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Rosa Amoroso
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy
| |
Collapse
|
15
|
Ammazzalorso A, De Filippis B, Giampietro L, Amoroso R. N-acylsulfonamides: Synthetic routes and biological potential in medicinal chemistry. Chem Biol Drug Des 2017. [PMID: 28632928 DOI: 10.1111/cbdd.13043] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Sulfonamide is a common structural motif in naturally occurring and synthetic medicinal compounds. The rising interest in sulfonamides and N-acyl derivatives is attested by the large number of drugs and lead compounds identified in last years, explored in different fields of medicinal chemistry and showing biological activity. Many acylsulfonamide derivatives were designed and synthesized as isosteres of carboxylic acids, being the characteristics of these functional groups very close. Starting from chemical routes to N-acylsulfonamides, this review explores compounds of pharmaceutical interest, developed as enzymatic inhibitors or targeting receptors.
Collapse
Affiliation(s)
| | | | | | - Rosa Amoroso
- Dipartimento di Farmacia, Università G. d'Annunzio, Chieti, Italy
| |
Collapse
|
16
|
Florio R, De Lellis L, di Giacomo V, Di Marcantonio MC, Cristiano L, Basile M, Verginelli F, Verzilli D, Ammazzalorso A, Prasad SC, Cataldi A, Sanna M, Cimini A, Mariani-Costantini R, Mincione G, Cama A. Effects of PPARα inhibition in head and neck paraganglioma cells. PLoS One 2017; 12:e0178995. [PMID: 28594934 PMCID: PMC5464765 DOI: 10.1371/journal.pone.0178995] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 05/22/2017] [Indexed: 01/21/2023] Open
Abstract
Head and neck paragangliomas (HNPGLs) are rare tumors that may cause important morbidity, because of their tendency to infiltrate the skull base. At present, surgery is the only therapeutic option, but radical removal may be difficult or impossible. Thus, effective targets and molecules for HNPGL treatment need to be identified. However, the lack of cellular models for this rare tumor hampers this task. PPARα receptor activation was reported in several tumors and this receptor appears to be a promising therapeutic target in different malignancies. Considering that the role of PPARα in HNPGLs was never studied before, we analyzed the potential of modulating PPARα in a unique model of HNPGL cells. We observed an intense immunoreactivity for PPARα in HNPGL tumors, suggesting that this receptor has an important role in HNPGL. A pronounced nuclear expression of PPARα was also confirmed in HNPGL-derived cells. The specific PPARα agonist WY14643 had no effect on HNPGL cell viability, whereas the specific PPARα antagonist GW6471 reduced HNPGL cell viability and growth by inducing cell cycle arrest and caspase-dependent apoptosis. GW6471 treatment was associated with a marked decrease of CDK4, cyclin D3 and cyclin B1 protein expression, along with an increased expression of p21 in HNPGL cells. Moreover, GW6471 drastically impaired clonogenic activity of HNPGL cells, with a less marked effect on cell migration. Notably, the effects of GW6471 on HNPGL cells were associated with the inhibition of the PI3K/GSK3β/β-catenin signaling pathway. In conclusion, the PPARα antagonist GW6471 reduces HNPGL cell viability, interfering with cell cycle and inducing apoptosis. The mechanisms affecting HNPGL cell viability involve repression of the PI3K/GSK3β/β-catenin pathway. Therefore, PPARα could represent a novel therapeutic target for HNPGL.
Collapse
Affiliation(s)
- Rosalba Florio
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Unit of General Pathology, CeSI-MeT, “G. d’Annunzio” University, Chieti, Italy
| | - Laura De Lellis
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Unit of General Pathology, CeSI-MeT, “G. d’Annunzio” University, Chieti, Italy
- * E-mail: (LDL); (AC)
| | - Viviana di Giacomo
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Maria Carmela Di Marcantonio
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Loredana Cristiano
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Mariangela Basile
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Fabio Verginelli
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Unit of General Pathology, CeSI-MeT, “G. d’Annunzio” University, Chieti, Italy
| | - Delfina Verzilli
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | | | | | - Amelia Cataldi
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Mario Sanna
- Department of Otology and Skull Base Surgery, Gruppo Otologico, Piacenza, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, United States of America
- Gran Sasso National Laboratory (LNGS), National Institute for Nuclear Physics (INFN), Assergi, Italy
| | - Renato Mariani-Costantini
- Unit of General Pathology, CeSI-MeT, “G. d’Annunzio” University, Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Gabriella Mincione
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Alessandro Cama
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Unit of General Pathology, CeSI-MeT, “G. d’Annunzio” University, Chieti, Italy
- * E-mail: (LDL); (AC)
| |
Collapse
|
17
|
Chen Y, Wang Y, Huang Y, Zeng H, Hu B, Guan L, Zhang H, Yu AM, Johnson CH, Gonzalez FJ, Huang M, Bi H. PPARα regulates tumor cell proliferation and senescence via a novel target gene carnitine palmitoyltransferase 1C. Carcinogenesis 2017; 38:474-483. [PMID: 28334197 DOI: 10.1093/carcin/bgx023] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 03/01/2017] [Indexed: 12/16/2022] Open
Abstract
Carnitine palmitoyltransferase 1C (CPT1C), an enzyme located in the outer mitochondria membrane, has a crucial role in fatty acid transport and oxidation. It is also involved in cell proliferation and is a potential driver for cancer cell senescence. However, its upstream regulatory mechanism is unknown. Peroxisome proliferator activated receptor α (PPARα) is a ligand-activated transcription factor that regulates lipid metabolism and tumor progression. The current study aimed to elucidate whether and how PPARα regulates CPT1C and then affects cancer cell proliferation and senescence. Here, for the first time we report that PPARα directly activated CPT1C transcription and CPT1C was a novel target gene of PPARα, as revealed by dual-luciferase reporter and chromatin immunoprecipitation (ChIP) assays. Moreover, regulation of CPT1C by PPARα was p53-independent. We further confirmed that depletion of PPARα resulted in low CPT1C expression and then inhibited proliferation and induced senescence of MDA-MB-231 and PANC-1 tumor cell lines in a CPT1C-dependent manner, while forced PPARα overexpression promoted cell proliferation and reversed cellular senescence. Taken together, these results indicate that CPT1C is a novel PPARα target gene that regulates cancer cell proliferation and senescence. The PPARα-CPT1C axis may be a new target for the intervention of cancer cellular proliferation and senescence.
Collapse
Affiliation(s)
- Yixin Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Yongtao Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Yaoyao Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Hang Zeng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Bingfang Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Lihuan Guan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Huizhen Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06520, USA and
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | - Huichang Bi
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| |
Collapse
|