1
|
Lengerli D, Bakht A, Çalışkan K, Dahlke P, Bal NB, Jordan PM, Çalışkan B, Werz O, Banoglu E. Phenyl-benzyl-ureas with pyridazinone motif: Potent soluble epoxide hydrolase inhibitors with enhanced pharmacokinetics and efficacy in a paclitaxel-induced neuropathic pain model. Eur J Med Chem 2025; 290:117510. [PMID: 40101448 DOI: 10.1016/j.ejmech.2025.117510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025]
Abstract
The severe pain linked to chemotherapy-induced peripheral neuropathy (CIPN) often becomes a critical factor limiting the effective dosage of life-saving chemotherapy treatments. This debilitating side effect not only hampers the effectiveness of cancer therapy but also poses challenges due to the adverse effects of current treatment options for managing CIPN-related pain complications. Soluble epoxide hydrolase (sEH) inhibitors, which elevate endogenous epoxy-fatty acid levels, have been shown to mitigate CIPN-related pain in different rodent models. In our quest to develop potent sEH inhibitors, we developed novel benzyl phenyl urea derivatives incorporating a pyridazinone ring alongside the urea group as a secondary pharmacophore. These compounds demonstrated remarkable potency in inhibiting sEH, with IC50 values ranging from 0.2 to 57 nM. Compound FP9 (IC50 = 0.2 nM), the most potent in this series, exhibited enhanced metabolic stability, translating into significantly improved oral bioavailability (F = 78 %). It consistently relieved pain perception in mice with paclitaxel-induced peripheral neuropathy, achieving a significant and sustained effect compared to gabapentin. In addition, docking studies and molecular dynamics simulations with FP9 provided valuable insights into the binding interactions between the inhibitor and the sEH binding site, offering direction for further optimization of new analogs. These findings align with recent research that highlights the beneficial effects of sEH inhibitors in reducing pain thresholds associated with CIPN.
Collapse
Affiliation(s)
- Deniz Lengerli
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Arooj Bakht
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Kübra Çalışkan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Philipp Dahlke
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Nur Banu Bal
- Department of Pharmacology, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Burcu Çalışkan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Erden Banoglu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey.
| |
Collapse
|
2
|
Alabbasi S, Tacconelli S, de Vries M, Gunnarsson I, Oke V, Kvarnström M, De Michele A, Gregorio PD, Patrignani P, Idborg H, Jakobsson PJ. Janus kinase inhibitors enhance prostanoid biosynthesis in human whole blood in vitro: implications for cardiovascular side effects and prevention strategies. Ann Rheum Dis 2025:S0003-4967(25)00882-9. [PMID: 40328613 DOI: 10.1016/j.ard.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/27/2025] [Accepted: 03/27/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVES Janus kinase inhibitors (JAKis) effectively treat chronic inflammatory diseases but are associated with cardiovascular side effects through unknown mechanisms. This study aimed to investigate the impact of JAKis on prothrombotic thromboxane (TX)A2 production in human whole blood (WB) as a possible mechanism. METHODS We evaluated the effects of 4 JAKis- tofacitinib, baricitinib, filgotinib, and upadacitinib (0.04-20.0 μM)-on TXB2 biosynthesis in clotting WB from healthy subjects, serving as a marker for platelet TXA2 generation. Additionally, we assessed the impact of these JAKis on TXB2 production in WB from healthy subjects, patients with systemic lupus erythematosus (SLE), and treatment-naïve patients with axial spondyloarthritis (axSpA) after 24-hour lipopolysaccharide (LPS) stimulation, as a marker of platelet and leukocyte prostanoid biosynthesis. RESULTS All JAKis increased serum TXB2 production in clotting WB, although not in a concentration-dependent manner. In LPS-stimulated WB, tofacitinib (1 μM) significantly increased TXB2 production in healthy subjects (HSs) (42% ± 33%, n = 17), patients with SLE (57% ± 39%, n = 12), and patients with axSpA (31% ± 23%, n = 15). Baricitinib (1 μM) also increased TXB2 in HSs (30% ± 22%, n = 10). Upadacitinib showed a trend towards increased TXB2 (46% ± 40%, n = 7), while filgotinib did not (21% ± 19%, n = 7). Aspirin (100 μM) almost completely reduced serum TXB2 in the presence of all JAKis. CONCLUSIONS The enhanced biosynthesis of TXA2 in platelets, with a minor contribution from leukocytes, may contribute to the increased cardiovascular risk associated with JAKis. Low-dose aspirin may offer a protective effect, warranting further investigations.
Collapse
Affiliation(s)
- Sabreen Alabbasi
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Stefania Tacconelli
- Systems Pharmacology and Translational Therapeutics Laboratory, Center for Advanced Studies and Technology (CAST), G. d'Annunzio University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, G. d'Annunzio University Medical School, Chieti, Italy
| | - Mirjam de Vries
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Iva Gunnarsson
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| | - Vilija Oke
- Center for Rheumatology (CFR), Academic Specialist Center, Stockholm Region and Division of Rheumatology, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Marika Kvarnström
- Center for Rheumatology (CFR), Academic Specialist Center, Stockholm Region and Division of Rheumatology, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Alessandra De Michele
- Systems Pharmacology and Translational Therapeutics Laboratory, Center for Advanced Studies and Technology (CAST), G. d'Annunzio University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, G. d'Annunzio University Medical School, Chieti, Italy
| | - Patrizia Di Gregorio
- Transfusion Medicine Service of the Azienda Sanitaria Locale (ASL) Lanciano-Vasto-Chieti and G. d'Annunzio University, Chieti, Italy
| | - Paola Patrignani
- Systems Pharmacology and Translational Therapeutics Laboratory, Center for Advanced Studies and Technology (CAST), G. d'Annunzio University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, G. d'Annunzio University Medical School, Chieti, Italy
| | - Helena Idborg
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden.
| | - Per-Johan Jakobsson
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
3
|
Tena-Garitaonaindia M, Rubio JM, Martínez-Plata E, Martínez-Augustin O, Sánchez de Medina F. Pharmacological bases of combining nonsteroidal antiinflammatory drugs and paracetamol. Biomed Pharmacother 2025; 187:118069. [PMID: 40306178 DOI: 10.1016/j.biopha.2025.118069] [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: 02/07/2025] [Revised: 04/13/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
Paracetamol and nonsteroidal antiinflammatory drugs (NSAIDs), particularly ibuprofen, are frequently administered together. A systematic review of clinical studies using combined or alternating regimes of NSAIDs was performed up to May 2023. Clinical evidence (77 studies) confirms that in many cases efficacy is enhanced by paracetamol + NSAID combinations, but quite a few studies show no added benefit. Synergism is more commonly found with combined regimens in analgesia for surgery, and with alternating regimes in antipyresis. In some instances the advantage may be related to the short duration of the effect of paracetamol. Mechanistically, central and peripheral actions associated with inhibition of cyclooxygenase have been documented for both paracetamol and NSAIDs, which are relevant for analgesia, antipyresis and closure of patent ductus arteriosus in neonates. In addition, paracetamol may achieve analgesia via different central pathways independently of cyclooxygenase. Hence, increased analgesia may result from NSAID and paracetamol acting at least partly via different mechanisms, while enhancement of antipyresis probably is explained simply by augmented or more prolongued inhibition of cyclooxygenase. Because of the inconsistencies found in the available evidence, added benefit should not be assumed for paracetamol/NSAID combinations. In addition, combining paracetamol and NSAIDs may lead to increased dosing errors, and may result in increased toxicity as a result of enhanced cyclooxygenase interference, a possibility that has barely been scrutinized. We conclude that combining paracetamol and NSAIDs may be justified in analgesia, but further studies are warranted to establish when and how an enhanced effect is achieved with this strategy.
Collapse
Affiliation(s)
- Mireia Tena-Garitaonaindia
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - José Manuel Rubio
- Department of Pharmacology, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Fermín Sánchez de Medina
- Department of Pharmacology, School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADA, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
4
|
Zhang Y, Yang L, Gan Y, Zhao C, Zhou C, Chen J, Yin Y, Xia S, Yang H, Bao X, Zhang M, Xu Y, Li J. Benzydamine attenuates microglia-mediated neuroinflammation and ischemic brain injury by targeting cathepsin s. Int Immunopharmacol 2025; 146:113824. [PMID: 39700961 DOI: 10.1016/j.intimp.2024.113824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Microglia, the primary immune cells of the central nervous system, play a crucial role in the neuroinflammatory processes following ischemic stroke. Targeting neuroinflammation is a promising strategy to enhance the outcomes of ischemic stroke. Benzydamine (BA), a well-known non-steroidal anti-inflammatory drug, has demonstrated potential in inhibiting pro-inflammatory cytokines across various disease models. However, the potential role of BA in microglial activation and post-stroke neuroinflammation remains unclear. Our study reveals that BA effectively suppresses the lipopolysaccharide (LPS)-stimulated pro-inflammatory responses of primary microglia, with high-dose BA (10 μM) suppressing LPS-induced inflammatory markers by up to 59.1 % in the mRNA levels of IL-1β. Furthermore, BA mitigated ischemic brain injury in experimental stroke mice. BA treatment also significantly attenuated neuroinflammatory responses and attenuates ischemic brain injury in experimental stroke mice. Further investigation revealed that BA reduces the release of the LPS-stimulated pro-inflammatory factors and activation of primary microglia by directly binding to and inhibiting the activity of cathepsin S (CTSS). In conclusion, our study identifies BA as a promising CTSS inhibitor with potential to suppress neuroinflammation following ischemic stroke. Our findings provide a theoretical basis for developing new neuroprotective strategies.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Lixuan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yonghui Gan
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Chenchen Zhao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Chao Zhou
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Jian Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yanping Yin
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Disease, Nanjing University, Nanjing 210008, China
| | - Haiyan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Disease, Nanjing University, Nanjing 210008, China
| | - Meijuan Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Disease, Nanjing University, Nanjing 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China; Nanjing Neurology Clinical Medical Center, Nanjing, 210008, China.
| | - Jingwei Li
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| |
Collapse
|
5
|
Patrignani P, De Michele A, Tacconelli S. New antiplatelet approach: inhibiting Pim kinase to reduce constitutive surface expression of thromboxane A 2 receptor. J Thromb Haemost 2025; 23:43-46. [PMID: 39798970 DOI: 10.1016/j.jtha.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 01/15/2025]
Affiliation(s)
- 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 Medical School, 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 Medical School, Chieti, Italy
| | - 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 Medical School, Chieti, Italy
| |
Collapse
|
6
|
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
|
7
|
Poblete J, Fernández-Martínez J, Aranda M, Quispe-Fuentes I. Green Recovery and Identification of Antioxidant and Enzyme Inhibitor Molecules from Pisco Grape Pomace by Targeted Effects Analysis Using Thin-Layer Chromatography, Bioassay, and Mass Spectrometry. Antioxidants (Basel) 2024; 13:1418. [PMID: 39594559 PMCID: PMC11591367 DOI: 10.3390/antiox13111418] [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: 10/23/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
The search and identification of inhibitory molecules from novel natural sources, such as pisco grape pomace extract obtained by green techniques, may help to develop agents with therapeutic potential that are beneficial to health with fewer adverse effects than drugs. Many drugs act as enzyme inhibitors, decreasing their activity and thus correcting a metabolic imbalance. This study aims to identify bioactive molecules with antioxidant and inhibitory activity over acetylcholinesterase and cyclooxygenase enzymes present in pisco grape pomace green extracts. Bioactive molecules were detected and identified applying directed effect analysis on planar chromatography coupled to mass spectrometry. For the first time, the presence of antioxidant molecules (quercetin-3-O-glucuronide, quercetin-3-O-glucoside, and gallic acid) and inhibitors of acetylcholinesterase (kaempferol-3-O-glucoside) and cyclooxygenase (gallic acid) enzymes are reported in pisco grape pomace. According to the results, grape pomace could be an alternative to develop novel functional foods and nutraceuticals that provide health benefits and, at the same time, generate a circular economy in the industry.
Collapse
Affiliation(s)
- Jacqueline Poblete
- Food Engineering Department, Universidad de La Serena, Av. Raúl Bitrán 1305, La Serena 1700000, Chile;
| | | | - Mario Aranda
- Department of Chemistry and Pharmacy, Pontificia Universidad Católica de Chile, Santiago 7810000, Chile
| | - Issis Quispe-Fuentes
- Food Engineering Department, Universidad de La Serena, Av. Raúl Bitrán 1305, La Serena 1700000, Chile;
| |
Collapse
|
8
|
Contursi A, Tacconelli S, Di Berardino S, De Michele A, Patrignani P. Platelets and extracellular vesicles in disease promotion via cellular cross-talk and eicosanoid biosynthesis. Prostaglandins Other Lipid Mediat 2024; 173:106848. [PMID: 38723943 DOI: 10.1016/j.prostaglandins.2024.106848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/12/2024] [Accepted: 05/03/2024] [Indexed: 06/17/2024]
Abstract
New insights have been gained on the role of platelets beyond thrombosis. Platelets can accumulate in damaged and inflamed tissues, acting as a sentinel to detect and repair tissue damage. However, by releasing several soluble factors, including thromboxane A2 (TXA2) and 12-hydroxyeicosatetraenoic acid, and extracellular vesicles (EVs), platelets can activate vascular cells, stromal, such as fibroblasts, immune cells, and cancer cells, leading to atherosclerosis, vascular restenosis, tissue fibrosis, and tumor metastasis. Platelet-derived extracellular vesicles (PEVs) are released when platelets are activated and can transfer their cargo to other cell types, thus contributing to the development of diseases. Inhibitors of the internalization of PEVs can potentially represent novel therapeutic tools. Both platelets and PEVs contain a significant number of different types of molecules, and their omics assessment and integration with clinical data using computational approaches have the potential to detect early disease development and monitor drug treatments.
Collapse
Affiliation(s)
- Annalisa Contursi
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University Medical School, Chieti, Italy
| | - Stefania Tacconelli
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy
| | - Sara Di Berardino
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University Medical School, Chieti, Italy
| | - Alessandra De Michele
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University Medical School, Chieti, Italy
| | - Paola Patrignani
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University Medical School, Chieti, Italy.
| |
Collapse
|
9
|
Vidal M, Roldán T. [Penetration capacity and local anti-inflammatory effect of a methyl salicylate gel with turpentine essence, camphor, and menthol for the symptomatic local treatment of muscular and joint pains]. FARMACEUTICOS COMUNITARIOS 2024; 16:5-11. [PMID: 39188782 PMCID: PMC11346264 DOI: 10.33620/fc.2173-9218.(2024).15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/27/2024] [Indexed: 08/28/2024]
Abstract
Introduction Oral non-steroidal anti-inflammatory drugs (NSAIDs) are effective in managing musculoskeletal pain, but often are associated with side effects. Therefore, topical anti-inflammatory drugs are considered a suitable alternative for certain patients. Objective To determine the skin penetration capacity and anti-inflammatory efficacy of a methyl salicylate gel (MS) with turpentine essence, camphor and menthol (GelSMETAM). Methods Human skin explants were used (three biological replicates with 3 technical replicates each were performed). To assess penetration capacity, methyl salicylate (MS) levels were quantified at different intervals using liquid chromatography with tandem mass spectrometry (LC-MS/MS). For the anti-inflammatory effect, the gene expression of major pro-inflammatory factors was quantified 30 minutes, 8 hours, and 12 hours after an impact; comparing GelSMETAM application with untreated controls. Results The results showed that approximately 0.8% of the total applied methyl salicylate penetrated the skin with a time-dependent concentration increase. The anti-inflammatory effect demonstrated a significant reduction in the gene expression of TNFα, IL1α, IL6, IL8, and NFKβ at 30 minutes (between -39% and -53.6% compared to control), which was maintained at 8 hours except for TNFα (between -59% and -92.6%) and persisted even at 12 hours (-69.4%) for IL1α. Conclusion The results support the hypothesis that GelSMETAM provides an effective and prolonged anti-inflammatory response, positioning it as a suitable option for the management of musculoskeletal pain.
Collapse
Affiliation(s)
| | - Taïs Roldán
- Responsable técnico y directora de asuntos regulatorios, médicos y de seguridad de Reckitt Benckiser Healthcare, EspañaEspaña
| |
Collapse
|
10
|
Nicolau ST, Tres DP, Ayala TS, Menolli RA. Nonsteroidal Anti-Inflammatory Drugs and Experimental Chagas Disease: An Unsolved Question. Parasite Immunol 2024; 46:e13057. [PMID: 39008292 DOI: 10.1111/pim.13057] [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: 12/08/2023] [Revised: 06/13/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
Chagas disease is a parasitic disease caused by the protozoan Trypanosoma cruzi with an acute, detectable blood parasites phase and a chronic phase, in which the parasitemia is not observable, but cardiac and gastrointestinal consequences are possible. Mice are the principal host used in experimental Chagas disease but reproduce the human infection depending on the animal and parasite strain, besides dose and route of administration. Lipidic mediators are tremendously involved in the pathogenesis of T. cruzi infection, meaning the prostaglandins and thromboxane, which participate in the immunosuppression characteristic of the acute phase. Thus, the eicosanoids inhibition caused by the nonsteroidal anti-inflammatory drugs (NSAIDs) alters the dynamic of the disease in the experimental models, both in vitro and in vivo, which can explain the participation of the different mediators in infection. However, marked differences are founded in the various NSAIDs existing because of the varied routes blocked by the drugs. So, knowing the results in the experimental models of Chagas disease with or without the NSAIDs helps comprehend the pathogenesis of this infection, which still needs a better understanding.
Collapse
Affiliation(s)
- Scheila Thaís Nicolau
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, Brazil
| | - Daniela Patrícia Tres
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, Brazil
| | - Thaís Soprani Ayala
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, Brazil
| | - Rafael Andrade Menolli
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, Brazil
| |
Collapse
|
11
|
Haque MF, El-Nashar HAS, Akbor MS, Alfaifi M, Bappi MH, Chowdhury AK, Hossain MK, El-Shazly M, Albayouk T, Saleh N, Islam MT. Anti-inflammatory activity of d-pinitol possibly through inhibiting COX-2 enzyme: in vivo and in silico studies. Front Chem 2024; 12:1366844. [PMID: 38690012 PMCID: PMC11058972 DOI: 10.3389/fchem.2024.1366844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/21/2024] [Indexed: 05/02/2024] Open
Abstract
Introduction: D-pinitol, a naturally occurring inositol, has diverse biological activities like antioxidant, antimicrobial and anticancer activities. This study aimed to evaluate anti-inflammatory effect of d-pinitol in a chick model. Additionally, in silico studies were performed to evaluate the molecular interactions with cyclooxygenase-2 (COX-2). Methods: The tested groups received d-pinitol (12.5, 25, and 50 mg/kg) and the standard drugs celecoxib and ketoprofen (42 mg/kg) via oral gavage prior to formalin injection. Then, the number of licks was counted for the first 10 min, and the paw edema diameter was measured at 60, 90, and 120 min. Results and Discussion: The d-pinitol groups significantly (p < 0.05) reduced the number of paw licks and paw edema diameters, compared to negative control. When d-pinitol was combined with celecoxib, it reduced inflammatory parameters more effectively than the individual groups. The in silico study showed a promising binding capacity of d-pinitol with COX-2. Taken together, d-pinitol exerted anti-inflammatory effects in a dose-dependent manner, possibly through COX-2 interaction pathway.
Collapse
Affiliation(s)
- Mst. Farjanamul Haque
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Heba A. S. El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Md. Showkoth Akbor
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Mohammed Alfaifi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mehedi Hasan Bappi
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | | | - Muhammad Kamal Hossain
- School of Pharmacy, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Pharmacy, University of Science and Technology Chittagiong, Chittagong, Bangladesh
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Tala Albayouk
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Na’il Saleh
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, Bangladesh
- BioLuster Research Center, Dhaka, Bangladesh
| |
Collapse
|
12
|
Osman EO, Khalil NA, Magdy A, El-Dash Y. Pyridazine and pyridazinone derivatives: Synthesis and in vitro investigation of their anti-inflammatory potential in LPS-induced RAW264.7 macrophages. Drug Dev Res 2024; 85:e22173. [PMID: 38515272 DOI: 10.1002/ddr.22173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 12/18/2023] [Accepted: 03/04/2024] [Indexed: 03/23/2024]
Abstract
New pyridazine and pyridazinone derivatives 3a-g, 4a-f, 6a, and 6b were designed and synthesized. Cell viability of all compounds was established based on the viability of lipopolysaccharide-induced RAW264.7 macrophage cells determined via the MTT assay. In vitro inhibition assays on human COX-1 and COX-2 enzymes were conducted to probe the newly synthesized compounds' anti-inflammatory activity. The half maximal inhibitory concentration values for the most active compounds, 3d, 3e, and 4e towards COX-2 were 0.425, 0.519, and 0.356 µM, respectively, in comparison with celecoxib. The newly synthesized compounds' ability to inhibit the production of certain proinflammatory cytokines, such as inducible nitric oxide synthase, tumor necrosis factor-α, interleukin-6, and prostaglandin-E2, was also estimated in lipopolysaccharide-induced macrophages (RAW264.7 cells). Compounds 3d and 3e were identified as the most potent cytokine production inhibitors. The results of molecular modeling studies suggested that these compounds were characterized by a reasonable binding affinity toward the active site of COX-2, when compared to a reference ligand. These results might be taken into consideration in further investigations into new anti-inflammatory agents.
Collapse
Affiliation(s)
- Eman O Osman
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nadia A Khalil
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Alaa Magdy
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Yara El-Dash
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
13
|
Ebmeyer J, Najjar A, Lange D, Boettcher M, Voß S, Brandmair K, Meinhardt J, Kuehnl J, Hewitt NJ, Krueger CT, Schepky A. Next generation risk assessment: an ab initio case study to assess the systemic safety of the cosmetic ingredient, benzyl salicylate, after dermal exposure. Front Pharmacol 2024; 15:1345992. [PMID: 38515841 PMCID: PMC10955127 DOI: 10.3389/fphar.2024.1345992] [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: 11/28/2023] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
We performed an ab initio next-generation risk assessment (NGRA) for a fragrance ingredient, benzyl salicylate (BSal), to demonstrate how cosmetic ingredients can be evaluated for systemic toxicity endpoints based on non-animal approaches. New approach methodologies (NAMs) used to predict the internal exposure included skin absorption assays, hepatocyte metabolism, and physiologically based pharmacokinetic (PBPK) modeling, and potential toxicodynamic effects were assessed using pharmacology profiling, ToxProfiler cell stress assay, transcriptomics in HepG2 and MCF-7 cells, ReproTracker developmental and reproductive toxicology (DART) assays, and cytotoxicity assays in human kidney cells. The outcome of the NGRA was compared to that of the traditional risk assessment approach based on animal data. The identification of the toxicologically critical entity was a critical step that directed the workflow and the selection of chemicals for PBPK modeling and testing in bioassays. The traditional risk assessment and NGRA identified salicylic acid (SA) as the "toxdriver." A deterministic PBPK model for a single-day application of 1.54 g face cream containing 0.5% BSal estimated the Cmax for BSal (1 nM) to be much lower than that of its major in vitro metabolite, SA (93.2 nM). Therefore, SA was tested using toxicodynamics bioassays. The lowest points of departure (PoDs) were obtained from the toxicogenomics assays. The interpretation of these results by two companies and methods were similar (SA only results in significant gene deregulation in HepG2 cells), but PoD differed (213 μM and 10.6 µM). A probabilistic PBPK model for repeated applications of the face cream estimated the highest Cmax of SA to be 630 nM. The resulting margins of internal exposure (MoIE) using the PoDs were 338 and 16, which were more conservative than those derived from external exposure and in vivo PoDs (margin of safety values were 9,705). In conclusion, both traditional and ab initio NGRA approaches concluded that the daily application of BSal in a cosmetic leave-on face cream at 0.5% is safe for humans. The processing and interpretation of toxicogenomics data can lead to different PoDs, which can subsequently affect the calculation of the MoIE. This case study supports the use of NAMs in a tiered NGRA ab initio approach.
Collapse
|
14
|
Patrignani P, Tacconelli S, Contursi A, Piazuelo E, Bruno A, Nobili S, Mazzei M, Milillo C, Hofling U, Hijos-Mallada G, Sostres C, Lanas A. Optimizing aspirin dose for colorectal cancer patients through deep phenotyping using novel biomarkers of drug action. Front Pharmacol 2024; 15:1362217. [PMID: 38495101 PMCID: PMC10941341 DOI: 10.3389/fphar.2024.1362217] [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: 12/27/2023] [Accepted: 02/15/2024] [Indexed: 03/19/2024] Open
Abstract
Background: Low-dose aspirin's mechanism of action for preventing colorectal cancer (CRC) is still debated, and the optimal dose remains uncertain. We aimed to optimize the aspirin dose for cancer prevention in CRC patients through deep phenotyping using innovative biomarkers for aspirin's action. Methods: We conducted a Phase II, open-label clinical trial in 34 CRC patients of both sexes randomized to receive enteric-coated aspirin 100 mg/d, 100 mg/BID, or 300 mg/d for 3 ± 1 weeks. Biomarkers were evaluated in blood, urine, and colorectal biopsies at baseline and after dosing with aspirin. Novel biomarkers of aspirin action were assessed in platelets and colorectal tissues using liquid chromatography-mass spectrometry to quantify the extent of cyclooxygenase (COX)-1 and COX-2 acetylation at Serine 529 and Serine 516, respectively. Results: All aspirin doses caused comparable % acetylation of platelet COX-1 at Serine 529 associated with similar profound inhibition of platelet-dependent thromboxane (TX)A2 generation ex vivo (serum TXB2) and in vivo (urinary TXM). TXB2 was significantly reduced in CRC tissue by aspirin 300 mg/d and 100 mg/BID, associated with comparable % acetylation of COX-1. Differently, 100 mg/day showed a lower % acetylation of COX-1 in CRC tissue and no significant reduction of TXB2. Prostaglandin (PG)E2 biosynthesis in colorectal tumors and in vivo (urinary PGEM) remained unaffected by any dose of aspirin associated with the variable and low extent of COX-2 acetylation at Serine 516 in tumor tissue. Increased expression of tumor-promoting genes like VIM (vimentin) and TWIST1 (Twist Family BHLH Transcription Factor 1) vs. baseline was detected with 100 mg/d of aspirin but not with the other two higher doses. Conclusion: In CRC patients, aspirin 300 mg/d or 100 mg/BID had comparable antiplatelet effects to aspirin 100 mg/d, indicating similar inhibition of the platelet's contribution to cancer. However, aspirin 300 mg/d and 100 mg/BID can have additional anticancer effects by inhibiting cancerous tissue's TXA2 biosynthesis associated with a restraining impact on tumor-promoting gene expression. EUDRACT number: 2018-002101-65. Clinical Trial Registration: ClinicalTrials.gov, identifier NCT03957902.
Collapse
Affiliation(s)
- Paola Patrignani
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Stefania Tacconelli
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Annalisa Contursi
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Elena Piazuelo
- Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Annalisa Bruno
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Stefania Nobili
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Matteo Mazzei
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Cristina Milillo
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Psychological Sciences, Health, and Territory, “G. d’Annunzio” University, Chieti, Italy
| | - Ulrika Hofling
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Gonzalo Hijos-Mallada
- University Hospital LB, Aragon Health Research Institute (IISAragon), CIBERehd, University of Zaragoza, Zaragoza, Spain
| | - Carlos Sostres
- University Hospital LB, Aragon Health Research Institute (IISAragon), CIBERehd, University of Zaragoza, Zaragoza, Spain
| | - Angel Lanas
- University Hospital LB, Aragon Health Research Institute (IISAragon), CIBERehd, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
15
|
Vashistha VK, Kumar T, Yadav S, Das DK. Enantioselective separation and determination of ibuprofen: Stereoselective pharmacokinetics, pharmacodynamics and analytical methods. Chirality 2024; 36:e23647. [PMID: 38356207 DOI: 10.1002/chir.23647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/13/2024] [Accepted: 01/14/2024] [Indexed: 02/16/2024]
Abstract
Ibuprofen (IBP), the 29th most prescribed drug in the United States in 2019, is a widely used nonsteroidal anti-inflammatory drug (NSAID) comprising two enantiomers, (R)-IBP and (S)-IBP, collectively known as (RS)-IBP. This critical review examines analytical techniques for the enantioselective separation and determination of IBP enantiomers, crucial for pharmaceutical and clinical applications. The review focuses on state-of-the-art methods, including chromatographic techniques including high-performance liquid chromatography, gas chromatography, liquid chromatography-tandem mass spectrometry, and some other techniques. This review addresses pharmacokinetics, pharmacology, and side effects of each enantiomer, ensuring safe drug usage. By consolidating diverse analytical methods and their applicability in different matrices, this review serves as a valuable resource for researchers, analysts, and practitioners in pharmaceutical analysis, pharmacology, and clinical studies.
Collapse
Affiliation(s)
- Vinod Kumar Vashistha
- Department of Chemistry, GLA University, Mathura, India
- Department of Chemistry, University of Lucknow, Lucknow, India
| | - Tarun Kumar
- Department of Applied Sciences, MIET Kumaon Haldwani Nainital, Haldwani, India
| | - Suman Yadav
- Department of Chemistry, Swami Shraddhanand College, University of Delhi, Delhi, India
| | | |
Collapse
|
16
|
Patrignani P, Schebb NH, Werz O, Steinhilber D. Editorial: Lipidomics of oxylipins in biological systems. Front Pharmacol 2023; 14:1342596. [PMID: 38125891 PMCID: PMC10731249 DOI: 10.3389/fphar.2023.1342596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Affiliation(s)
| | | | - Oliver Werz
- Oliver Werz, Friedrich Schiller University Jena, Jena, Germany
| | | |
Collapse
|
17
|
da Silva PR, Apolinário NDM, da Silva SÂS, Araruna MEC, Costa TB, e Silva YMSDM, da Silva TG, de Moura RO, dos Santos VL. Anti-Inflammatory Activity of N'-(3-(1H-indol-3-yl)benzylidene)-2-cyanoacetohydrazide Derivative via sGC-NO/Cytokine Pathway. Pharmaceuticals (Basel) 2023; 16:1415. [PMID: 37895886 PMCID: PMC10610422 DOI: 10.3390/ph16101415] [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: 08/17/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
The N-acylhydrazone function has been reported as a pharmacophore group of molecules with diverse pharmacological activities, including anti-inflammatory effects. Therefore, this study was designed to evaluate the anti-inflammatory potential of the compound N'-(3-(1H-indol-3-yl)benzylidene)-2-cyanoacetohydrazide (JR19) in vivo. The study started with the carrageenan-induced peritonitis model, followed by an investigation of leukocyte migration using the subcutaneous air pouch test and an assessment of the antinociceptive profile using formalin-induced pain. A preliminary molecular docking study focusing on the crystallographic structures of NFκB, iNOS, and sGC was performed to determine the likely mechanism of action. The computational study revealed satisfactory interaction energies with the selected targets, and the same peritonitis model was used to validate the involvement of the nitric oxide pathway and cytokine expression in the peritoneal exudate of mice pretreated with L-NAME or methylene blue. In the peritonitis assay, JR19 (10 and 20 mg/kg) reduced leukocyte migration by 59% and 52%, respectively, compared to the vehicle group, with the 10 mg/kg dose used in subsequent assays. In the subcutaneous air pouch assay, the reduction in cell migration was 66%, and the response to intraplantar formalin was reduced by 39%, particularly during the inflammatory phase, suggesting that the compound lacks central analgesic activity. In addition, a reversal of the anti-inflammatory effect was observed in mice pretreated with L-NAME or methylene blue, indicating the involvement of iNOS and sGC in the anti-inflammatory response of JR19. The compound effectively and significantly decreased the levels of IL-6, TNF-α, IL-17, and IFN-γ, and this effect was reversed in animals pretreated with L-NAME, supporting a NO-dependent anti-inflammatory effect. In contrast, pretreatment with methylene blue only reversed the reduction in TNF-α levels. Therefore, these results demonstrate the pharmacological potential of the novel N-acylhydrazone derivative, which acts through the nitric oxide pathway and cytokine signaling, making it a strong candidate as an anti-inflammatory and immunomodulatory agent.
Collapse
Affiliation(s)
- Pablo Rayff da Silva
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
- Laboratório de Desenvolvimento e Síntese de Fármacos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Nadjaele de Melo Apolinário
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Simone Ângela Soares da Silva
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Maria Elaine Cristina Araruna
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Thássia Borges Costa
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Yvnni M. S. de Medeiros e Silva
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Desenvolvimento e Síntese de Fármacos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Teresinha Gonçalves da Silva
- Departamento de Antibióticos, Centro de Biociências, Universidade Federal de Pernambuco, Recife 50740-520, PE, Brazil;
| | - Ricardo Olímpio de Moura
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Desenvolvimento e Síntese de Fármacos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Vanda Lucia dos Santos
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| |
Collapse
|
18
|
Vijay K, Ambedkar R, Sowmya PRR, Ramaiah S, Ranga Rao A, Gundamaraju R, Hanumanthappa M, Malarvili MB, Manikam R, Lakshminarayana R. Prevention of aspirin-mediated secondary toxicity by combined treatment of carotenoids in macrophages. 3 Biotech 2023; 13:223. [PMID: 37292139 PMCID: PMC10244315 DOI: 10.1007/s13205-023-03632-w] [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: 08/24/2022] [Accepted: 04/26/2023] [Indexed: 06/10/2023] Open
Abstract
Upon understanding the boosting role of carotenoids on the endogenous anti-inflammatory system, it is vital to explore their role in reducing the use of high doses of non-steroidal anti-inflammatory drug (NSAIDs), and their mediated secondary toxicity during the treatment of chronic diseases. The current study investigates the carotenoids potential on inhibition of secondary complications induced by NSAIDs, aspirin (ASA) against lipopolysaccharide (LPS) stimulated inflammation. Initially, this study evaluated a minimal cytotoxic dose of ASA and carotenoids (β-carotene, BC/lutein, LUT/astaxanthin, AST/fucoxanthin FUCO) in Raw 264.7, U937, and peripheral blood mononuclear cells (PBMCs). In all three cells, carotenoids + ASA treatment reduced the LDH release, NO, and PGE2 efficiently than an equivalent dose of carotenoid or ASA treated alone. Based on cytotoxicity and sensitivity results, RAW 264.7 cells were selected for further cell-based assay. Among carotenoids, FUCO + ASA exhibited an efficient reduction of LDH release, NO, and PGE2 than the other carotenoids (BC + ASA, LUT + ASA, and AST + ASA) treatment. FUCO + ASA combination decreased LPS/ASA induced oxidative stress, pro-inflammatory mediators (iNOS, COX-2, and NF-κB), and cytokines (IL-6, TNF-α, and IL-1β) efficiently. Further, apoptosis was inhibited by 69.2% in FUCO + ASA, and 46.7% in ASA than LPS treated cells. A drastic decrease in intracellular ROS generation with the increase in GSH was observed in FUCO + ASA compared to LPS/ASA groups. The results documented on the low dose of ASA with a relative physiological concentration of FUCO suggested greater importance for alleviating secondary complications and optimize prolonged chronic disease treatments with NSAID's associated side effects. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03632-w.
Collapse
Affiliation(s)
- Kariyappa Vijay
- Department of Microbiology and Biotechnology, Jnana Bharathi Campus, Bangalore University, Bangalore, 560 056 India
| | - Rudrappa Ambedkar
- Department of Microbiology and Biotechnology, Jnana Bharathi Campus, Bangalore University, Bangalore, 560 056 India
| | | | - Suresh Ramaiah
- Department of Statistics, Jnana Bharathi Campus, Bangalore University, Bangalore, 560 056 India
| | - Ambati Ranga Rao
- Department of Biotechnology, Vignan’s Foundation of Science, Technology and Research (Deemed to be University), Vadlamudi, Guntur, Andhra Pradesh 522213 India
| | - Rohit Gundamaraju
- ER Stress and Mucosal Immunology Lab, School of Health Sciences, University of Tasmania, Launceston, TAS 7248 Australia
| | - Manjunatha Hanumanthappa
- Department of Biochemistry, Jnana Bharathi Campus, Bangalore University, Bangalore, 560 056 India
| | - M. B. Malarvili
- Trauma and Emergency, University of Malaya, Kuala Lumpur, Malaysia
| | - Rishya Manikam
- School of Biomedical and Health Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor Bahru Malaysia
| | - Rangaswamy Lakshminarayana
- Department of Microbiology and Biotechnology, Jnana Bharathi Campus, Bangalore University, Bangalore, 560 056 India
| |
Collapse
|
19
|
Bruno A, Tacconelli S, Contursi A, Ballerini P, Patrignani P. Cyclooxygenases and platelet functions. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 97:133-165. [PMID: 37236757 DOI: 10.1016/bs.apha.2022.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Cyclooxygenase (COX) isozymes, i.e., COX-1 and COX-2, are encoded by separate genes and are involved in the generation of the same products, prostaglandin (PG)G2 and PGH2 from arachidonic acid (AA) by the COX and peroxidase activities of the enzymes, respectively. PGH2 is then transformed into prostanoids in a tissue-dependent fashion due to the different expression of downstream synthases. Platelets present almost exclusively COX-1, which generates large amounts of thromboxane (TX)A2, a proaggregatory and vasoconstrictor mediator. This prostanoid plays a central role in atherothrombosis, as shown by the benefit of the antiplatelet agent low-dose aspirin, a preferential inhibitor of platelet COX-1. Recent findings have shown the relevant role played by platelets and TXA2 in developing chronic inflammation associated with several diseases, including tissue fibrosis and cancer. COX-2 is induced in response to inflammatory and mitogenic stimuli to generate PGE2 and PGI2 (prostacyclin), in inflammatory cells. However, PGI2 is constitutively expressed in vascular cells in vivo and plays a crucial role in protecting the cardiovascular systems due to its antiplatelet and vasodilator effects. Here, platelets' role in regulating COX-2 expression in cells of the inflammatory microenvironment is described. Thus, the selective inhibition of platelet COX-1-dependent TXA2 by low-dose aspirin prevents COX-2 induction in stromal cells leading to antifibrotic and antitumor effects. The biosynthesis and functions of other prostanoids, such as PGD2, and isoprostanes, are reported. In addition to aspirin, which inhibits platelet COX-1 activity, possible strategies to affect platelet functions by influencing platelet prostanoid receptors or synthases are discussed.
Collapse
Affiliation(s)
- Annalisa Bruno
- Center for Advanced Studies and Technology (CAST), Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy
| | - Stefania Tacconelli
- Center for Advanced Studies and Technology (CAST), Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy
| | - Annalisa Contursi
- Center for Advanced Studies and Technology (CAST), Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy
| | - Patrizia Ballerini
- Center for Advanced Studies and Technology (CAST), Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, "G.d'Annunzio" University, Chieti, Italy
| | - Paola Patrignani
- Center for Advanced Studies and Technology (CAST), Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G. d'Annunzio" University, Chieti, Italy.
| |
Collapse
|
20
|
Kuczyńska J, Nieradko-Iwanicka B. Comparison of the effects of ketoprofen and ketoprofen lysine salt on the Wistar rats' nervous system, kidneys and liver after ethyl alcohol intoxication. Biomed Pharmacother 2023; 161:114456. [PMID: 36870283 DOI: 10.1016/j.biopha.2023.114456] [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: 01/05/2023] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Side effects of Ketoprofen and ketoprofen lysine salt (KLS) may be inter alia from the central nervous system, kidneys and liver. After binge drinking people often use ketoprofen, which increases the risk for the occurrence of side effects. The aim of the study was to compere effects of ketoprofen and KLS on the nervous system, kidneys and liver after ethyl alcohol intoxication. There were 6 groups of 6 male rats which received: ethanol; 0.9%NaCl; 0.9%NaCl and ketoprofen; ethanol and ketoprofen; 0.9%NaCl and KLS; ethanol and KLS. On day 2, the motor coordination test on a rotary rod and memory and motor activity test in the Y-maze were performed. Hot plate test was performed on day 6. After euthanasia brains, livers and kidneys were taken to histopathological tests. Motor coordination was significant worse in group 5 vs 1,3, p 0.05. Spontaneous motor activity of group 6 was significant better than that of groups 1,5. Pain tolerance of group 6 was significant worse than that of groups 1,4,5. Liver and kidney mass were significantly lower in group 6 vs group 3,5 and vs group 1,3, respectively. The histopathologic examination of the brains and kidneys revealed normal picture in all groups, without signs of inflammation. In the histopathologic examination of the livers in one animal in group 3 some of the specimens showed perivascular inflammation. After alcohol ketoprofen is a better painkiller than KLS. Spontaneous motor activity is better after KLS after alcohol. Both drugs have a similar effect on the kidneys and liver.
Collapse
Affiliation(s)
- Joanna Kuczyńska
- Department of Hygiene and Epidemiology, Medical University of Lublin, Chodźki 7 Street, 20-093 Lublin, Poland; Doctoral School, Medical University of Lublin, Poland.
| | - Barbara Nieradko-Iwanicka
- Department of Hygiene and Epidemiology, Medical University of Lublin, Chodźki 7 Street, 20-093 Lublin, Poland.
| |
Collapse
|
21
|
Yang Y, Geng Y, Cheng X, Gao J, Shi Z, Zhao M. Cyclooxygenase‑2 contributes to the hypoxia‑induced aggravation of the neuroinflammation response stimulated by lipopolysaccharide in microglia. Exp Ther Med 2023; 25:123. [PMID: 36845947 PMCID: PMC9947573 DOI: 10.3892/etm.2023.11822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
Hypoxia and neuroinflammation are key risk factors involved in various pathophysiological neural disorders. Hypoxia can aggravate neuroinflammation in vitro and in vivo; however, the underlying mechanisms remain unknown. In the present study, hypoxia [either 3 or 1% oxygen (O2)] increased lipopolysaccharide (LPS)-induced expression of the IL-6, IL-1β and TNF-α proinflammatory cytokines in BV2 cells. At the molecular level, both hypoxia and FG-4592, an hypoxia inducible factor 1 pathway activator, effectively induced cyclooxygenase-2 (COX-2) expression. The COX-2 inhibitor celecoxib significantly reduced the expression of cytokines induced by LPS under hypoxic conditions. Additionally, the administration of celecoxib inhibited the activation of microglia as well as cytokine expression in mice administered with hypoxia exposure and LPS injection. The present data demonstrated that COX-2 is involved in the hypoxia-induced aggravation of neuroinflammation stimulated by LPS.
Collapse
Affiliation(s)
- Yifan Yang
- Department of Pain Medicine, Peking University People's Hospital, Beijing 100044, P.R. China,Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Yanan Geng
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Xiang Cheng
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Jiayue Gao
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Zibi Shi
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Ming Zhao
- Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China,Correspondence to: Dr Ming Zhao, Department of Brain Plasticity, Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian, Beijing 100850, P.R. China
| |
Collapse
|
22
|
Gur Maz T, Koc B, Jordan PM, İbiş K, Çalışkan B, Werz O, Banoglu E. Benzoxazolone-5-Urea Derivatives as Human Soluble Epoxide Hydrolase (sEH) Inhibitors. ACS OMEGA 2023; 8:2445-2454. [PMID: 36687110 PMCID: PMC9850727 DOI: 10.1021/acsomega.2c06936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/23/2022] [Indexed: 06/17/2023]
Abstract
Inhibition of soluble epoxide hydrolase (sEH) is indicated as a new therapeutic modality against a variety of inflammatory diseases, including metabolic, renal, and cardiovascular disorders. In our ongoing research on sEH inhibitors, we synthesized novel benzoxazolone-5-urea analogues with highly potent sEH inhibitory properties inspired by the crystallographic fragment scaffolds incorporating a single H-bond donor/acceptor pair. The tractable SAR results indicated that the aryl or benzyl fragments flanking the benzoxazolone-urea scaffold conferred potent sEH inhibition, and compounds 31-39 inhibited the sEH activity with IC50 values in the range of 0.39-570 nM. Docking studies and molecular dynamics simulations with the most potent analogue 33 provided valuable insights into potential binding interactions of the inhibitor in the sEH binding region. In conclusion, benzoxazolone-5-ureas furnished with benzyl groups on the urea function can be regarded as novel lead structures, which allow the development of advanced analogues with enhanced properties against sEH.
Collapse
Affiliation(s)
- Tugce Gur Maz
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No:3 Yenimahalle, 06560 Ankara, Turkey
| | - Beyzanur Koc
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No:3 Yenimahalle, 06560 Ankara, Turkey
| | - Paul M. Jordan
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-7743 Jena, Germany
| | - Kübra İbiş
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No:3 Yenimahalle, 06560 Ankara, Turkey
| | - Burcu Çalışkan
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No:3 Yenimahalle, 06560 Ankara, Turkey
| | - Oliver Werz
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-7743 Jena, Germany
| | - Erden Banoglu
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No:3 Yenimahalle, 06560 Ankara, Turkey
| |
Collapse
|
23
|
Sharif N. Neuroaxonal and cellular damage/protection by prostanoid receptor ligands, fatty acid derivatives and associated enzyme inhibitors. Neural Regen Res 2023; 18:5-17. [PMID: 35799502 PMCID: PMC9241399 DOI: 10.4103/1673-5374.343887] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Cellular and mitochondrial membrane phospholipids provide the substrate for synthesis and release of prostaglandins in response to certain chemical, mechanical, noxious and other stimuli. Prostaglandin D2, prostaglandin E2, prostaglandin F2α, prostaglandin I2 and thromboxane-A2 interact with five major receptors (and their sub-types) to elicit specific downstream cellular and tissue actions. In general, prostaglandins have been associated with pain, inflammation, and edema when they are present at high local concentrations and involved on a chronic basis. However, in acute settings, certain endogenous and exogenous prostaglandins have beneficial effects ranging from mediating muscle contraction/relaxation, providing cellular protection, regulating sleep, and enhancing blood flow, to lowering intraocular pressure to prevent the development of glaucoma, a blinding disease. Several classes of prostaglandins are implicated (or are considered beneficial) in certain central nervous system dysfunctions (e.g., Alzheimer’s, Parkinson’s, and Huntington’s diseases; amyotrophic lateral sclerosis and multiple sclerosis; stroke, traumatic brain injuries and pain) and in ocular disorders (e.g., ocular hypertension and glaucoma; allergy and inflammation; edematous retinal disorders). This review endeavors to address the physiological/pathological roles of prostaglandins in the central nervous system and ocular function in health and disease, and provides insights towards the therapeutic utility of some prostaglandin agonists and antagonists, polyunsaturated fatty acids, and cyclooxygenase inhibitors.
Collapse
|
24
|
Marotta C, Giorgi E, Binacchi F, Cirri D, Gabbiani C, Pratesi A. An overview of recent advancements in anticancer Pt(IV) prodrugs: New smart drug combinations, activation and delivery strategies. Inorganica Chim Acta 2023. [DOI: 10.1016/j.ica.2023.121388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
25
|
Hofling U, Tacconelli S, Contursi A, Bruno A, Mucci M, Ballerini P, Cohen S, Patrignani P. Characterization of the acetylation of cyclooxygenase-isozymes and targeted lipidomics of eicosanoids in serum and colon cancer cells by the new aspirin formulation IP1867B versus aspirin in vitro. Front Pharmacol 2022; 13:1070277. [PMID: 36588714 PMCID: PMC9795017 DOI: 10.3389/fphar.2022.1070277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022] Open
Abstract
Background: Aspirin(acetylsalicylic acid, ASA) is recommended for the secondary prevention of atherothrombotic events and has shown anticancer effects. The current enteric-coated drug formulation may reduce aspirin bioavailability. Liquid formulations could improve aspirin pharmacokinetics and pharmacodynamics. IP1867B is a liquid-aspirin formulation that combines three ingredients, ASA/triacetin/saccharin. Methods: ASA and IP1867B(L-ASA) were assessed in human serum(obtained by allowing to clot human whole blood at 37 °C for 1h), washed platelets, and colonic adenocarcinoma HCA7 cells on eicosanoid generation and COX-isozyme acetylation at Serine529 and 516 by LC-MS/MS. Results: In serum, ASA and L-ASA acted by selectively affecting COX-1-derived eicosanoids, including thromboxane(TX)B2. L-ASA was more potent in inhibiting serum TXB2, a known biomarker of aspirin antiplatelet effect, than ASA. However, ASA and L-ASA were equipotent to acetylate COX-1 in washed platelets and COX-2 in HCA7 cells. In HCA7 cells, ASA and L-ASA acted by inhibiting prostaglandin(PG)E2(the most abundant prostanoid) and TXB2 biosynthesis. In the presence of a high arachidonic acid concentration(100 μM), 15R-hydroxyeicosatetraenoic acid(HETE) was generated at baseline by cancer cell COX-2 and was only slightly enhanced by supratherapeutic concentrations of ASA(1 mM). In whole blood and HCA7 cells treated with ASA or L-ASA, 15-epi-lipoxin(LX)A4 were undetectable. Conclusion: IP1867B was more potent in affecting serum TXB2 generation than ASA. The relevance of this finding deserves evaluation in vivo in humans. In cancer cells, ASA and IP1867B acted by inhibiting PGE2 and TXB2 generation via the acetylation of COX-2. ASA and IP867B at clinically relevant concentrations did not substantially induce the biosynthesis of 15R-HETE and 15-epi-LXA4.
Collapse
Affiliation(s)
- Ulrika Hofling
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, Medical School, “G. D’Annunzio” University, Chieti, Italy
| | - Stefania Tacconelli
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, Medical School, “G. D’Annunzio” University, Chieti, Italy
| | - Annalisa Contursi
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, Medical School, “G. D’Annunzio” University, Chieti, Italy
| | - Annalisa Bruno
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, Medical School, “G. D’Annunzio” University, Chieti, Italy
| | - Matteo Mucci
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, Medical School, “G. D’Annunzio” University, Chieti, Italy
| | - Patrizia Ballerini
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, “G. D’Annunzio” University, Chieti, Italy
| | - Simon Cohen
- Innovate Pharmaceuticals, Manchester, United Kingdom
| | - Paola Patrignani
- Center for Advanced Studies and Technology (CAST), “G. D’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, Medical School, “G. D’Annunzio” University, Chieti, Italy
| |
Collapse
|
26
|
Bruno A, Contursi A, Tacconelli S, Sacco A, Hofling U, Mucci M, Lamolinara A, Del Pizzo F, Ballerini P, Di Gregorio P, Yu Y, Patrignani P. The specific deletion of cyclooxygenase-1 in megakaryocytes/platelets reduces intestinal polyposis in Apc Min/+ mice. Pharmacol Res 2022; 185:106506. [PMID: 36241001 DOI: 10.1016/j.phrs.2022.106506] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/26/2022] [Accepted: 10/09/2022] [Indexed: 10/31/2022]
Abstract
Clinical and experimental evidence sustain the role of cyclooxygenase (COX)-1 in intestinal tumorigenesis. However, the cell type expressing the enzyme involved and molecular mechanism(s) have not been clarified yet. We aimed to elucidate the role of platelet COX-1 (the target of low-dose aspirin in humans) in intestinal tumorigenesis of ApcMin/+ mice, considered a clinically relevant model. To realize this objective, we generated an ApcMin/+ mouse with a specific deletion of Ptgs1(COX-1 gene name) in megakaryocytes/platelets (ApcMin/+;pPtgs1-/-mice) characterized by profound inhibition of thromboxane(TX)A2 biosynthesis ex vivo (serum TXB2; by 99%) and in vivo [urinary 2,3-dinor-TXB2(TXM), by 79%]. ApcMin/+ mice with the deletion of platelet COX-1 showed a significantly reduced number (67%) and size (32%) of tumors in the small intestine. The intestinal adenomas of these mice had decreased proliferative index associated with reduced COX-2 expression and systemic prostaglandin(PG)E2 biosynthesis (urinary PGEM) vs. ApcMin/+mice. Extravasated platelets were detected in the intestine of ApcMin/+mice. Thus, we explored their contribution to COX-2 induction in fibroblasts, considered the primary polyp cell type expressing the protein. In the coculture of human platelets and myofibroblasts, platelet-derived TXA2 was involved in the induction of COX-2-dependent PGE2 in myofibroblasts since it was prevented by the selective inhibition of platelet COX-1 by aspirin or by a specific antagonist of TXA2 receptors. In conclusion, our results support the platelet hypothesis of intestinal tumorigenesis and provide experimental evidence that selective inhibition of platelet COX-1 can mitigate early events of intestinal tumorigenesis by restraining COX-2 induction.
Collapse
Affiliation(s)
- Annalisa Bruno
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Annalisa Contursi
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Stefania Tacconelli
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Angela Sacco
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Ulrika Hofling
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Matteo Mucci
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Alessia Lamolinara
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Francesco Del Pizzo
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Patrizia Ballerini
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, "G.d'Annunzio" University, 66100 Chieti, Italy
| | - Patrizia Di Gregorio
- Institute of Transfusion Medicine, "Ss. Annunziata" Hospital, 66100 Chieti, Italy
| | - Ying Yu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Paola Patrignani
- Center for Advanced Studies and Technology (CAST), "G.d'Annunzio" University, 66100 Chieti, Italy; Department of Neuroscience, Imaging and Clinical Science, "G.d'Annunzio" University, 66100 Chieti, Italy.
| |
Collapse
|
27
|
Turanlı S, Ergül AG, Jordan PM, Olğaç A, Çalışkan B, Werz O, Banoglu E. Quinazoline-4(3 H)-one-7-carboxamide Derivatives as Human Soluble Epoxide Hydrolase Inhibitors with Developable 5-Lipoxygenase Activating Protein Inhibition. ACS OMEGA 2022; 7:36354-36365. [PMID: 36278102 PMCID: PMC9583330 DOI: 10.1021/acsomega.2c04039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/22/2022] [Indexed: 06/16/2023]
Abstract
Soluble epoxide hydrolase (sEH) metabolizes epoxyeicosatrienoic acids (EETs), which are endowed with beneficial biological activities as they reduce inflammation, regulate endothelial tone, improve mitochondrial function, and decrease oxidative stress. Therefore, inhibition of sEH for maintaining high EET levels is implicated as a new therapeutic modality with broad clinical applications for metabolic, renal, and cardiovascular disorders. In our search for new sEH inhibitors, we designed and synthesized novel amide analogues of the quinazolinone-7-carboxylic acid derivative 5, a previously discovered 5-lipoxygenase-activating protein (FLAP) inhibitor, to evaluate their potential for inhibiting sEH. As a result, we identified new quinazolinone-7-carboxamides that demonstrated selective sEH inhibition with decreased FLAP inhibitor properties. The tractable SAR results indicated that the amide and thiobenzyl fragments flanking the quinazolinone nucleus are critical features governing the potent sEH inhibition, and compounds 34, 35, 37, and 43 inhibited the sEH activity with IC50 values of 0.30-0.66 μM. Compound 34 also inhibited the FLAP-mediated leukotriene biosynthesis (IC50 = 2.91 μM). In conclusion, quinazolinone-7-carboxamides can be regarded as novel lead structures, and newer analogues with improved efficiency against sEH along with or without FLAP inhibition can be generated.
Collapse
Affiliation(s)
- Sümeyye Turanlı
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No: 3 Yenimahalle, 06560 Ankara, Turkey
| | - Azize Gizem Ergül
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No: 3 Yenimahalle, 06560 Ankara, Turkey
| | - Paul M. Jordan
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-7743 Jena, Germany
| | - Abdurrahman Olğaç
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No: 3 Yenimahalle, 06560 Ankara, Turkey
| | - Burcu Çalışkan
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No: 3 Yenimahalle, 06560 Ankara, Turkey
| | - Oliver Werz
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-7743 Jena, Germany
| | - Erden Banoglu
- Department
of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Taç Sok. No: 3 Yenimahalle, 06560 Ankara, Turkey
| |
Collapse
|
28
|
Schulte B, Tergast TL, Griemsmann M, Menti D, Deveci N, Kahlhöfer J, Dörge P, Hüffner L, Kraft ARM, Behrendt P, Wedemeyer H, Cornberg M, Stichtenoth DO, Maasoumy B. Metamizole-Associated Risks in Decompensated Hepatic Cirrhosis. DEUTSCHES ARZTEBLATT INTERNATIONAL 2022; 119:687-693. [PMID: 35912424 PMCID: PMC9830680 DOI: 10.3238/arztebl.m2022.0280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/11/2022] [Accepted: 07/04/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Because of the increased risk of acute renal failure (ARF), the use of cyclooxygenase (COX) inhibitors is not recommended in patients with decompensated hepatic cirrhosis. Metamizole is not a classic COX inhibitor, but there are insufficient data to support its safe use. In this study, we investigate the effect of metamizole on the risk of ARF in these patients. METHODS Metamizole use, ARF incidence, and patient mortality were examined in a large, retrospective, exploratory cohort and validated with data from a prospective registry. RESULTS 523 patients were evaluated in the exploratory cohort. Metamizole use at baseline was documented in 110 cases (21%) and was independently associated with the development of ARF, severe (grade 3) ARF, and lower survival without liver transplantation at follow-up on day 28 (HR: 2.2, p < 0.001; HR: 2.8, p < 0.001; and HR: 2.6, p < 0.001, respectively). Interestingly, the risk of ARF depended on the dose of metamizole administered (HR: 1.038, p < 0.001). Compared to patients who were treated with opioids, the rate of ARF was higher in the metamizole group (49% vs. 79%, p = 0.014). An increased risk of ARF with metamizole use was also demonstrated in the independent validation cohort (p < 0.001). CONCLUSION Metamizole therapy, especially at high doses, should only be used with a high level of caution in patients with decompensated cirrhosis.
Collapse
Affiliation(s)
| | - Tammo L. Tergast
- * Joint first authors,Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School
| | - Marie Griemsmann
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School
| | - Denise Menti
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School,Center for Individualized Infection Medicine (CiiM), Hannover
| | - Neslihan Deveci
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School
| | - Julia Kahlhöfer
- German Center for Infection Research (DZIF), German Liver Foundation, HepNet, Hannover
| | - Petra Dörge
- German Center for Infection Research (DZIF), German Liver Foundation, HepNet, Hannover
| | - Lucas Hüffner
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School,Twincore, Center for Experimental and Clinical Infection Research, Hannover
| | - Anke R. M. Kraft
- * Joint first authors,German Center for Infection Research (DZIF), Hannover-Braunschweig
| | - Patrick Behrendt
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School,German Center for Infection Research (DZIF), Hannover-Braunschweig
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School,German Center for Infection Research (DZIF), Hannover-Braunschweig
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School,Center for Individualized Infection Medicine (CiiM), Hannover,Twincore, Center for Experimental and Clinical Infection Research, Hannover,German Center for Infection Research (DZIF), Hannover-Braunschweig
| | | | - Benjamin Maasoumy
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School,German Center for Infection Research (DZIF), Hannover-Braunschweig,*Klinik für Gastroenterologie, Hepatologie und Endokrinologie Medizinische Hochschule Hannover Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
29
|
Patrono C. Aspirin: 1A @ 125. Eur Heart J 2022; 43:3194-3195. [PMID: 35938360 DOI: 10.1093/eurheartj/ehac416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Carlo Patrono
- Pharmacology Section, Department of Safety and Bioethics, Catholic University School of Medicine, Largo F. Vito 1, 00168 Rome, Italy
| |
Collapse
|
30
|
Patrignani P. Editorial: Women in Inflammation Pharmacology: 2021. Front Pharmacol 2022; 13:973691. [PMID: 35910361 PMCID: PMC9326490 DOI: 10.3389/fphar.2022.973691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
|
31
|
Pagano M, Savoca S, Impellitteri F, Albano M, Capillo G, Faggio C. Toxicological Evaluation of Acetylsalicylic Acid in Non-Target Organisms: Chronic Exposure on Mytilus galloprovincialis (Lamarck, 1819). Front Physiol 2022; 13:920952. [PMID: 35899021 PMCID: PMC9309544 DOI: 10.3389/fphys.2022.920952] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/23/2022] [Indexed: 11/17/2022] Open
Abstract
Pharmaceuticals are now considered to be established contaminants, and their presence in water poses a real risk not only to the marine ecosystem, as they may adversely affect non-target organisms that are exposed to them, but also indirectly to humans. This is particularly true for the model organism considered in this work, Mytilus galloprovincialis (Lamarck, 1819), a suspensivore and bioaccumulating organism that enters the human food chain. Among the most commonly used over-the-counter medicines, anti-inflammatory drugs certainly feature prominently, with acetylsalicylic acid (ASA) at the top. In this work, M. galloprovincialis specimens were exposed to two concentrations of ASA (10 and 100 μg/L) for 10 and 20 days to evaluate possible alterations in the decrease in regulatory volume (RVD) in digestive gland cells and cell viability of both these cells and hemocytes. In addition, the histopathological condition index of the gills and digestive gland was evaluated. The data obtained showed that chronic exposure to ASA did not alter the cell viability of hemocytes and digestive gland cells but alters the physiological mechanisms of volume regulation in the digestive gland and, in addition, a time-dose reaction to ASA in the gills and digestive gland showing numerous alterations such as lipofuscin deposits and hemocyte infiltration was found. These results confirm the potential toxicity to the marine biota, highlighting the necessity to deepen the knowledge regarding the link between over-the-counter pharmaceuticals and non-target organisms.
Collapse
Affiliation(s)
- M. Pagano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - S. Savoca
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Messina, Italy
- Institute for Marine Biological Resources and Biotechnology (IRBIM), National Research Council (CNR), Messina, Italy
| | - F. Impellitteri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - M. Albano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - G. Capillo
- Institute for Marine Biological Resources and Biotechnology (IRBIM), National Research Council (CNR), Messina, Italy
- Department of Veterinary Sciences, Polo Universitario Dell’Annunziata, University of Messina, Messina, Italy
| | - C. Faggio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
- *Correspondence: C. Faggio,
| |
Collapse
|
32
|
Luan X, Cong Z, Anastassiades TP, Gao Y. N-Butyrylated Hyaluronic Acid Achieves Anti-Inflammatory Effects In Vitro and in Adjuvant-Induced Immune Activation in Rats. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103267. [PMID: 35630747 PMCID: PMC9145605 DOI: 10.3390/molecules27103267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 12/20/2022]
Abstract
Previously synthesized N-butyrylated hyaluronic acid (BHA) provides anti-inflammatory effects in rat models of acute gouty arthritis and hyperuricemia. However, the mechanism of action remains to be elucidated. Herein, the anti-inflammatory and antioxidative activities of BHA and the targeted signaling pathways were explored with LPS-induced RAW264.7 and an adjuvant-induced inflammation in a rat model. Results indicated that BHA inhibited the generation of pro-inflammatory cytokines TNFα, IL-1β and IL-6, reduced ROS production and down-regulated JAK1-STAT1/3 signaling pathways in LPS-induced RAW264.7. In vivo, BHA alleviated paw and joint swelling, decreased inflammatory cell infiltration in paw tissues, suppressed gene expressions of p38 and p65, down-regulated the NF-κB and MAPK signaling pathways and reduced protein levels of TNFα, IL-1β and IL-6 in joint tissues of arthritis rats. This study demonstrated the pivotal role of BHA in anti-inflammation and anti-oxidation, suggesting the potential clinical value of BHA in the prevention of inflammatory arthritis and is worthy for development as a new pharmacological treatment.
Collapse
Affiliation(s)
- Xue Luan
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China; (X.L.); (Z.C.)
| | - Zhongcheng Cong
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China; (X.L.); (Z.C.)
| | - Tassos P. Anastassiades
- Division of Rheumatology, Department of Medicine, Queen’s University, Kingston, ON K7L 4B4, Canada;
| | - Yin Gao
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China; (X.L.); (Z.C.)
- Correspondence: ; Fax: +86-431-85168175
| |
Collapse
|
33
|
New uses of ketoprofen – a review of studies from 2015 to 2021. CURRENT ISSUES IN PHARMACY AND MEDICAL SCIENCES 2022. [DOI: 10.2478/cipms-2022-0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abstract
Ketoprofen (K) belongs to the family of nonsteroidal anti-inflammatory drugs (NSAIDs) and demonstrates analgesic, anti-inflammatory and antipyretic properties. K is one of the most commonly used NSAIDs because of the speed and effectiveness of its activity. K is currently used for the treatment of pain and treatment of symptoms in rheumatic diseases, however, many researchers are looking for new uses of K. The aim of the review was to present the possible applications of K as indicated in current literature. We searched research literature and compiled all the reports (2015 onwards) we could find about new possible employments of K in health practices. Many studies have been aimed at obtaining new uses of K. This article describes the use of ketoprofen lysine salt for treating injured gastric mucosa, the anti-allergic potential of K, the employment of K in treating nonalcoholic fatty liver disease, human lymphedema and seizures, as well as the antidepressant and anxiolytic effects of K, prospects for the use of K in oncology and transplantology. The findings of the review confirm that K, its derivatives and complexes have many newly discovered effects. It is likely that in the future, K will have more indications than it has today.
Collapse
|
34
|
Çapan İ, Jordan PM, Olğaç A, Çalışkan B, Kretzer C, Werz O, Banoglu E. Discovery and optimization of piperazine urea derivatives as soluble epoxide hydrolase (sEH) inhibitors. ChemMedChem 2022; 17:e202200137. [DOI: 10.1002/cmdc.202200137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/21/2022] [Indexed: 11/05/2022]
Affiliation(s)
- İrfan Çapan
- Gazi University: Gazi Universitesi Department of Material and Material Processing Technologies TURKEY
| | - Paul M. Jordan
- Friedrich Schiller University Jena: Friedrich-Schiller-Universitat Jena Medicinal Chemsitry GERMANY
| | | | - Burcu Çalışkan
- Gazi University: Gazi Universitesi Pharmaceutical Chemistry TURKEY
| | | | - Oliver Werz
- Friedrich Schiller University Jena: Friedrich-Schiller-Universitat Jena Medicinal Chemistry GERMANY
| | - Erden Banoglu
- Gazi Universitesi Eczacilik Fakultesi Pharmaceutical Chemistry Tac Sokak No 3 06580 Ankara TURKEY
| |
Collapse
|
35
|
Maternal Inflammation Exaggerates Offspring Susceptibility to Cerebral Ischemia–Reperfusion Injury via the COX-2/PGD2/DP2 Pathway Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1571705. [PMID: 35437456 PMCID: PMC9013311 DOI: 10.1155/2022/1571705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 12/04/2022]
Abstract
The pathogenesis of cerebral ischemia–reperfusion (I/R) injury is complex and does not exhibit an effective strategy. Maternal inflammation represents one of the most important factors involved in the etiology of brain injury in newborns. We aimed to investigate the effect of maternal inflammation on offspring susceptibility to cerebral I/R injury and the mechanisms by which it exerts its effects. Pregnant SD rats were intraperitoneally injected with LPS (300 μg/kg/day) at gestational days 11, 14, and 18. Pups were subjected to MCAO/R on postnatal day 60. Primary neurons were obtained from postnatal day 0 SD rats and subjected to OGD/R. Neurological deficits, brain injury, neuronal viability, neuronal damage, and neuronal apoptosis were assessed. Oxidative stress and inflammation were evaluated, and the expression levels of COX-2/PGD2/DP pathway-related proteins and apoptotic proteins were detected. Maternal LPS exposure significantly increased the levels of oxidative stress and inflammation, significantly activated the COX-2/PGD2/DP2 pathway, and increased proapoptotic protein expression. However, maternal LPS exposure significantly decreased the antiapoptotic protein expression, which subsequently increased neurological deficits and cerebral I/R injury in offspring rats. The corresponding results were observed in primary neurons. Moreover, these effects of maternal LPS exposure were reversed by a COX-2 inhibitor and DP1 agonist but exacerbated by a DP2 agonist. In conclusion, maternal inflammatory exposure may increase offspring susceptibility to cerebral I/R injury. Moreover, the underlying mechanism might be related to the activation of the COX-2/PGD2/DP2 pathway. These findings provide a theoretical foundation for the development of therapeutic drugs for cerebral I/R injury.
Collapse
|
36
|
Boizet-Bonhoure B, Déjardin S, Rossitto M, Poulat F, Philibert P. Using Experimental Models to Decipher the Effects of Acetaminophen and NSAIDs on Reproductive Development and Health. FRONTIERS IN TOXICOLOGY 2022; 4:835360. [PMID: 35295217 PMCID: PMC8915900 DOI: 10.3389/ftox.2022.835360] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/07/2022] [Indexed: 01/04/2023] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs), such as aspirin (acetylsalicylic acid), diclofenac and ibuprofen (IBU), and analgesic drugs, such as acetaminophen (APAP, or paracetamol), are widely used to treat inflammation and pain. APAP and IBU are over-the-counter drugs and are among the most commonly taken drugs in the first trimester of pregnancy, even in combination. Furthermore, these drugs and their metabolites are released in the environment, and can be frequently detected in wastewater, surface water, and importantly in drinking water. Although their environmental concentrations are much lower than the therapeutics doses, this suggests an uncontrolled low-dose exposure of the general population, including pregnant women and young children, two particularly at risk populations. Epidemiological studies show that exposure to these molecules in the first and second trimester of gestation can favor genital malformations in new-born boys. To investigate the cellular, molecular and mechanistic effects of exposure to these molecules, ex vivo studies with human or rodent gonadal explants and in vivo experiments in rodents have been performed in the past years. This review recapitulates recent data obtained in rodent models after in utero or postnatal exposure to these drugs. The first part of this review discusses the mechanisms by which NSAIDs and analgesics may impair gonadal development and maturation, puberty development, sex hormone production, maturation and function of adult organs, and ultimately fertility in the exposed animals and their offspring. Like other endocrine disruptors, NSAIDs and APAP interfere with endocrine gland function and may have inter/transgenerational adverse effects. Particularly, they may target germ cells, resulting in reduced quality of male and female gametes, and decreased fertility of exposed individuals and their descendants. Then, this review discusses the effects of exposure to a single drug (APAP, aspirin, or IBU) or to combinations of drugs during early embryogenesis, and the consequences on postnatal gonadal development and adult reproductive health. Altogether, these data may increase medical and public awareness about these reproductive health concerns, particularly in women of childbearing age, pregnant women, and parents of young children.
Collapse
Affiliation(s)
- Brigitte Boizet-Bonhoure
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
- *Correspondence: Brigitte Boizet-Bonhoure,
| | - Stéphanie Déjardin
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
| | | | - Francis Poulat
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
| | - Pascal Philibert
- Institute of Human Genetics, CNRS, University of Montpellier, Montpellier, France
- Laboratory of Biochemistry and Molecular Biology, Carèmeau Hospital, Nîmes University Hospital, Nîmes, France
| |
Collapse
|
37
|
Ballerini P, Contursi A, Bruno A, Mucci M, Tacconelli S, Patrignani P. Inflammation and Cancer: From the Development of Personalized Indicators to Novel Therapeutic Strategies. Front Pharmacol 2022; 13:838079. [PMID: 35308229 PMCID: PMC8927697 DOI: 10.3389/fphar.2022.838079] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/09/2022] [Indexed: 12/15/2022] Open
Abstract
Colorectal (CRC) and hepatocellular carcinoma (HCC) are associated with chronic inflammation, which plays a role in tumor development and malignant progression. An unmet medical need in these settings is the availability of sensitive and specific noninvasive biomarkers. Their use will allow surveillance of high-risk populations, early detection, and monitoring of disease progression. Moreover, the characterization of specific fingerprints of patients with nonalcoholic fatty liver disease (NAFLD) without or with nonalcoholic steatohepatitis (NASH) at the early stages of liver fibrosis is necessary. Some lines of evidence show the contribution of platelets to intestinal and liver inflammation. Thus, low-dose Aspirin, an antiplatelet agent, reduces CRC and liver cancer incidence and mortality. Aspirin also produces antifibrotic effects in NAFLD. Activated platelets can trigger chronic inflammation and tissue fibrosis via the release of soluble mediators, such as thromboxane (TX) A2 and tumor growth factor (TGF)-β, and vesicles containing genetic material (including microRNA). These platelet-derived products contribute to cyclooxygenase (COX)-2 expression and prostaglandin (PG)E2 biosynthesis by tumor microenvironment cells, such as immune and endothelial cells and fibroblasts, alongside cancer cells. Enhanced COX-2-dependent PGE2 plays a crucial role in chronic inflammation and promotes tumor progression, angiogenesis, and metastasis. Antiplatelet agents can indirectly prevent the induction of COX-2 in target cells by inhibiting platelet activation. Differently, selective COX-2 inhibitors (coxibs) block the activity of COX-2 expressed in the tumor microenvironment and cancer cells. However, coxib chemopreventive effects are hampered by the interference with cardiovascular homeostasis via the coincident inhibition of vascular COX-2-dependent prostacyclin biosynthesis, resulting in enhanced risk of atherothrombosis. A strategy to improve anti-inflammatory agents' use in cancer prevention could be to develop tissue-specific drug delivery systems. Platelet ability to interact with tumor cells and transfer their molecular cargo can be employed to design platelet-mediated drug delivery systems to enhance the efficacy and reduce toxicity associated with anti-inflammatory agents in these settings. Another peculiarity of platelets is their capability to uptake proteins and transcripts from the circulation. Thus, cancer patient platelets show specific proteomic and transcriptomic expression profiles that could be used as biomarkers for early cancer detection and disease monitoring.
Collapse
Affiliation(s)
- Patrizia Ballerini
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, Chieti, Italy
| | - Annalisa Contursi
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, G. d’Annunzio University, Chieti, Italy
| | - Annalisa Bruno
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, G. d’Annunzio University, Chieti, Italy
| | - Matteo Mucci
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, G. d’Annunzio University, Chieti, Italy
| | - Stefania Tacconelli
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, G. d’Annunzio University, Chieti, Italy
| | - Paola Patrignani
- Center for Advanced Studies and Technology (CAST), Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, G. d’Annunzio University, Chieti, Italy
| |
Collapse
|
38
|
Serrano D, Patrignani P, Stigliano V, Turchetti D, Sciallero S, Roviello F, D’Arpino A, Grattagliano I, Testa S, Oliani C, Bertario L, Bonanni B. Aspirin Colorectal Cancer Prevention in Lynch Syndrome: Recommendations in the Era of Precision Medicine. Genes (Basel) 2022; 13:460. [PMID: 35328014 PMCID: PMC8952565 DOI: 10.3390/genes13030460] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer prevention in the era of precision medicine has to consider integrated therapeutic approaches. Therapeutic cancer prevention should be offered to selected cohorts with increased cancer risk. Undoubtedly, carriers of hereditary cancer syndromes have a well-defined high cancer risk. Lynch Syndrome is one of the most frequent hereditary syndromes; it is mainly associated with colorectal cancer (CRC). Nonsteroidal anti-inflammatory drugs and, in particular, aspirin use, has been associated with reduced CRC risk in several studies, initially with contradictory results; however, longer follow-up confirmed a reduced CRC incidence and mortality. The CAPP2 study recruited 861 Lynch syndrome participants randomly assigned to 600 mg of aspirin versus placebo. Like sporadic CRCs, a significant CRC risk reduction was seen after an extended follow-up, with a median treatment time that was relatively short (2 years). The ongoing CAPP3 will address whether lower doses are equally effective. Based on pharmacology and clinical data on sporadic CRCs, the preventive effect should also be obtained with low-dose aspirin. The leading international guidelines suggest discussing with Lynch syndrome carriers the possibility of using low-dose aspirin for CRC prevention. We aim systematically promote this intervention with all Lynch syndrome carriers.
Collapse
Affiliation(s)
- Davide Serrano
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.S.); (B.B.)
| | - Paola Patrignani
- Department of Neuroscience, Imaging and Clinical Sciences, and CAST, “G. d’Annunzio” University, 66100 Chieti, Italy;
| | - Vittoria Stigliano
- Division of Gastroenterology and Digestive Endoscopy, IRCCS, Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Daniela Turchetti
- Center for Hereditary Cancer, Department of Medical and Surgical Sciences, University of Bologna, 40100 Bologna, Italy;
| | | | - Franco Roviello
- Unit of General Surgery and Surgical Oncology, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy;
| | - Alessandro D’Arpino
- Hospital Pharmacy Unit, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, 06100 Perugia, Italy;
| | | | | | - Cristina Oliani
- Ambulatorio Familiarita’ Neoplastica UOC Oncologia Medica ULSS5 Polesana, 45100 Rovigo, Italy;
| | - Lucio Bertario
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.S.); (B.B.)
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.S.); (B.B.)
| |
Collapse
|
39
|
Rovati G, Contursi A, Bruno A, Tacconelli S, Ballerini P, Patrignani P. Antiplatelet Agents Affecting GPCR Signaling Implicated in Tumor Metastasis. Cells 2022; 11:725. [PMID: 35203374 PMCID: PMC8870128 DOI: 10.3390/cells11040725] [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: 01/17/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 11/16/2022] Open
Abstract
Metastasis requires that cancer cells survive in the circulation, colonize distant organs, and grow. Despite platelets being central contributors to hemostasis, leukocyte trafficking during inflammation, and vessel stability maintenance, there is significant evidence to support their essential role in supporting metastasis through different mechanisms. In addition to their direct interaction with cancer cells, thus forming heteroaggregates such as leukocytes, platelets release molecules that are necessary to promote a disseminating phenotype in cancer cells via the induction of an epithelial-mesenchymal-like transition. Therefore, agents that affect platelet activation can potentially restrain these prometastatic mechanisms. Although the primary adhesion of platelets to cancer cells is mainly independent of G protein-mediated signaling, soluble mediators released from platelets, such as ADP, thromboxane (TX) A2, and prostaglandin (PG) E2, act through G protein-coupled receptors (GPCRs) to cause the activation of more additional platelets and drive metastatic signaling pathways in cancer cells. In this review, we examine the contribution of the GPCRs of platelets and cancer cells in the development of cancer metastasis. Finally, the possible use of agents affecting GPCR signaling pathways as antimetastatic agents is discussed.
Collapse
Affiliation(s)
- Gianenrico Rovati
- Department of Pharmaceutical Sciences, University of Milan, 20122 Milan, Italy;
| | - Annalisa Contursi
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Annalisa Bruno
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Stefania Tacconelli
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Patrizia Ballerini
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Paola Patrignani
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| |
Collapse
|
40
|
Meng Q, Li B, Huang N, Wei S, Ren Q, Wu S, Li X, Chen R. Folic acid targets splenic extramedullary hemopoiesis to attenuate carbon black-induced coagulation-thrombosis potential. JOURNAL OF HAZARDOUS MATERIALS 2022; 424:127354. [PMID: 34634699 DOI: 10.1016/j.jhazmat.2021.127354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/14/2021] [Accepted: 09/23/2021] [Indexed: 06/13/2023]
Abstract
Due to its wide applications in tire and rubber products, carbon black (CB) implicates concerns on its safety during production, collection, and handling. Here we report that exposure CB, increases coagulation-thrombosis potential in a splenic extramedullary hemopoiesis (EMH)-dependent manner. Adult C57BL/6 mice are kept in whole-body inhalation chambers, and exposed to filtered room air (FRA) or CB for 28 consecutive days. CB exposure resulted in splenic EMH characterized with platelet precursor cells, megakaryocytes (MKs), hyperplasia and enhanced in vivo blood coagulation ability. Metabolomics analysis suggests significant enhance in PGE2 production but reduction in folic acid (FA) levels in murine serum following CB exposure. Mechanistically, activation of COX-dependent PGE2 production promotes IL-6 expression in splenic macrophages, which subsequently results in splenic EMH and increased platelet counts in circulation. Administration of FA protects the mice against CB-induced splenic EMH through inhibiting prostaglandin-endoperoxide synthase 2 (Ptgs2 or Cox2) and prostaglandin E synthase (Ptges) expression in splenic macrophages, eventually recover the coagulation capacity to normal level. The results strongly suggest the involvement of splenic EMH in response to CB exposure and subsequently increased coagulation-thrombosis potential. Supplementation with FA may be a candidate to prevent thrombosis potential attributable to CB exposure.
Collapse
Affiliation(s)
- Qingtao Meng
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, PR China
| | - Bin Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Nannan Huang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Shengnan Wei
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China
| | - Quanzhong Ren
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, PR China
| | - Shenshen Wu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, PR China
| | - Xiaobo Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, PR China.
| | - Rui Chen
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, PR China; School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, PR China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
41
|
Zhai Z, Zhang X, Ding Y, Huang Z, Li Q, Zheng M, Cho K, Dong Z, Fu W, Chen Z, Jiang B. Eugenol restrains abdominal aortic aneurysm progression with down‐regulations on
NF‐κB
and
COX
‐2. Phytother Res 2022; 36:928-937. [PMID: 35132703 DOI: 10.1002/ptr.7358] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 11/14/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Ziyi Zhai
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- College of Pharmacy, China Medical University, Liaoning, China
| | - Xianjing Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yuchao Ding
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- College of Pharmacy, China Medical University, Liaoning, China
| | - Ziming Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qian Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- College of Pharmacy, China Medical University, Liaoning, China
| | - Mingyue Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Kenka Cho
- Department of Acupuncture, Takarazuka University of Medical and Health Care, Takarazuka-city, Japan
| | - Zhihui Dong
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiguo Fu
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zaixing Chen
- College of Pharmacy, China Medical University, Liaoning, China
| | - Baohong Jiang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
42
|
Kretzer C, Jordan PM, Bilancia R, Rossi A, Gür Maz T, Banoglu E, Schubert US, Werz O. Shifting the Biosynthesis of Leukotrienes Toward Specialized Pro-Resolving Mediators by the 5-Lipoxygenase-Activating Protein (FLAP) Antagonist BRP-201. J Inflamm Res 2022; 15:911-925. [PMID: 35173459 PMCID: PMC8842732 DOI: 10.2147/jir.s345510] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
Background and Purpose Lipid mediators (LM) play crucial roles in the complex inflammation process with respect to initiation, maintenance, and resolution. Proinflammatory leukotrienes (LTs), generated by 5-lipoxygenase (LOX) and the 5-LOX-activating protein (FLAP), initiate and maintain inflammation while specialized pro-resolving mediators (SPMs) formed by various LOXs as key enzymes promote inflammation resolution and the return to homeostasis. Since 5-LOX also contributes to SPM biosynthesis, smart pharmacological manipulation of the 5-LOX pathway and accompanied activation of 12-/15-LOXs may accomplish suppression of LT formation but maintain or even elevate SPM formation. Here, we demonstrated that the FLAP antagonist BRP-201 possesses such pharmacological profile and causes a switch from LT toward SPM formation. Methods and Results Comprehensive LM metabololipidomics with activated human monocyte-derived macrophages (MDM) of M1 or M2 phenotype showed that BRP-201 strongly inhibits LT formation induced by bacterial exotoxins. In parallel, SPM levels and 12/15-LOX-derived products were markedly elevated, in particular in M2-MDM. Intriguingly, in unstimulated MDM, BRP-201 induced formation of 12/15-LOX products including SPM and caused 15-LOX-1 subcellular redistribution without affecting 5-LOX. Experiments with HEK293 cells stably expressing either 5-LOX with or without FLAP, 15-LOX-1 or 15-LOX-2 confirmed suppression of 5-LOX product formation due to FLAP antagonism by BRP-201 but activated 15-LOX-1 in the absence of FLAP. Finally, in zymosan-induced murine peritonitis, BRP-201 (2 mg/kg, ip) lowered LT levels but elevated 12/15-LOX products including SPMs. Conclusion BRP-201 acts as FLAP antagonist but also as 12/15-LOX activator switching formation of pro-inflammatory LTs toward inflammation-resolving SPM, which reflects a beneficial pharmacological profile for intervention in inflammation.
Collapse
Affiliation(s)
- Christian Kretzer
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, 07743, Germany
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, 07743, Germany
| | - Rossella Bilancia
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, I-80131, Italy
| | - Antonietta Rossi
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, I-80131, Italy
| | - Tuğçe Gür Maz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Erden Banoglu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Yenimahalle, 06560, Ankara, Turkey
| | - Ulrich S Schubert
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Jena, 07743, Germany
- Jena Center for Soft Matter (JCSM) Friedrich Schiller University Jena, Jena, 07743, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, 07743, Germany
- Jena Center for Soft Matter (JCSM) Friedrich Schiller University Jena, Jena, 07743, Germany
- Correspondence: Oliver Werz, Email
| |
Collapse
|
43
|
Magni A, Agostoni P, Bonezzi C, Massazza G, Menè P, Savarino V, Fornasari D. Management of Osteoarthritis: Expert Opinion on NSAIDs. Pain Ther 2021; 10:783-808. [PMID: 33876393 PMCID: PMC8586433 DOI: 10.1007/s40122-021-00260-1] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/27/2021] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is a leading cause of disability among older adults worldwide. Treatment aims are to alleviate inflammatory pain and improve physical function through non-pharmacological and pharmacological interventions. Non-steroidal anti-inflammatory drugs (NSAIDs) are recommended as first-line therapy. However, selection is challenged by patient age, comorbidities and polypharmacy, and by the drug's benefit/risk balance, all of which together influence the risk of cardiovascular (CV), gastrointestinal (GI) and renal adverse events (AEs). While the efficacy profile of the various NSAIDs is delineated, the differences in their safety profile are not straightforward. This narrative review provides practical indications by a multidisciplinary Italian expert panel for general practitioners and specialists managing OA patients with chronic inflammatory pain; the goal is to maximize therapy efficacy while reducing untoward effects caused by inappropriate NSAID use. The discussion on the best approach to NSAIDs spanned the following topics: (1) patient evaluation: investigate pain origin, duration and components together with possible risk factors for CV, GI and renal AEs; (2) non-pharmacological interventions: the physiatrist provides a person-centered, holistic approach accounting for all patient aspects; (3) pharmacological interventions: patient profile and drugs' pharmacological properties affect NSAID selection, which drugs to be used in combination or to be avoided, formulation and therapy duration; (4) the pharmacologist's, general practitioner's and pain therapist's points of view; (5) NSAID safety: the individual baseline risk and the drug's safety profile are major determinants of CV, GI and renal risk; consider possible drug-drug interactions; (6) periodical re-evaluation of treatment response and adherence, using scales to assess pain and function.
Collapse
Affiliation(s)
- Alberto Magni
- Italian College of General Practitioners and Primary Care, Via Del Sansovino 179, Florence, Italy
| | - Piergiuseppe Agostoni
- Centro Cardiologico Monzino, IRCCS, Via Carlo Parea, 4, Milan, Italy ,Dipartimento di scienze cliniche e di comunità, Università degli Studi di Milano, Via Carlo Parea 4, Milan, Italy
| | - Cesare Bonezzi
- Unità di Terapia del dolore, Istituti Clinici Scientifici Maugeri, Via Salvatore Maugeri 10, Pavia, Italy
| | - Giuseppe Massazza
- Division of Physical Medicine and Rehabilitation, Department of Surgical Sciences, University of Turin, Via Zuretti 29, Turin, Italy ,“Città della Salute e della Scienza” University Hospital, Corso Bramante, 88, Turin, Italy
| | - Paolo Menè
- Division of Nephrology and Dialysis, Sant’Andrea University Hospital, “Sapienza” University of Rome, Via di Grottarossa, 1035/1039, Rome, Italy
| | - Vincenzo Savarino
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, Genoa, Italy
| | - Diego Fornasari
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Vanvitelli, 32, 20133, Milan, Italy.
| |
Collapse
|
44
|
Hoshijima H, Hunt M, Nagasaka H, Yaksh T. Systematic Review of Systemic and Neuraxial Effects of Acetaminophen in Preclinical Models of Nociceptive Processing. J Pain Res 2021; 14:3521-3552. [PMID: 34795520 PMCID: PMC8594782 DOI: 10.2147/jpr.s308028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/11/2021] [Indexed: 12/29/2022] Open
Abstract
Acetaminophen (APAP) in humans has robust effects with a high therapeutic index in altering postoperative and inflammatory pain states in clinical and experimental pain paradigms with no known abuse potential. This review considers the literature reflecting the preclinical actions of acetaminophen in a variety of pain models. Significant observations arising from this review are as follows: 1) acetaminophen has little effect upon acute nociceptive thresholds; 2) acetaminophen robustly reduces facilitated states as generated by mechanical and thermal hyperalgesic end points in mouse and rat models of carrageenan and complete Freund’s adjuvant evoked inflammation; 3) an antihyperalgesic effect is observed in models of facilitated processing with minimal inflammation (eg, phase II intraplantar formalin); and 4) potent anti-hyperpathic effects on the thermal hyperalgesia, mechanical and cold allodynia, allodynic thresholds in rat and mouse models of polyneuropathy and mononeuropathies and bone cancer pain. These results reflect a surprisingly robust drug effect upon a variety of facilitated states that clearly translate into a wide range of efficacy in preclinical models and to important end points in human therapy. The specific systems upon which acetaminophen may act based on targeted delivery suggest both a spinal and a supraspinal action. Review of current targets for this molecule excludes a role of cyclooxygenase inhibitor but includes effects that may be mediated through metabolites acting on the TRPV1 channel, or by effect upon cannabinoid and serotonin signaling. These findings suggest that the mode of action of acetaminophen, a drug with a long therapeutic history of utilization, has surprisingly robust effects on a variety of pain states in clinical patients and in preclinical models with a good therapeutic index, but in spite of its extensive use, its mechanisms of action are yet poorly understood.
Collapse
Affiliation(s)
- Hiroshi Hoshijima
- Department of Anesthesiology, Saitama Medical University Hospital, Saitama, Japan
| | - Matthew Hunt
- Departments of Anesthesiology and Pharmacology, University of California, San Diego Anesthesia Research Laboratory, La Jolla, CA, USA
| | - Hiroshi Nagasaka
- Department of Anesthesiology, Saitama Medical University Hospital, Saitama, Japan
| | - Tony Yaksh
- Departments of Anesthesiology and Pharmacology, University of California, San Diego Anesthesia Research Laboratory, La Jolla, CA, USA
| |
Collapse
|
45
|
The Natural Combination Medicine Traumeel (Tr14) Improves Resolution of Inflammation by Promoting the Biosynthesis of Specialized Pro-Resolving Mediators. Pharmaceuticals (Basel) 2021; 14:ph14111123. [PMID: 34832905 PMCID: PMC8623904 DOI: 10.3390/ph14111123] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 02/06/2023] Open
Abstract
The resolution of inflammation is an integral part of the acute inflammatory response and eventually leads to the return to homeostasis. It is supported by specialized pro-resolving mediators (SPMs) that act as immunoresolvents via specific G-protein-coupled receptors. In contrast to classical non-steroidal anti-inflammatory drugs (NSAIDs) that suppress the formation of pro-inflammatory lipid mediators such as prostaglandins, novel pharmacotherapeutic concepts propose to foster the biosynthesis of beneficial SPMs. Here, we demonstrate that the natural combination medicine Traumeel (Tr14) improves resolution of inflammation by promoting SPM formation. Tr14 enhanced the biosynthesis of 12-/15-lipoxygenase (LOX) products and of SPMs in zymosan-induced mouse peritonitis as well as in human monocyte-derived macrophages challenged with Staphylococcus aureus. Importantly, in the peritonitis model, Tr14 supported the recruitment of innate leukocytes and the efferocytotic capacity of macrophages, and positively influenced the inflammation resolution index. Taken together, we suggest that based on these properties Tr14 may possess therapeutic potential as an enhancer for the resolution of inflammatory processes.
Collapse
|
46
|
Chávez-Castillo M, Ortega Á, Cudris-Torres L, Duran P, Rojas M, Manzano A, Garrido B, Salazar J, Silva A, Rojas-Gomez DM, De Sanctis JB, Bermúdez V. Specialized Pro-Resolving Lipid Mediators: The Future of Chronic Pain Therapy? Int J Mol Sci 2021; 22:ijms221910370. [PMID: 34638711 PMCID: PMC8509014 DOI: 10.3390/ijms221910370] [Citation(s) in RCA: 9] [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: 08/31/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022] Open
Abstract
Chronic pain (CP) is a severe clinical entity with devastating physical and emotional consequences for patients, which can occur in a myriad of diseases. Often, conventional treatment approaches appear to be insufficient for its management. Moreover, considering the adverse effects of traditional analgesic treatments, specialized pro-resolving lipid mediators (SPMs) have emerged as a promising alternative for CP. These include various bioactive molecules such as resolvins, maresins, and protectins, derived from ω-3 polyunsaturated fatty acids (PUFAs); and lipoxins, produced from ω-6 PUFAs. Indeed, SPMs have been demonstrated to play a central role in the regulation and resolution of the inflammation associated with CP. Furthermore, these molecules can modulate neuroinflammation and thus inhibit central and peripheral sensitizations, as well as long-term potentiation, via immunomodulation and regulation of nociceptor activity and neuronal pathways. In this context, preclinical and clinical studies have evidenced that the use of SPMs is beneficial in CP-related disorders, including rheumatic diseases, migraine, neuropathies, and others. This review integrates current preclinical and clinical knowledge on the role of SPMs as a potential therapeutic tool for the management of patients with CP.
Collapse
Affiliation(s)
- Mervin Chávez-Castillo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Ángel Ortega
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Lorena Cudris-Torres
- Programa de Psicología, Fundación Universitaria del Área Andina sede Valledupar, Valledupar 200001, Colombia;
| | - Pablo Duran
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Milagros Rojas
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Alexander Manzano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Bermary Garrido
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Juan Salazar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Aljadis Silva
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela; (M.C.-C.); (Á.O.); (P.D.); (M.R.); (A.M.); (B.G.); (J.S.); (A.S.)
| | - Diana Marcela Rojas-Gomez
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Santiago 8370035, Chile;
| | - Juan B. De Sanctis
- Institute of Molecular and Translational Medicine, Palacký University Olomouc, 77900 Olomouc, Czech Republic;
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080002, Colombia
- Correspondence:
| |
Collapse
|
47
|
Silveira TL, Pang LY, Di Domenico A, Veloso ES, Silva ILD, Puerto HLD, Ferreria E, Argyle DJ. COX-2 Silencing in Canine Malignant Melanoma Inhibits Malignant Behaviour. Front Vet Sci 2021; 8:633170. [PMID: 34513965 PMCID: PMC8427276 DOI: 10.3389/fvets.2021.633170] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 07/27/2021] [Indexed: 12/22/2022] Open
Abstract
Metastatic melanoma is a very aggressive form of cancer in both humans and dogs. Dogs primarily develop oral melanoma of mucosal origin. Although oral melanoma in humans is rare, both diseases are highly aggressive with frequent metastases. This disease represents a “One Health” opportunity to improve molecular and mechanistic understanding of melanoma progression. Accumulating evidence suggests that cyclooxygenase-2 (COX-2) may play a critical role in the malignant behaviour of melanoma. In this study we analysed 85 histologically confirmed melanomas from canine patients and showed that COX-2 is overexpressed in both oral and cutaneous melanomas and that COX-2 expression correlates with established markers of poor prognosis. To determine the role of COX-2 in melanoma we developed two melanoma cell lines with stable integration of an inducible doxycycline-regulated expression vector containing a COX-2 targeted micro-RNA (miRNA). Using this system, we showed that cellular proliferation, migration and invasion are COX-2 dependent, establishing a direct relationship between COX-2 expression and malignant behaviour in canine melanoma. We have also developed a powerful molecular tool to aid further dissection of the mechanisms by which COX-2 regulates melanoma progression.
Collapse
Affiliation(s)
- Tatiany L Silveira
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Royal (Dick) School of Veterinary Studies, Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Lisa Y Pang
- Royal (Dick) School of Veterinary Studies, Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Alexandra Di Domenico
- Royal (Dick) School of Veterinary Studies, Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Emerson S Veloso
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Istéfani L D Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Helen L Del Puerto
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Enio Ferreria
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - David J Argyle
- Royal (Dick) School of Veterinary Studies, Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
48
|
Gao L, Yu Q, Zhang H, Wang Z, Zhang T, Xiang J, Yu S, Zhang S, Wu H, Xu Y, Wang Z, Shen L, Shu G, Chen YG, Liu H, Shen L, Li B. A resident stromal cell population actively restrains innate immune response in the propagation phase of colitis pathogenesis in mice. Sci Transl Med 2021; 13:13/603/eabb5071. [PMID: 34290057 DOI: 10.1126/scitranslmed.abb5071] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 12/09/2020] [Accepted: 05/10/2021] [Indexed: 12/22/2022]
Abstract
Inflammatory bowel disease (IBD) affects 0.3% of the global population, yet the etiology remains poorly understood. Anti-inflammation therapy has shown great success, but only 60% of patients with IBD benefit from it, indicating that new targets are needed. Here, we report the discovery of an intrinsic counter regulatory mechanism in colitis pathogenesis that may be targeted for IBD treatment. In response to microbial invasion, resident Vimentin+ stromal cells, connective tissue cells genetically marked by Twist2, are activated during the propagation phase of the disease, but not during initiation and resolution phases, and become a primary source of prostaglandin E2 (PGE2). PGE2 induction requires a nuclear factor κB-independent, TLR4-p38MAPK-Cox2 pathway activation. Ablation of each of the pathway genes, but not Rela or Tgfb1, in Twist2 cells enhanced M1 macrophage polarization and granulocyte/T helper 1 (TH1)/TH17 infiltration and aggravated colitis development. PGE2 administration ameliorated colitis in mouse models with defective PGE2 production but not in animals with normal PGE2 induction. Analysis of clinical samples and public domain data revealed increased expression of Cox2, the rate-limiting enzyme of PGE2 biosynthesis, in inflamed tissues, and especially in colon Vimentin+Twist2+ stromal cells, in about 60% of patients with active Crohn's disease or ulcerative colitis. Moreover, Cox2 protein expression was negatively correlated with disease severity, suggesting an involvement of stromal cells in IBD pathogenesis. Thus, the study uncovers an active immune pathway in colitic inflammation that may be targeted to treat patients with IBD with defects in PGE2 production.
Collapse
Affiliation(s)
- Liang Gao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qian Yu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Huasheng Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhengting Wang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tianyu Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jinnan Xiang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuxiang Yu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shaoyang Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hongguang Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yizhou Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhuo Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lu Shen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutritional Control, South China Agricultural University, Guangzhou 510642, China
| | - Ye-Guang Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Lei Shen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China. .,Institute of Traditional Chinese Medicine and Stem Cell Research, School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
49
|
Friedrich L, Cingolani G, Ko Y, Iaselli M, Miciaccia M, Perrone MG, Neukirch K, Bobinger V, Merk D, Hofstetter RK, Werz O, Koeberle A, Scilimati A, Schneider G. Learning from Nature: From a Marine Natural Product to Synthetic Cyclooxygenase-1 Inhibitors by Automated De Novo Design. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100832. [PMID: 34176236 PMCID: PMC8373093 DOI: 10.1002/advs.202100832] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/16/2021] [Indexed: 05/03/2023]
Abstract
The repertoire of natural products offers tremendous opportunities for chemical biology and drug discovery. Natural product-inspired synthetic molecules represent an ecologically and economically sustainable alternative to the direct utilization of natural products. De novo design with machine intelligence bridges the gap between the worlds of bioactive natural products and synthetic molecules. On employing the compound Marinopyrrole A from marine Streptomyces as a design template, the algorithm constructs innovative small molecules that can be synthesized in three steps, following the computationally suggested synthesis route. Computational activity prediction reveals cyclooxygenase (COX) as a putative target of both Marinopyrrole A and the de novo designs. The molecular designs are experimentally confirmed as selective COX-1 inhibitors with nanomolar potency. X-ray structure analysis reveals the binding of the most selective compound to COX-1. This molecular design approach provides a blueprint for natural product-inspired hit and lead identification for drug discovery with machine intelligence.
Collapse
Affiliation(s)
- Lukas Friedrich
- Department of Chemistry and Applied BiosciencesETH ZurichVladimir‐Prelog‐Weg 4Zurich8093Switzerland
| | - Gino Cingolani
- Department of Biochemistry and Molecular BiologySidney Kimmel Cancer CenterThomas Jefferson University1020 Locust StreetPhiladelphiaPA19107USA
| | - Ying‐Hui Ko
- Department of Biochemistry and Molecular BiologySidney Kimmel Cancer CenterThomas Jefferson University1020 Locust StreetPhiladelphiaPA19107USA
| | - Mariaclara Iaselli
- Department of Pharmacy – Pharmaceutical SciencesUniversity of BariVia E. Orabona 4Bari70125Italy
| | - Morena Miciaccia
- Department of Pharmacy – Pharmaceutical SciencesUniversity of BariVia E. Orabona 4Bari70125Italy
| | - Maria Grazia Perrone
- Department of Pharmacy – Pharmaceutical SciencesUniversity of BariVia E. Orabona 4Bari70125Italy
| | - Konstantin Neukirch
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruck6020Austria
| | - Veronika Bobinger
- Department of Chemistry and Applied BiosciencesETH ZurichVladimir‐Prelog‐Weg 4Zurich8093Switzerland
| | - Daniel Merk
- Department of Chemistry and Applied BiosciencesETH ZurichVladimir‐Prelog‐Weg 4Zurich8093Switzerland
- Institute of Pharmaceutical ChemistryGoethe‐UniversityMax‐von‐Laue Straße 9Frankfurt am Main60438Germany
| | - Robert Klaus Hofstetter
- Department of Pharmaceutical/Medicinal ChemistryFriedrich‐Schiller‐University JenaPhilosophenweg 14Jena07743Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal ChemistryFriedrich‐Schiller‐University JenaPhilosophenweg 14Jena07743Germany
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruck6020Austria
| | - Antonio Scilimati
- Department of Pharmacy – Pharmaceutical SciencesUniversity of BariVia E. Orabona 4Bari70125Italy
| | - Gisbert Schneider
- Department of Chemistry and Applied BiosciencesETH ZurichVladimir‐Prelog‐Weg 4Zurich8093Switzerland
- ETH Singapore SEC Ltd1 CREATE Way, #06‐01 CREATE TowerSingapore138602Singapore
| |
Collapse
|
50
|
Kuczyńska J, Nieradko-Iwanicka B. Future prospects of ketoprofen in improving the safety of the gastric mucosa. Biomed Pharmacother 2021; 139:111608. [DOI: 10.1016/j.biopha.2021.111608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/02/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
|