1
|
Zhang W, Shi X, Huang S, Yu Q, Wu Z, Xie W, Li B, Xu Y, Gao Z, Li G, Qian Q, He T, Zheng J, Zhang T, Tong Y, Deng D, Gao X, Tian H, Yao W. NitraTh epitope-based neoantigen vaccines for effective tumor immunotherapy. Cancer Immunol Immunother 2024; 73:245. [PMID: 39358493 PMCID: PMC11447171 DOI: 10.1007/s00262-024-03830-2] [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: 07/20/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
Neoantigen vaccines represent an emerging and promising strategy in the field of tumor immunotherapy. Despite their potential, designing an effective neoantigen vaccine remains a challenge due to the current limitations in predicting CD4+ T cell epitopes with high accuracy. Here, we introduce a novel approach to neoantigen vaccine design that does not rely on computational prediction of CD4+ T cell epitopes. Utilizing nitrated helper T cell epitope containing p-nitrophenylalanine, termed "NitraTh epitope," we have successfully engineered a series of tumor neoantigen vaccines capable of eliciting robust neoantigen-specific immune responses. With the help of NitraTh epitope, even mutations with low predicted affinity for MHC class I molecules were successfully induced to elicit neoantigen-specific responses. In H22 cell allograft and patient-derived xenograft (PDX) liver cancer mouse models, the NitraTh epitope-based neoantigen vaccines significantly suppressed tumor progression. More strikingly, through single-cell sequencing we found that the NitraTh epitope-based neoantigen vaccines regulate macrophage reprogramming and modulate macrophages to decrease the levels of the immunosuppressive molecule prostaglandin E2 (PGE2), which in turn reshapes the tumor immunosuppressive microenvironment. In summary, NitraTh epitope-based neoantigen vaccines possess the dual effects of potently activating neoantigen-specific immunity and alleviating immunosuppression, potentially providing a new paradigm for the design of tumor neoantigen vaccines.
Collapse
Grants
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- (No. 82073754, No.82273840, No.81973222) National Natural Science Foundation of China
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- 2020B03003 the Key R&D Program of Xinjiang Uygur Autonomous Region
- the Key R&D Program of Xinjiang Uygur Autonomous Region
Collapse
Affiliation(s)
- Wanli Zhang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Xupeiyao Shi
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Shitong Huang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Qiumin Yu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Zijie Wu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Wenbin Xie
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Binghua Li
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Yanchao Xu
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Zheng Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Guozhi Li
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Qianqian Qian
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Tiandi He
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Jiaxue Zheng
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Tingran Zhang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Yue Tong
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Danni Deng
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, 213003, Jiangsu, People's Republic of China
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Hong Tian
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Wenbing Yao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
2
|
Huang Q, Lai T, Wang Q, Luo L. mPGES-1 Inhibitor Discovery Based on Computer-Aided Screening: Pharmacophore Models, Molecular Docking, ADMET, and MD Simulations. Molecules 2023; 28:6059. [PMID: 37630311 PMCID: PMC10458489 DOI: 10.3390/molecules28166059] [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: 07/02/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
mPGES-1 is an enzyme, which, when activated by inflammatory factors, can cause prostaglandin E synthesis. Traditional non-steroidal anti-inflammatory drugs are capable of inhibiting prostaglandin production, yet they can also cause gastrointestinal reactions and coagulation disorders. mPGES-1, the enzyme at the conclusion of prostaglandin production, does not cause any adverse reactions when inhibited. Numerous studies have demonstrated that mPGES-1 is more abundant in cancerous cells than in healthy cells, indicating that decreasing the expression of mPGES-1 could be a potential therapeutic strategy for cancer. Consequently, the invention of mPGES-1 inhibitors presents a fresh avenue for the treatment of inflammation and cancer. Incorporating a database of TCM compounds, we collected a batch of compounds that had an inhibitory effect on mPGES-1 and possessed IC50 value. Firstly, a pharmacophore model was constructed, and the TCM database was screened, and the compounds with score cut-off values of more than 1 were retained. Then, the compounds retained after being screened via the pharmacodynamic model were screened for docking at the mPGES-1 binding site, followed by high-throughput virtual screening [HTVS] and standard precision [SP] and super-precision [XP] docking, and the compounds in the top 20% of the XP docking score were selected to calculate the total free binding energy of MM-GBSA. The best ten compounds were chosen by comparing their score against the reference ligand 4U9 and the MM-GBSA_dG_Bind score. ADMET analysis resulted in the selection of ten compounds, three of which had desirable medicinal properties. Finally, the binding energy of the target protein mPGES-1 and the candidate ligand compound was analyzed using a 100 ns molecular dynamics simulation of the reference ligand 4U9 and three selected compounds. After a gradual screening study and analysis, we identified a structure that is superior to the reference ligand 4U9 in all aspects, namely compound 15643. Taken together, the results of this study reveal a structure that can be used to inhibit mPGES-1 compound 15643, thereby providing a new option for anti-inflammatory and anti-tumor drugs.
Collapse
Affiliation(s)
- Qiqi Huang
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China; (Q.H.); (T.L.)
| | - Tianli Lai
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China; (Q.H.); (T.L.)
| | - Qu Wang
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China;
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China;
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
| |
Collapse
|
3
|
Ganesh T. Targeting EP2 Receptor for Drug Discovery: Strengths, Weaknesses, Opportunities, and Threats (SWOT) Analysis. J Med Chem 2023; 66:9313-9324. [PMID: 37458373 PMCID: PMC10388357 DOI: 10.1021/acs.jmedchem.3c00655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Indexed: 07/28/2023]
Abstract
Cyclooxygenase-1 and -2 (COX1 and COX2) derived endogenous ligand prostaglandin-E2 (PGE2) triggers several physiological and pathological conditions. It mediates signaling through four G-protein coupled receptors, EP1, EP2, EP3, and EP4. Among these, EP2 is expressed throughout the body including the brain and uterus. The functional role of EP2 has been extensively studied using EP2 gene knockout mice, cellular models, and selective small molecule agonists and antagonists for this receptor. The efficacy data from in vitro and in vivo animal models indicate that EP2 receptor is a major proinflammatory mediator with deleterious functions in a variety of diseases suggesting a path forward for EP2 inhibitors as the next generation of selective anti-inflammatory and antiproliferative agents. Interestingly in certain diseases, EP2 action is beneficial; therefore, EP2 agonists seem to be clinically useful. Here, we highlight the strengths, weaknesses, opportunities, and potential threats (SWOT analysis) for targeting EP2 receptor for therapeutic development for a variety of unmet clinical needs.
Collapse
Affiliation(s)
- Thota Ganesh
- Department of Pharmacology and Chemical
Biology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| |
Collapse
|
4
|
Sluter MN, Li Q, Yasmen N, Chen Y, Li L, Hou R, Yu Y, Yang CY, Meibohm B, Jiang J. The inducible prostaglandin E synthase (mPGES-1) in neuroinflammatory disorders. Exp Biol Med (Maywood) 2023; 248:811-819. [PMID: 37515545 PMCID: PMC10468642 DOI: 10.1177/15353702231179926] [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] [Indexed: 07/31/2023] Open
Abstract
The cyclooxygenase (COX)/prostaglandin E2 (PGE2) signaling pathway has emerged as a critical target for anti-inflammatory therapeutic development in neurological diseases. However, medical use of COX inhibitors in the treatment of various neurological disorders has been limited due to well-documented cardiovascular and cerebrovascular complications. It has been widely proposed that modulation of downstream microsomal prostaglandin E synthase-1 (mPGES-1) enzyme may provide more specificity for inhibiting PGE2-elicited neuroinflammation. Heightened levels of mPGES-1 have been detected in a variety of brain diseases such as epilepsy, stroke, glioma, and neurodegenerative diseases. Subsequently, elevated levels of PGE2, the enzymatic product of mPGES-1, have been demonstrated to modulate a multitude of deleterious effects. In epilepsy, PGE2 participates in retrograde signaling to augment glutamate release at the synapse leading to neuronal death. The excitotoxic demise of neurons incites the activation of microglia, which can become overactive upon further stimulation by PGE2. A selective mPGES-1 inhibitor was able to reduce gliosis and the expression of proinflammatory cytokines in the hippocampus following status epilepticus. A similar mechanism has also been observed in stroke, where the overactivation of microglia by PGE2 upregulated the expression and secretion of proinflammatory cytokines. This intense activation of neuroinflammatory processes triggered the secondary injury commonly observed in stroke, and blockade of mPGES-1 reduced infarction size and edema, suppressed induction of proinflammatory cytokines, and improved post-stroke well-being and cognition. Furthermore, elevated levels of PGE2 have been shown to intensify the proliferation of glioma cells, mediate P-glycoprotein expression at the blood-brain barrier (BBB) and facilitate breakdown of the BBB. For these reasons, targeting mPGES-1, the central and inducible enzyme of the COX cascade, may provide a more specific therapeutic strategy for treating neuroinflammatory diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Ruida Hou
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ying Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Chao-Yie Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
5
|
Sluter M, Bhuniya R, Yuan X, Ramaraju A, Chen Y, Yu Y, Parmar KR, Temrikar ZH, Srivastava A, Meibohm B, Jiang J, Yang CY. Novel, Brain-Permeable, Cross-Species Benzothiazole Inhibitors of Microsomal Prostaglandin E Synthase-1 (mPGES-1) Dampen Neuroinflammation In Vitro and In Vivo. ACS Pharmacol Transl Sci 2023; 6:587-599. [PMID: 37082746 PMCID: PMC10111624 DOI: 10.1021/acsptsci.2c00241] [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: 12/02/2022] [Indexed: 04/22/2023]
Abstract
Microsomal prostaglandin E synthase-1 (mPGES-1) is an inducible enzyme of the cyclooxygenase (COX) cascade that generates prostaglandin E2 (PGE2) during inflammatory conditions. PGE2 is known to be a potent immune signaling molecule that mediates both peripheral and central inflammations. Inhibition of mPGES-1, rather than COX, may overcome the cardiovascular side effects associated with long-term COX inhibition by providing a more specific strategy to target inflammation. However, mPGES-1 inhibitor development is hampered by the large differences in cross-species activity due to the structural differences between the human and murine mPGES-1. Here, we report that our thiazole-based mPGES-1 inhibitors, compounds 11 (UT-11) and 19 derived from two novel scaffolds, were able to suppress PGE2 production in human (SK-N-AS) and murine (BV2) cells. The IC50 values of inhibiting PGE2 production in human and murine cells were 0.10 and 2.00 μM for UT-11 and 0.43 and 1.55 μM for compound 19, respectively. Based on in vitro and in vivo pharmacokinetic data, we selected UT-11 for evaluation in a lipopolysaccharide (LPS)-induced inflammation model. We found that our compound significantly suppressed proinflammatory cytokines and chemokines in the hippocampus but not in the kidney. Taken together, we demonstrated the potential of UT-11 in treating neuroinflammatory conditions, including epilepsy and stroke, and warrant further optimization.
Collapse
Affiliation(s)
- Madison
N. Sluter
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- College
of Graduate Health Sciences, University
of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Rajib Bhuniya
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Xinrui Yuan
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Andhavaram Ramaraju
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Yu Chen
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Ying Yu
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Keyur R. Parmar
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Zaid H. Temrikar
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Ashish Srivastava
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Bernd Meibohm
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Jianxiong Jiang
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Chao-Yie Yang
- Departments
of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
6
|
Potenza M, Giordano A, Chini MG, Saviano A, Kretzer C, Raucci F, Russo M, Lauro G, Terracciano S, Bruno I, Iorizzi M, Hofstetter RK, Pace S, Maione F, Werz O, Bifulco G. Identification of 2-Aminoacyl-1,3,4-thiadiazoles as Prostaglandin E 2 and Leukotriene Biosynthesis Inhibitors. ACS Med Chem Lett 2022; 14:26-34. [PMID: 36655121 PMCID: PMC9841589 DOI: 10.1021/acsmedchemlett.2c00343] [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: 07/25/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
The application of a multi-step scientific workflow revealed an unprecedented class of PGE2/leukotriene biosynthesis inhibitors with in vivo activity. Specifically, starting from a combinatorial virtual library of ∼4.2 × 105 molecules, a small set of compounds was identified for the synthesis. Among these, four novel 2-aminoacyl-1,3,4-thiadiazole derivatives (3, 6, 7, and 9) displayed marked anti-inflammatory properties in vitro by strongly inhibiting PGE2 biosynthesis, with IC50 values in the nanomolar range. The hit compounds also efficiently interfered with leukotriene biosynthesis in cell-based systems and modulated IL-6 and PGE2 biosynthesis in a lipopolysaccharide-stimulated J774A.1 macrophage cell line. The most promising compound 3 showed prominent in vivo anti-inflammatory activity in a mouse model, with efficacy comparable to that of dexamethasone, attenuating zymosan-induced leukocyte migration in mouse peritoneum with considerable modulation of the levels of typical pro-/anti-inflammatory cytokines.
Collapse
Affiliation(s)
- Marianna Potenza
- Department
of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy,The
FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Assunta Giordano
- Department
of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy,Institute
of Biomolecular Chemistry (ICB), Consiglio
Nazionale delle Ricerche (CNR), Via Campi Flegrei 34, Pozzuoli, 80078 Napoli, Italy
| | - Maria G. Chini
- Department
of Biosciences and Territory, University
of Molise, Contrada Fonte Lappone, Pesche, 86090 Isernia, Italy
| | - Anella Saviano
- ImmunoPharmaLab,
Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Christian Kretzer
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743 Jena, Germany
| | - Federica Raucci
- ImmunoPharmaLab,
Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Marina Russo
- ImmunoPharmaLab,
Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Gianluigi Lauro
- Department
of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Stefania Terracciano
- Department
of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Ines Bruno
- Department
of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy
| | - Maria Iorizzi
- Department
of Biosciences and Territory, University
of Molise, Contrada Fonte Lappone, Pesche, 86090 Isernia, Italy
| | - Robert K. Hofstetter
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743 Jena, Germany
| | - Simona Pace
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743 Jena, Germany
| | - Francesco Maione
- ImmunoPharmaLab,
Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy
| | - Oliver Werz
- Department
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, 07743 Jena, Germany,
| | - Giuseppe Bifulco
- Department
of Pharmacy, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, Italy,
| |
Collapse
|
7
|
Mikhail DS, El-Nassan HB, Mahmoud ST, Fahim SH. Nonacidic thiophene-based derivatives as potential analgesic and design, synthesis, biological evaluation, and metabolic stability study. Drug Dev Res 2022; 83:1739-1757. [PMID: 36074734 DOI: 10.1002/ddr.21992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/23/2022] [Accepted: 08/02/2022] [Indexed: 12/29/2022]
Abstract
Nonsteroidal anti-inflammatory drugs represent one of the most popularly used classes of drugs. However, their long-term administration is associated with various side effects including gastrointestinal ulceration. One of the major reasons of NSAIDs ulcerogenicity is direct damage of the epithelial lining cells by the acidic moieties present in many drugs. Another drawback for this acidic group is its rapid metabolism and clearance through Phase II conjugation. Three series of thiophene and thienopyrimidine derivatives were designed and synthesized as nonacidic anti-inflammatory agents. In vivo testing of their analgesic activity indicated that compounds 2b and 7a-d showed higher PI values than that of the positive control drugs, indomethacin and celecoxib. The latter compounds 2b and 7a-d were subjected to further anti-inflammatory activity testing where they showed comparable percentage edema inhibition to that of indomethacin and celecoxib. Compounds 2b, 7a, 7c, and 7d inhibited PGE2 synthesis by 61.10%-74.54% (71.47% for indomethacin, and 80.11% for celecoxib). The same compounds inhibited the expression of rat mPGES-1 and cPGES3 by 74%-83% (77% for indomethacin, and 82% for celecoxib) and 48%-70% (62% for indomethacin, and 70% for celecoxib), respectively. The stability of the most active compound 2b in Nonenzymatic gastrointestinal fluids and in human plasma was tested. Additionally, studying the metabolic stability of compound 2b in S9 rat liver fraction showed that it displayed a slow in vitro clearance with half-life time 1.5-fold longer than indomethacin. The metabolites of 2b were predicted via UPLC-MS/MS. In silico ADMET profiling study was also included.
Collapse
Affiliation(s)
- Demiana S Mikhail
- Department, of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hala B El-Nassan
- Department, of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Sally T Mahmoud
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Samar H Fahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
8
|
Santos Nascimento IJD, de Aquino TM, da Silva Júnior EF. Computer-Aided Drug Design of Anti-inflammatory Agents Targeting Microsomal Prostaglandin E2 Synthase-1 (mPGES-1). Curr Med Chem 2022; 29:5397-5419. [PMID: 35301943 DOI: 10.2174/0929867329666220317122948] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 11/22/2022]
Abstract
Inflammation is a natural process in response to external stimuli associated with organism protection. However, this reaction could be exaggerated, leading to severe damages related to physiopathological processes, such as rheumatoid arthritis, cancer, diabetes, allergies, infections, among others. Inflammation is mainly characterized by pain, increased temperature, flushing, and edema, which can be controlled using anti-inflammatory drugs. In this context, prostaglandin E2 (PGE2) inhibition has been targeted for designing new compounds with anti-inflammatory properties. It is a bioactive lipid overproduced during an inflammatory process, in which its increased production is carried out mainly by COX-1, COX-2, and microsomal prostaglandin E2 synthase-1 (mPGES-1). Recently, studies have demonstrated that mPGES-1 inhibition is a safe strategy to develop anti-inflammatory agents, which could protect against pain, acute inflammation, arthritis, autoimmune diseases, and different types of cancers. To decrease production costs and increase the probability of discovering active substances, computer-aided drug design (CADD) approaches have been increasingly used for designing new inhibitors. Thus, this review will cover all aspects involving high-throughput virtual screening, molecular docking, dynamics, fragment-based drug design, quantitative structure-activity relationship in seeking new promising mPGES-1 inhibitors.
Collapse
Affiliation(s)
- Igor José Dos Santos Nascimento
- Laboratory of Synthesis and Research in Medicinal Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Brazil.
- Department of Pharmacy, Estácio of Alagoas College, Maceió, Brazil
| | - Thiago Mendonça de Aquino
- Laboratory of Synthesis and Research in Medicinal Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Brazil.
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Brazil
| | - Edeildo Ferreira da Silva Júnior
- Laboratory of Synthesis and Research in Medicinal Chemistry, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Brazil.
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Brazil
| |
Collapse
|
9
|
N-Acylated and N-Alkylated 2-Aminobenzothiazoles Are Novel Agents That Suppress the Generation of Prostaglandin E2. Biomolecules 2022; 12:biom12020267. [PMID: 35204768 PMCID: PMC8961538 DOI: 10.3390/biom12020267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 11/23/2022] Open
Abstract
The quest for novel agents to regulate the generation of prostaglandin E2 (PGE2) is of high importance because this eicosanoid is a key player in inflammatory diseases. We synthesized a series of N-acylated and N-alkylated 2-aminobenzothiazoles and related heterocycles (benzoxazoles and benzimidazoles) and evaluated their ability to suppress the cytokine-stimulated generation of PGE2 in rat mesangial cells. 2-Aminobenzothiazoles, either acylated by the 3-(naphthalen-2-yl)propanoyl moiety (GK510) or N-alkylated by a chain carrying a naphthalene (GK543) or a phenyl moiety (GK562) at a distance of three carbon atoms, stand out in inhibiting PGE2 generation, with EC50 values ranging from 118 nM to 177 nM. Both GK510 and GK543 exhibit in vivo anti-inflammatory activity greater than that of indomethacin. Thus, N-acylated or N-alkylated 2-aminobenzothiazoles are novel leads for the regulation of PGE2 formation.
Collapse
|
10
|
Ergül AG, Maz TG, Kretzer C, Olğaç A, Jordan PM, Çalışkan B, Werz O, Banoglu E. Novel potent benzimidazole-based microsomal prostaglandin E2 Synthase-1 (mPGES-1) inhibitors derived from BRP-201 that also inhibit leukotriene C4 synthase. Eur J Med Chem 2022; 231:114167. [DOI: 10.1016/j.ejmech.2022.114167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/22/2022] [Accepted: 01/29/2022] [Indexed: 01/27/2023]
|
11
|
Nur S, Hossain MM, Islam N, Tareq AM, Hanif NB, Khatun R, Sayeed MA. Scrutinizing pharmacological efficiency for Acacia auriculiformis by experimental and computational approach. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The study sought to investigate the biological efficacy of methanol leave extract of Acacia auriculiformis (MEAA) via in vitro, in vivo, in silico approaches. The in vitro cytotoxicity was evaluated through brine shrimp lethality assay, and anti-inflammatory activity was determined by membrane stabilisation and protein denaturation methods (BSA and egg albumin). The in vivo antipyretic activity was examined via Brewer’s yeast induced pyrexia model.
Results
A. auriculiformis extract unveiled moderate cytotoxicity with significant anti-inflammatory efficacy (p < 0.001) compared to standard drug. This extract also exhibited dose-dependent time of paralysis and death for the worm (p < 0.001) in the anthelmintic test which was directly proportional to employed concentrations. A notable percentage of clot lysis effect (36.42 ± 1.95%, p < 0.001) was also observed for MEAA in human blood compared to control. However, this extract significantly (p < 0.05) reduced fever in a dose-dependent manner during the antipyretic experiment. Besides, in computer-aided investigations, two compounds (2,4-ditert-butylphenol and 3-hydroxy-β-damascone) revealed the best binding interaction with six proteins for cytotoxicity, inflammation, helminthic, thrombolytic and pyretic effect. Moreover, these two compounds satisfy Lipinski’s ‘Rule of Five’ and revealed drug-likeness profiles in the toxicological study.
Conclusions
These findings disclosed that methanol leaves extract of A. auriculiformis might be a potent source for anti-inflammatory, anti-helminthic, thrombolytic and antipyretic agents.
Collapse
|
12
|
Di Micco S, Terracciano S, Ruggiero D, Potenza M, Vaccaro MC, Fischer K, Werz O, Bruno I, Bifulco G. Identification of 2-(thiophen-2-yl)acetic Acid-Based Lead Compound for mPGES-1 Inhibition. Front Chem 2021; 9:676631. [PMID: 34046398 PMCID: PMC8144515 DOI: 10.3389/fchem.2021.676631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/07/2021] [Indexed: 11/13/2022] Open
Abstract
We report the implementation of our in silico/synthesis pipeline by targeting the glutathione-dependent enzyme mPGES-1, a valuable macromolecular target in both cancer therapy and inflammation therapy. Specifically, by using a virtual fragment screening approach of aromatic bromides, straightforwardly modifiable by the Suzuki-Miyaura reaction, we identified 3-phenylpropanoic acid and 2-(thiophen-2-yl)acetic acid to be suitable chemical platforms to develop tighter mPGES-1 inhibitors. Among these, compounds 1c and 2c showed selective inhibitory activity against mPGES-1 in the low micromolar range in accordance with molecular modeling calculations. Moreover, 1c and 2c exhibited interesting IC50 values on A549 cell lines compared to CAY10526, selected as reference compound. The most promising compound 2c induced the cycle arrest in the G0/G1 phase at 24 h of exposure, whereas at 48 and 72 h, it caused an increase of subG0/G1 fraction, suggesting an apoptosis/necrosis effect.
Collapse
Affiliation(s)
- Simone Di Micco
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| | | | - Dafne Ruggiero
- Dipartimento di Farmacia, University degli Studi di Salerno, Fisciano, Italy
| | - Marianna Potenza
- Dipartimento di Farmacia, University degli Studi di Salerno, Fisciano, Italy
| | - Maria C Vaccaro
- Dipartimento di Farmacia, University degli Studi di Salerno, Fisciano, Italy
| | - Katrin Fischer
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Oliver Werz
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Jena, Germany
| | - Ines Bruno
- Dipartimento di Farmacia, University degli Studi di Salerno, Fisciano, Italy
| | - Giuseppe Bifulco
- Dipartimento di Farmacia, University degli Studi di Salerno, Fisciano, Italy
| |
Collapse
|
13
|
Psarra A, Theodoropoulou MA, Erhardt M, Mertiri M, Mantzourani C, Vasilakaki S, Magrioti V, Huwiler A, Kokotos G. α-Ketoheterocycles Able to Inhibit the Generation of Prostaglandin E 2 (PGE 2) in Rat Mesangial Cells. Biomolecules 2021; 11:biom11020275. [PMID: 33668480 PMCID: PMC7918003 DOI: 10.3390/biom11020275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/28/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
Prostaglandin E2 (PGE2) is a key mediator of inflammation, and consequently huge efforts have been devoted to the development of novel agents able to regulate its formation. In this work, we present the synthesis of various α-ketoheterocycles and a study of their ability to inhibit the formation of PGE2 at a cellular level. A series of α-ketobenzothiazoles, α-ketobenzoxazoles, α-ketobenzimidazoles, and α-keto-1,2,4-oxadiazoles were synthesized and chemically characterized. Evaluation of their ability to suppress the generation of PGE2 in interleukin-1β plus forskolin-stimulated mesangial cells led to the identification of one α-ketobenzothiazole (GK181) and one α-ketobenzoxazole (GK491), which are able to suppress the PGE2 generation at a nanomolar level.
Collapse
Affiliation(s)
- Anastasia Psarra
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (A.P.); (M.A.T.); (M.M.); (C.M.); (S.V.); (V.M.)
| | - Maria A. Theodoropoulou
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (A.P.); (M.A.T.); (M.M.); (C.M.); (S.V.); (V.M.)
| | - Martin Erhardt
- Institute of Pharmacology, University of Bern, CH-3010 Bern, Switzerland; (M.E.); (A.H.)
| | - Marina Mertiri
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (A.P.); (M.A.T.); (M.M.); (C.M.); (S.V.); (V.M.)
| | - Christiana Mantzourani
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (A.P.); (M.A.T.); (M.M.); (C.M.); (S.V.); (V.M.)
| | - Sofia Vasilakaki
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (A.P.); (M.A.T.); (M.M.); (C.M.); (S.V.); (V.M.)
| | - Victoria Magrioti
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (A.P.); (M.A.T.); (M.M.); (C.M.); (S.V.); (V.M.)
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, CH-3010 Bern, Switzerland; (M.E.); (A.H.)
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Athens, Greece; (A.P.); (M.A.T.); (M.M.); (C.M.); (S.V.); (V.M.)
- Correspondence: ; Tel.: +30-210-727-4462
| |
Collapse
|
14
|
Mechanism of action and potential applications of selective inhibition of microsomal prostaglandin E synthase-1-mediated PGE 2 biosynthesis by sonlicromanol's metabolite KH176m. Sci Rep 2021; 11:880. [PMID: 33441600 PMCID: PMC7806836 DOI: 10.1038/s41598-020-79466-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/08/2020] [Indexed: 01/29/2023] Open
Abstract
Increased prostaglandin E2 (PGE2) levels were detected in mitochondrial disease patient cells harboring nuclear gene mutations in structural subunits of complex I, using a metabolomics screening approach. The increased levels of this principal inflammation mediator normalized following exposure of KH176m, an active redox-modulator metabolite of sonlicromanol (KH176). We next demonstrated that KH176m selectively inhibited lipopolysaccharide (LPS) or interleukin-1β (IL-1β)-induced PGE2 production in control skin fibroblasts. Comparable results were obtained in the mouse macrophage-like cell line RAW264.7. KH176m selectively inhibited mPGES-1 activity, as well as the inflammation-induced expression of mPGES-1. Finally, we showed that the effect of KH176m on mPGES-1 expression is due to the inhibition of a PGE2-driven positive feedback control-loop of mPGES-1 transcriptional regulation. Based on the results obtained we discuss potential new therapeutic applications of KH176m and its clinical stage parent drug candidate sonlicromanol in mitochondrial disease and beyond.
Collapse
|
15
|
Dos Santos LD, Froes TQ, Contin de Melo MC, Petto de Souza GE, Soares DDM, Castilho MS. Triazol-phenyl antipyretic derivatives inhibit mPGES-1 mRNA levels in LPS-Induced RAW 264.7 macrophage cells. Antiinflamm Antiallergy Agents Med Chem 2020; 20:271-281. [PMID: 33292158 DOI: 10.2174/1871523019999201208202831] [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: 07/13/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Microsomal prostaglandin E synthase-1 (mPGES-1) catalyzes the terminal step of prostaglandin E2 (PGE2) production, which plays an important role in the regulation of febrile response. In our previous work, ligand-based pharmacophore models, built with mPGES-1 inhibitors, were employed to identify a novel series of compounds that reduce the febrile response in rats. OBJECTIVES Evaluate the mechanism of action of the most active compound (1). METHODS For in vivo assays, rats were pretreated with the antipyretic compounds 1-8, 30 min before LPS injection. For in vitro assays, RAW 264.7 macrophage cells were incubated with the antipyretic compounds 1-8 for 1 hour before LPS stimu-lus. After 16 h, quantitative real-time PCR was carried out. Additionally, the PGE2 concentration in hypothalamus was quantified by ELISA and the inhibitory effect of N-cyclopentyl-N'-[3-(3-cyclopropyl-1H-1,2,4-triazol-5-yl)phenyl]ethanediamide (1) over human COX-2 enzymatic activity was determined with a COX Colorimetric Inhibitor Screening Assay Kit. RESULTS Compound 1 and CAY10526 have comparable efficacy to reduce the febrile response when injected i.v. (com-pound 1: 63.10%, CAY10526: 70.20%). Moreover, compound 1 significantly reduces the mPGES-1 mRNA levels, in RAW264.7 cells, under inflammatory conditions. A chemically-similar compound (8- ) also significantly reduces the mRNA levels of the gene target. On the other hand, compounds 6 and 7, which are also somewhat similar to compound 1, do not, significantly, impact mPGES-1 mRNA levels. CONCLUSIONS PGE2 concentration reduction in hypothalamus, due to compound 1 central injection, is related to decreased mPGES-1 mRNA levels but not to COX-2 inhibition (IC50> 50 μM). Therefore, compound 1 is a promising lead for inno-vative antipyretic drug development.
Collapse
Affiliation(s)
- Lenisa Dandara Dos Santos
- Laboratory of Pharmacology of inflammation and fever, Faculty of Pharmacy, Federal University of Bahia, Av. Barão de Jeremoabo s/n, Salvador, BA,. Brazil
| | - Thamires Quadros Froes
- Laboratory of Pharmacology of inflammation and fever, Faculty of Pharmacy, Federal University of Bahia, Av. Barão de Jeremoabo s/n, Salvador, BA,. Brazil
| | - Miriam Cristina Contin de Melo
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n, 14040-903, Ribeirão Preto, SP,. Brazil
| | - Gloria Emília Petto de Souza
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n, 14040-903, Ribeirão Preto, SP,. Brazil
| | - Denis de Melo Soares
- Laboratory of Pharmacology of inflammation and fever, Faculty of Pharmacy, Federal University of Bahia, Av. Barão de Jeremoabo s/n, Salvador, BA,. Brazil
| | - Marcelo Santos Castilho
- Laboratory of Bioinformatics and Molecular Modeling, Faculty of Pharmacy, Federal University of Bahia, Av. Barão de Jeremoabo s/n, Salvador, BA,. Brazil
| |
Collapse
|
16
|
Tuure L, Pemmari A, Hämäläinen M, Moilanen T, Moilanen E. Regulation of gene expression by MF63, a selective inhibitor of microsomal PGE synthase 1 (mPGES1) in human osteoarthritic chondrocytes. Br J Pharmacol 2020; 177:4134-4146. [PMID: 32449517 PMCID: PMC7443472 DOI: 10.1111/bph.15142] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 01/15/2020] [Accepted: 03/17/2020] [Indexed: 12/18/2022] Open
Abstract
Background and Purpose mPGES1 catalyses the production of PGE2, the most abundant prostanoid related to inflammation and pain in arthritis. mPGES1 is suggested to be a safer and more selective drug target in inflammatory conditions compared to the COX enzymes inhibited by NSAIDs. In the present study, we investigated the effects of the selective mPGES1 inhibitor MF63 on gene expression in primary human chondrocytes from patients with osteoarthritis (OA). Experimental Approach Chondrocytes were isolated from articular cartilage obtained from osteoarthritis patients undergoing knee replacement surgery. The effects of MF63 were studied in the primary chondrocytes with RNA‐sequencing based genome‐wide expression analysis. The main results were confirmed with qRT‐PCR and compared with the effects of the NSAID ibuprofen. Functional analysis was performed with the GO database and interactions between the genes were studied with STRING. Key Results MF63 enhanced the expression of multiple metallothionein 1 (MT1) isoforms as well as endogenous antagonists of IL‐1 and IL‐36. The expression of IL‐6, by contrast, was down‐regulated. These genes were also essential in functional and interaction network analyses. The effects of MF63 were consistent in qRT‐PCR analysis, whereas the effects of ibuprofen overlapped only partly with MF63. There were no evident findings of catabolic effects by MF63. Conclusion and Implications Metallothionein 1 has been suggested to have anti‐inflammatory and protective effects in cartilage. Up‐regulation of the antagonists of IL‐1 superfamily and down‐regulation of the pro‐inflammatory cytokine IL‐6 also support novel anti‐inflammatory and possibly disease‐modifying effects of mPGES1 inhibitors in arthritis.
Collapse
Affiliation(s)
- Lauri Tuure
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Antti Pemmari
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Teemu Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland.,Coxa Hospital for Joint Replacement, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| |
Collapse
|
17
|
Thulasingam M, Haeggström JZ. Integral Membrane Enzymes in Eicosanoid Metabolism: Structures, Mechanisms and Inhibitor Design. J Mol Biol 2020; 432:4999-5022. [PMID: 32745470 DOI: 10.1016/j.jmb.2020.07.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/14/2022]
Abstract
Eicosanoids are potent lipid mediators involved in central physiological processes such as hemostasis, renal function and parturition. When formed in excess, eicosanoids become critical players in a range of pathological conditions, in particular pain, fever, arthritis, asthma, cardiovascular disease and cancer. Eicosanoids are generated via oxidative metabolism of arachidonic acid along the cyclooxygenase (COX) and lipoxygenase (LOX) pathways. Specific lipid species are formed downstream of COX and LOX by specialized synthases, some of which reside on the nuclear and endoplasmic reticulum, including mPGES-1, FLAP, LTC4 synthase, and MGST2. These integral membrane proteins are members of the family "membrane-associated proteins in eicosanoid and glutathione metabolism" (MAPEG). Here we focus on this enzyme family, which encompasses six human members typically catalyzing glutathione dependent transformations of lipophilic substrates. Enzymes of this family have evolved to combat the topographical challenge and unfavorable energetics of bringing together two chemically different substrates, from cytosol and lipid bilayer, for catalysis within a membrane environment. Thus, structural understanding of these enzymes are of utmost importance to unravel their molecular mechanisms, mode of substrate entry and product release, in order to facilitate novel drug design against severe human diseases.
Collapse
Affiliation(s)
- Madhuranayaki Thulasingam
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| | - Jesper Z Haeggström
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
18
|
Park JC, Hagiwara A, Park HG, Lee JS. The glutathione S-transferase genes in marine rotifers and copepods: Identification of GSTs and applications for ecotoxicological studies. MARINE POLLUTION BULLETIN 2020; 156:111080. [PMID: 32510351 DOI: 10.1016/j.marpolbul.2020.111080] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 06/11/2023]
Abstract
Various xenobiotics are constantly being released and accumulated into the aquatic environments and consequently, the aquatic organisms are continuously being exposed to exogenous stressors. Among various xenobiotic detoxifying enzymes, Glutathione S-transferase (GST) is one of the major xenobiotic detoxifying enzyme which is widely distributed among living organisms and thus, understanding of the nature of GSTs is crucial. Previous studies have shown GST activity in response to various xenobiotics yet, full identification of GSTs in marine invertebrates is still limited. This review covers information on the importance of GSTs as a biomarker for emerging chemicals and their response to wide ranges of environmental pollutants as well as in-depth phylogenetic analysis of marine invertebrates, including recently identified GSTs belonging to rotifers (Brachionus spp.) and copepods (Tigriopus japonicus and Paracyclopina nana), with unique class-specific features of GSTs, as well as a new suggestion of GST evolutionary pathway.
Collapse
Affiliation(s)
- Jun Chul Park
- Department of Biological Science, College of Science, Sungkyunkwan University (SKKU), Suwon 16419, South Korea
| | - Atsushi Hagiwara
- Graduate School of Fisheries and Environmental Sciences, Nagasaki University, Nagasaki 852-8521, Japan; Organization for Marine Science and Technology, Nagasaki University, Nagasaki 852-8521, Japan
| | - Heum Gi Park
- Department of Marine Resource Development, College of Life Sciences, Gangneung-Wonju National University, Gangneung 25457, South Korea
| | - Jae-Seong Lee
- Department of Biological Science, College of Science, Sungkyunkwan University (SKKU), Suwon 16419, South Korea.
| |
Collapse
|
19
|
Di Francesco L, Bruno A, Ricciotti E, Tacconelli S, Dovizio M, Guillem-Llobat P, Alisi MA, Garrone B, Coletta I, Mangano G, Milanese C, FitzGerald GA, Patrignani P. Pharmacological Characterization of the Microsomal Prostaglandin E 2 Synthase-1 Inhibitor AF3485 In Vitro and In Vivo. Front Pharmacol 2020; 11:374. [PMID: 32317963 PMCID: PMC7147323 DOI: 10.3389/fphar.2020.00374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/12/2020] [Indexed: 12/14/2022] Open
Abstract
Rationale The development of inhibitors of microsomal prostaglandin (PG)E2 synthase-1 (mPGES-1) was driven by the promise of attaining antiinflammatory agents with a safe cardiovascular profile because of the possible diversion of the accumulated substrate, PGH2, towards prostacyclin (PGI2). Objectives We studied the effect of the human mPGES-1 inhibitor, AF3485 (a benzamide derivative) on prostanoid biosynthesis in human whole blood in vitro. To characterize possible off-target effects of the compound, we evaluated: i)the impact of its administration on the systemic biosynthesis of prostanoids in a model of complete Freund's adjuvant (CFA)-induced monoarthritis in rats; ii) the effects on cyclooxygenase (COX)-2 expression and the biosynthesis of prostanoids in human monocytes and human umbilical vein endothelial cells (HUVECs) in vitro. Methods Prostanoids were assessed in different cellular models by immunoassays. The effect of the administration of AF3485 (30 and 100 mg/kg,i.p.) or celecoxib (20mg/kg, i.p.), for 3 days, on the urinary levels of enzymatic metabolites of prostanoids, PGE-M, PGI-M, and TX-M were assessed by LC-MS. Results In LPS-stimulated whole blood, AF3485 inhibited PGE2 biosynthesis, in a concentration-dependent fashion. At 100μM, PGE2 levels were reduced by 66.06 ± 3.30%, associated with a lower extent of TXB2 inhibition (40.56 ± 5.77%). AF3485 administration to CFA-treated rats significantly reduced PGE-M (P < 0.01) and TX-M (P < 0.05) similar to the selective COX-2 inhibitor, celecoxib. In contrast, AF3485 induced a significant (P < 0.05) increase of urinary PGI-M while it was reduced by celecoxib. In LPS-stimulated human monocytes, AF3485 inhibited PGE2 biosynthesis with an IC50 value of 3.03 µM (95% CI:0.5–8.75). At 1μM, AF3485 enhanced TXB2 while at higher concentrations, the drug caused a concentration-dependent inhibition of TXB2. At 100 μM, maximal inhibition of the two prostanoids was associated with the downregulation of COX-2 protein by 86%. These effects did not involve AMPK pathway activation, IkB stabilization, or PPARγ activation. In HUVEC, AF3485 at 100 μM caused a significant (P < 0.05) induction of COX-2 protein associated with enhanced PGI2 production. These effects were reversed by the PPARγ antagonist GW9662. Conclusions The inhibitor of human mPGES-1 AF3485 is a novel antiinflammatory compound which can also modulate COX-2 induction by inflammatory stimuli. The compound also induces endothelial COX-2-dependent PGI2 production via PPARγ activation, both in vitro and in vivo, which might translate into a protective effect for the cardiovascular system.
Collapse
Affiliation(s)
- Luigia Di Francesco
- Department of Neuroscience, Imaging and Clinical Sciences, and Center for Advanced Studies and Technology (CAST), School of Medicine, G. d'Annunzio University, Chieti, Italy
| | - Annalisa Bruno
- Department of Neuroscience, Imaging and Clinical Sciences, and Center for Advanced Studies and Technology (CAST), School of Medicine, G. d'Annunzio University, Chieti, Italy
| | - Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| | - Stefania Tacconelli
- Department of Neuroscience, Imaging and Clinical Sciences, and Center for Advanced Studies and Technology (CAST), School of Medicine, G. d'Annunzio University, Chieti, Italy
| | - Melania Dovizio
- Department of Neuroscience, Imaging and Clinical Sciences, and Center for Advanced Studies and Technology (CAST), School of Medicine, G. d'Annunzio University, Chieti, Italy
| | - Paloma Guillem-Llobat
- Department of Neuroscience, Imaging and Clinical Sciences, and Center for Advanced Studies and Technology (CAST), School of Medicine, G. d'Annunzio University, Chieti, Italy
| | | | | | | | | | | | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
| | - Paola Patrignani
- Department of Neuroscience, Imaging and Clinical Sciences, and Center for Advanced Studies and Technology (CAST), School of Medicine, G. d'Annunzio University, Chieti, Italy
| |
Collapse
|
20
|
|
21
|
Abstract
Prostaglandins (PGs) are highly bioactive fatty acids. PGs, especially prostaglandin E2 (PGE2), are abundantly produced by cells of both the bone-forming (osteoblast) lineage and the bone-resorbing (osteoclast) lineage. The inducible cyclooxygenase, COX-2, is largely responsible for most PGE2 production in bone, and once released, PGE2 is rapidly degraded in vivo. COX-2 is induced by multiple agonists - hormones, growth factors, and proinflammatory factors - and the resulting PGE2 may mediate, amplify, or, as we have recently shown for parathyroid hormone (PTH), inhibit responses to these agonists. In vitro, PGE2 can directly stimulate osteoblast differentiation and, indirectly via stimulation of RANKL in osteoblastic cells, stimulate the differentiation of osteoclasts. The net balance of these two effects of PGE2 in vivo on bone formation and bone resorption has been hard to predict and, as expected for such a widespread local factor, hard to study. Some of the complexity of PGE2 actions on bone can be explained by the fact that there are four receptors for PGE2 (EP1-4). Some of the major actions of PGE2 in vitro occur via EP2 and EP4, both of which can stimulate cAMP signaling, but there are other distinct signaling pathways, important in other tissues, which have not yet been fully elucidated in bone cells. Giving PGE2 or agonists of EP2 and EP4 to accelerate bone repair has been examined with positive results. Further studies to clarify the pathways of PGE2 action in bone may allow us to identify new and more effective ways to deliver the therapeutic benefits of PGE2 in skeletal disorders.
Collapse
Affiliation(s)
- Carol Pilbeam
- Department of Medicine and Musculoskeletal Institute, UConn Health, Farmington, CT, USA.
| |
Collapse
|
22
|
A review on mPGES-1 inhibitors: From preclinical studies to clinical applications. Prostaglandins Other Lipid Mediat 2019; 147:106383. [PMID: 31698145 DOI: 10.1016/j.prostaglandins.2019.106383] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/16/2019] [Accepted: 09/09/2019] [Indexed: 02/06/2023]
Abstract
Prostaglandin E2 (PGE2) is a lipid mediator of inflammation and cancer progression. It is mainly formed via metabolism of arachidonic acid by cyclooxygenases (COX) and the terminal enzyme microsomal prostaglandin E synthase-1 (mPGES-1). Widely used non-steroidal anti-inflammatory drugs (NSAIDs) inhibit COX activity, resulting in decreased PGE2 production and symptomatic relief. However, NSAIDs block the production of many other lipid mediators that have important physiological and resolving actions, and these drugs cause gastrointestinal bleeding and/or increase the risk for severe cardiovascular events. Selective inhibition of downstream mPGES-1 for reduction in only PGE2 biosynthesis is suggested as a safer therapeutic strategy. This review covers the recent advances in characterization of new mPGES-1 inhibitors in preclinical models and their future clinical applications.
Collapse
|
23
|
Affiliation(s)
- Tony Avril
- Inserm U1242, University of Rennes, Rennes, France.,Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Eric Chevet
- Inserm U1242, University of Rennes, Rennes, France. .,Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| |
Collapse
|
24
|
Kanikarla-Marie P, Kopetz S, Hawk ET, Millward SW, Sood AK, Gresele P, Overman M, Honn K, Menter DG. Bioactive lipid metabolism in platelet "first responder" and cancer biology. Cancer Metastasis Rev 2019; 37:439-454. [PMID: 30112590 DOI: 10.1007/s10555-018-9755-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Platelets can serve as "first responders" in cancer and metastasis. This is partly due to bioactive lipid metabolism that drives both platelet and cancer biology. The two primary eicosanoid metabolites that maintain platelet rapid response homeostasis are prostacyclin made by endothelial cells that inhibits platelet function, which is counterbalanced by thromboxane produced by platelets during activation, aggregation, and platelet recruitment. Both of these arachidonic acid metabolites are inherently unstable due to their chemical structure. Tumor cells by contrast predominantly make more chemically stable prostaglandin E2, which is the primary bioactive lipid associated with inflammation and oncogenesis. Pharmacological, clinical, and epidemiologic studies demonstrate that non-steroidal anti-inflammatory drugs (NSAIDs), which target cyclooxygenases, can help prevent cancer. Much of the molecular and biological impact of these drugs is generally accepted in the field. Cyclooxygenases catalyze the rate-limiting production of substrate used by all synthase molecules, including those that produce prostaglandins along with prostacyclin and thromboxane. Additional eicosanoid metabolites include lipoxygenases, leukotrienes, and resolvins that can also influence platelets, inflammation, and carcinogenesis. Our knowledge base and technology are now progressing toward identifying newer molecular and cellular interactions that are leading to revealing additional targets. This review endeavors to summarize new developments in the field.
Collapse
Affiliation(s)
- Preeti Kanikarla-Marie
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Scott Kopetz
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Ernest T Hawk
- Office of the Vice President Cancer Prevention and Population Science, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Steven W Millward
- Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Anil K Sood
- Gynocologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.,Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Paolo Gresele
- Department of Medicine, Section of Internal and Cardiovascular Medicine, University of Perugia, Via E. Dal Pozzo, 06126, Perugia, Italy
| | - Michael Overman
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Kenneth Honn
- Bioactive Lipids Research Program, Department of Pathology, Wayne State University, 5101 Cass Ave. 430 Chemistry, Detroit, MI, 48202, USA.,Department of Pathology, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA.,Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA.,Department of Gastrointestinal Medical Oncology, M. D. Anderson Cancer Center, 1515 Holcombe Boulevard--Unit 0426, Houston, TX, 77030, USA
| | - David G Menter
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.
| |
Collapse
|
25
|
Kim SH, Roszik J, Cho SN, Ogata D, Milton DR, Peng W, Menter DG, Ekmekcioglu S, Grimm EA. The COX2 Effector Microsomal PGE2 Synthase 1 is a Regulator of Immunosuppression in Cutaneous Melanoma. Clin Cancer Res 2019; 25:1650-1663. [PMID: 30538110 PMCID: PMC6397703 DOI: 10.1158/1078-0432.ccr-18-1163] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 08/16/2018] [Accepted: 12/07/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE Microsomal prostaglandin E2 synthase 1 (mPGES1) was evaluated as an important downstream effector of the COX2 pathway responsible for tumor-mediated immunosuppression in melanoma. EXPERIMENTAL DESIGN The analysis of a stage III melanoma tissue microarray (n = 91) was performed to assess the association between mPGES1, COX2, CD8, and patient survival. Pharmacologic inhibitors and syngeneic mouse models using PTGES-knockout (KO) mouse melanoma cell lines were used to evaluate the mPGES1-mediated immunosuppressive function. RESULTS We observed correlations in expression and colocalization of COX2 and mPGES1, which are associated with increased expression of immunosuppressive markers in human melanoma. In a syngeneic melanoma mouse model, PTGES KO increased melanoma expression of PD-L1, increased infiltration of CD8a+ T cells, and CD8a+ dendritic cells into tumors and suppressed tumor growth. Durable tumor regression was observed in mice bearing PTGES KO tumors that were given anti-PD-1 therapy. Analysis of a stage III melanoma tissue microarray revealed significant associations between high mPGES1 expression and low CD8+ infiltration, which correlated with a shorter patient survival. CONCLUSIONS Our results are the first to illustrate a potential role for mPGES1 inhibition in melanoma immune evasion and selective targeting in supporting the durability of response to PD-1 checkpoint immunotherapy. More research effort in this drug development space is needed to validate the use of mPGES1 inhibitors as safe treatment options.
Collapse
Affiliation(s)
- Sun-Hee Kim
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sung-Nam Cho
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dai Ogata
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Denái R Milton
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Weiyi Peng
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David G Menter
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Suhendan Ekmekcioglu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth A Grimm
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
26
|
Larsson K, Kock A, Kogner P, Jakobsson PJ. Targeting the COX/mPGES-1/PGE 2 Pathway in Neuroblastoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1161:89-100. [PMID: 31562624 DOI: 10.1007/978-3-030-21735-8_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The importance of prostaglandin E2 in cancer progression is well established, but research on its role in cancer has so far mostly been focused on epithelial cancer in adults while the knowledge about the contribution of prostaglandin E2 to childhood malignancies is limited. Neuroblastoma, an extracranial solid tumor of the sympathetic nervous system, mainly affects young children. Patients with tumors classified as high-risk have poor survival despite receiving intensive treatment, illustrating a need for new treatments complimenting existing ones. The basis of neuroblastoma treatment e.g. chemotherapy and radiation therapy, target the proliferating genetically unstable tumor cells leading to treatment resistance and relapses. The tumor microenvironment is an avenue, still to a great extent, unexplored and lacking effective targeted therapies. Cancer-associated fibroblasts is the main source of prostaglandin E2 in neuroblastoma contributing to angiogenesis, immunosuppression and tumor growth. Prostaglandin E2 is formed from its precursor arachidonic acid in a two-step enzymatic reaction. Arachidonic acid is first converted by cyclooxygenases into prostaglandin H2 and then further converted by microsomal prostaglandin E synthase-1 into prostaglandin E2. We believe targeting of microsomal prostaglandin E synthase-1 in cancer-associated fibroblasts will be an effective future therapeutic strategy in fighting neuroblastoma.
Collapse
Affiliation(s)
- Karin Larsson
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden.
| | - Anna Kock
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Lauro G, Cantone V, Potenza M, Fischer K, Koeberle A, Werz O, Riccio R, Bifulco G. Discovery of 3-hydroxy-3-pyrrolin-2-one-based mPGES-1 inhibitors using a multi-step virtual screening protocol. MEDCHEMCOMM 2018; 9:2028-2036. [PMID: 30746063 DOI: 10.1039/c8md00497h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/17/2018] [Indexed: 12/20/2022]
Abstract
Targeting microsomal prostaglandin E2 synthase-1 (mPGES-1) represents an efficient strategy for the development of novel drugs against inflammation and cancer with potentially reduced side effects. With this aim, a virtual screening was performed on a large library of commercially available molecules using the X-ray structure of mPGES-1 co-complexed with a potent inhibitor. Combining fast ligand-based shape alignment, molecular docking experiments, and qualitative analysis of the binding poses, a small set of molecules was selected for the subsequent steps of validation of the biological activity. Compounds 2 and 3, bearing the 3-hydroxy-3-pyrrolin-2-one nucleus, showed mPGES-1-inhibitory activity in the low micromolar range. These data highlighted the applicability of the reported virtual screening protocol for the selection of new mPGES-1 inhibitors as promising anti-inflammatory/anti-cancer drugs.
Collapse
Affiliation(s)
- Gianluigi Lauro
- Department of Pharmacy , University of Salerno , via Giovanni Paolo II 132 , 84084 Fisciano , Italy . ; ; Tel: +39 (0)89 969741
| | - Vincenza Cantone
- Department of Pharmacy , University of Salerno , via Giovanni Paolo II 132 , 84084 Fisciano , Italy . ; ; Tel: +39 (0)89 969741
| | - Marianna Potenza
- Department of Pharmacy , University of Salerno , via Giovanni Paolo II 132 , 84084 Fisciano , Italy . ; ; Tel: +39 (0)89 969741
| | - Katrin Fischer
- Department of Pharmaceutical/Medicinal Chemistry , Institute of Pharmacy , Friedrich-Schiller-University Jena , Philosophenweg 14 , D-07743 Jena , Germany
| | - Andreas Koeberle
- Department of Pharmaceutical/Medicinal Chemistry , Institute of Pharmacy , Friedrich-Schiller-University Jena , Philosophenweg 14 , D-07743 Jena , Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry , Institute of Pharmacy , Friedrich-Schiller-University Jena , Philosophenweg 14 , D-07743 Jena , Germany
| | - Raffaele Riccio
- Department of Pharmacy , University of Salerno , via Giovanni Paolo II 132 , 84084 Fisciano , Italy . ; ; Tel: +39 (0)89 969741
| | - Giuseppe Bifulco
- Department of Pharmacy , University of Salerno , via Giovanni Paolo II 132 , 84084 Fisciano , Italy . ; ; Tel: +39 (0)89 969741
| |
Collapse
|
28
|
Napagoda M, Gerstmeier J, Butschek H, Lorenz S, Kanatiwela D, Qader M, Nagahawatte A, De Soyza S, Wijayaratne GB, Svatoš A, Jayasinghe L, Koeberle A, Werz O. Lipophilic extracts of Leucas zeylanica, a multi-purpose medicinal plant in the tropics, inhibit key enzymes involved in inflammation and gout. JOURNAL OF ETHNOPHARMACOLOGY 2018; 224:474-481. [PMID: 29727733 DOI: 10.1016/j.jep.2018.04.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/24/2018] [Accepted: 04/27/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Leucas zeylanica (L.) W.T. Aiton is a popular, multi-purpose medicinal plant in Sri Lanka but the pharmacological potential and the chemical profile have not been systematically investigated to understand and rationalize the reported ethnobotanical significance. AIM OF THE STUDY The present study was undertaken to scientifically validate the traditional usage of this plant for the treatment of inflammatory conditions, gout and microbial infections. Inhibition of 5-lipoxygenase (5-LO), microsomal prostaglandin E2 synthase (mPGES)-1 and xanthine oxidase (XO) by different extracts of L. zeylanica was investigated to determine the anti-inflammatory and anti-gout activity, respectively. The antibacterial and antifungal activities were also studied and the relevant constituents in the bioactive extracts were tentatively identified. MATERIALS AND METHODS Cell-free and/or cell-based assays were employed in order to investigate the effects of the extracts against the activity of human 5-LO, mPGES-1 and XO as well as to assess antioxidant properties. The antibacterial activity of the extracts was determined by the broth micro-dilution method against Gram positive and Gram negative bacteria including methicillin-resistant Staphylococcus aureus while the agar dilution method was employed to determine the anti-Candida activity. Gas chromatography coupled to mass spectrometric (GC-MS) analysis enabled the characterization of secondary metabolites in the extracts. RESULTS The dichloromethane extract of L. zeylanica efficiently inhibited 5-LO activity in stimulated human neutrophils (IC50 = 5.5 µg/mL) and isolated human 5-LO and mPGES-1 (IC50 = 2.2 and 0.4 µg/mL). Potent inhibition of XO was observed by the same extract (IC50 = 47.5 μg/mL), which is the first report of XO-inhibitory activity of a Sri Lankan medicinal plant. Interestingly, significant radical scavenging activity was not observed by this extract. Only the n-hexane extract exhibited antibacterial activity against Staphylococcus aureus and Staphylococcus saprophyticus with a MIC of 250 µg/mL while the anti-Candida activity was moderate. GC-MS analysis revealed the presence of phytosterols, fatty acids, sesquiterpenes, diterpenes and several other types of secondary metabolites. CONCLUSIONS Potent inhibition of 5-LO, mPGES-1 and XO rationalizes the ethnopharmacological use of L. zeylanica as anti-inflammatory and anti-gout remedy. Interestingly, the antimicrobial activities were not prominent, despite its wide utility as an antimicrobial medication.
Collapse
Affiliation(s)
- Mayuri Napagoda
- Department of Biochemistry, Faculty of Medicine, University of Ruhuna, Galle 80000, Sri Lanka.
| | - Jana Gerstmeier
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743 Jena, Germany.
| | - Hannah Butschek
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743 Jena, Germany.
| | - Sybille Lorenz
- Research Group Mass Spectrometry and Proteomics, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, D-07745 Jena, Germany.
| | - Dinusha Kanatiwela
- National Institute of Fundamental Studies, Hantana Road, Kandy 20000, Sri Lanka.
| | - Mallique Qader
- National Institute of Fundamental Studies, Hantana Road, Kandy 20000, Sri Lanka.
| | - Ajith Nagahawatte
- Department of Microbiology, Faculty of Medicine, University of Ruhuna, Galle 80000, Sri Lanka.
| | - Sudhara De Soyza
- Department of Biochemistry, Faculty of Medicine, University of Ruhuna, Galle 80000, Sri Lanka.
| | | | - Aleš Svatoš
- Research Group Mass Spectrometry and Proteomics, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, D-07745 Jena, Germany.
| | - Lalith Jayasinghe
- National Institute of Fundamental Studies, Hantana Road, Kandy 20000, Sri Lanka.
| | - Andreas Koeberle
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743 Jena, Germany.
| | - Oliver Werz
- Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, D-07743 Jena, Germany.
| |
Collapse
|
29
|
Gupta A, Chaudhary N, Aparoy P. MM-PBSA and per-residue decomposition energy studies on 7-Phenyl-imidazoquinolin-4(5H)-one derivatives: Identification of crucial site points at microsomal prostaglandin E synthase-1 (mPGES-1) active site. Int J Biol Macromol 2018; 119:352-359. [PMID: 30031079 DOI: 10.1016/j.ijbiomac.2018.07.050] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 12/29/2022]
Abstract
The huge therapeutic potential and the market share of painkillers are well-known. Due to the side effects associated with traditional NSAIDs and selective cyclooxygenase (COX-2) inhibitors, a new generation of painkillers is the need of the hour. In this regard, microsomal prostaglandin E synthase-1 (mPGES-1) offers great potential as an alternative drug target against inflammatory disorders. The present study is aimed at identifying the amino acids crucial in effective inhibitor binding at the mPGES-1 active site by performing molecular dynamics based studies on a series of 7-Phenyl-imidazoquinolin-4(5H)-one derivatives. Molecular dynamics (MD) simulations, MM-PBSA, per-residue energy decomposition and Dimensionality Reduction through Covariance matrix Transformation for Identification of Differences in dynamics (DIRECT-ID) analysis were performed to get insights into the structural details that can aid in novel drug design against mPGES-1. The high correlations of electrostatic and polar energy terms with biological activity highlight their importance and applicability in in silico screening studies. Further, per-residue energy decomposition studies revealed that Lys42, Arg52, Arg122, Pro124, Ser127, Val128 and Thr131 were contributing more towards inhibitor binding energy. The results clearly show that MM-PBSA can act as a filter in virtual screening experiments and can play major role in facilitating various mPGES-1 drug discovery studies.
Collapse
Affiliation(s)
- Ashish Gupta
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Dharamshala, Himachal Pradesh 176215, India
| | - Neha Chaudhary
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Dharamshala, Himachal Pradesh 176215, India
| | - Polamarasetty Aparoy
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Dharamshala, Himachal Pradesh 176215, India.
| |
Collapse
|
30
|
Muthukaman N, Deshmukh S, Tambe M, Pisal D, Tondlekar S, Shaikh M, Sarode N, Kattige VG, Sawant P, Pisat M, Karande V, Honnegowda S, Kulkarni A, Behera D, Jadhav SB, Sangana RR, Gudi GS, Khairatkar-Joshi N, Gharat LA. Alleviating CYP and hERG liabilities by structure optimization of dihydrofuran-fused tricyclic benzo[d]imidazole series - Potent, selective and orally efficacious microsomal prostaglandin E synthase-1 (mPGES-1) inhibitors: Part-2. Bioorg Med Chem Lett 2018. [PMID: 29519738 DOI: 10.1016/j.bmcl.2018.02.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In an effort to identify CYP and hERG clean mPGES-1 inhibitors from the dihydrofuran-fused tricyclic benzo[d]imidazole series lead 7, an extensive structure-activity relationship (SAR) studies were performed. Optimization of A, D and E-rings in 7 afforded many potent compounds with human whole blood potency in the range of 160-950 nM. Selected inhibitors 21d, 21j, 21m, 21n, 21p and 22b provided selectivity against COX-enzymes and mPGES-1 isoforms (mPGES-2 and cPGES) along with sufficient selectivity against prostanoid synthases. Most of the tested analogs demonstrated required metabolic stability in liver microsomes, low hERG and CYP liability. Oral pharmacokinetics and bioavailability of lead compounds 21j, 21m and 21p are discussed in multiple species like rat, guinea pig, dog, and cynomolgus monkey. Besides, these compounds revealed low to moderate activity against human pregnane X receptor (hPXR). The selected lead 21j further demonstrated in vivo efficacy in acute hyperalgesia (ED50: 39.6 mg/kg) and MIA-induced osteoarthritic pain models (ED50: 106 mg/kg).
Collapse
Affiliation(s)
- Nagarajan Muthukaman
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Sanjay Deshmukh
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Macchindra Tambe
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Dnyandeo Pisal
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Shital Tondlekar
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Mahamadhanif Shaikh
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Neelam Sarode
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Vidya G Kattige
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Pooja Sawant
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Monali Pisat
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Vikas Karande
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Srinivasa Honnegowda
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Abhay Kulkarni
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Dayanidhi Behera
- Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Satyawan B Jadhav
- Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Ramchandra R Sangana
- Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Girish S Gudi
- Drug Metabolism and Pharmacokinetics, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Neelima Khairatkar-Joshi
- Biological Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India
| | - Laxmikant A Gharat
- Chemical Research, Glenmark Pharmaceuticals Limited, Glenmark Research Center, Navi Mumbai, Maharashtra 400709, India.
| |
Collapse
|
31
|
Koeberle A, Werz O. Natural products as inhibitors of prostaglandin E 2 and pro-inflammatory 5-lipoxygenase-derived lipid mediator biosynthesis. Biotechnol Adv 2018; 36:1709-1723. [PMID: 29454981 DOI: 10.1016/j.biotechadv.2018.02.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/19/2018] [Accepted: 02/14/2018] [Indexed: 12/31/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) inhibit prostanoid formation and represent prevalent therapeutics for treatment of inflammatory disorders. However, NSAIDs are afflicted with severe side effects, which might be circumvented by more selective suppression of pro-inflammatory eicosanoid biosynthesis. This concept led to dual inhibitors of microsomal prostaglandin E2 synthase (mPGES)-1 and 5-lipoxygenase that are crucial enzymes in the biosynthesis of pro-inflammatory prostaglandin E2 and leukotrienes. The potential of their dual inhibition in light of superior efficacy and safety is discussed. Focus is placed on natural products, for which direct inhibition of mPGES-1 and leukotriene biosynthesis has been confirmed.
Collapse
Affiliation(s)
- Andreas Koeberle
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, Jena 07743, Germany.
| | - Oliver Werz
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, Jena 07743, Germany.
| |
Collapse
|
32
|
Allocati N, Masulli M, Di Ilio C, Federici L. Glutathione transferases: substrates, inihibitors and pro-drugs in cancer and neurodegenerative diseases. Oncogenesis 2018; 7:8. [PMID: 29362397 PMCID: PMC5833873 DOI: 10.1038/s41389-017-0025-3] [Citation(s) in RCA: 373] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/12/2017] [Indexed: 12/12/2022] Open
Abstract
Glutathione transferase classical GSH conjugation activity plays a critical role in cellular detoxification against xenobiotics and noxious compounds as well as against oxidative stress. However, this feature is also exploited by cancer cells to acquire drug resistance and improve their survival. As a result, various members of the family were found overexpressed in a number of different cancers. Moreover several GST polymorphisms, ranging from null phenotypes to point mutations, were detected in members of the family and found to correlate with the onset of neuro-degenerative diseases. In the last decades, a great deal of research aimed at clarifying the role played by GSTs in drug resistance, at developing inhibitors to counteract this activity but also at exploiting GSTs for prodrugs specific activation in cancer cells. Here we summarize some of the most important achievements reached in this lively area of research.
Collapse
Affiliation(s)
- Nerino Allocati
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy.
| | - Michele Masulli
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Carmine Di Ilio
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Luca Federici
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy.,CESI-MET, University "G. d'Annunzio", Chieti, Italy
| |
Collapse
|
33
|
Small Peptides Able to Suppress Prostaglandin E₂ Generation in Renal Mesangial Cells. Molecules 2018; 23:molecules23010158. [PMID: 29342835 PMCID: PMC6017359 DOI: 10.3390/molecules23010158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/10/2018] [Accepted: 01/10/2018] [Indexed: 11/17/2022] Open
Abstract
Peptide drug discovery may play a key role in the identification of novel medicinal agents. Here, we present the development of novel small peptides able to suppress the production of PGE₂ in mesangial cells. The new compounds were generated by structural alterations applied on GK115, a novel inhibitor of secreted phospholipase A₂, which has been previously shown to reduce PGE₂ synthesis in rat renal mesangial cells. Among the synthesized compounds, the tripeptide derivative 11 exhibited a nice dose-dependent suppression of PGE₂ production, similar to that observed for GK115.
Collapse
|
34
|
Mozos I, Malainer C, Horbańczuk J, Gug C, Stoian D, Luca CT, Atanasov AG. Inflammatory Markers for Arterial Stiffness in Cardiovascular Diseases. Front Immunol 2017; 8:1058. [PMID: 28912780 PMCID: PMC5583158 DOI: 10.3389/fimmu.2017.01058] [Citation(s) in RCA: 237] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/15/2017] [Indexed: 01/13/2023] Open
Abstract
Arterial stiffness predicts an increased risk of cardiovascular events. Inflammation plays a major role in large arteries stiffening, related to atherosclerosis, arteriosclerosis, endothelial dysfunction, smooth muscle cell migration, vascular calcification, increased activity of metalloproteinases, extracellular matrix degradation, oxidative stress, elastolysis, and degradation of collagen. The present paper reviews main mechanisms explaining the crosstalk between inflammation and arterial stiffness and the most common inflammatory markers associated with increased arterial stiffness, considering the most recent clinical and experimental studies. Diverse studies revealed significant correlations between the severity of arterial stiffness and inflammatory markers, such as white blood cell count, neutrophil/lymphocyte ratio, adhesion molecules, fibrinogen, C-reactive protein, cytokines, microRNAs, and cyclooxygenase-2, in patients with a broad variety of diseases, such as metabolic syndrome, diabetes, coronary heart disease, peripheral arterial disease, malignant and rheumatic disorders, polycystic kidney disease, renal transplant, familial Mediterranean fever, and oral infections, and in women with preeclampsia or after menopause. There is strong evidence that inflammation plays an important and, at least, partly reversible role in the development of arterial stiffness, and inflammatory markers may be useful additional tools in the assessment of the cardiovascular risk in clinical practice. Combined assessment of arterial stiffness and inflammatory markers may improve non-invasive assessment of cardiovascular risk, enabling selection of high-risk patients for prophylactic treatment or more regular medical examination. Development of future destiffening therapies may target pro-inflammatory mechanisms.
Collapse
Affiliation(s)
- Ioana Mozos
- Department of Functional Sciences, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania.,Center for Translational Research and Systems Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | | | - Jarosław Horbańczuk
- The Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzębiec, Poland
| | - Cristina Gug
- Department of Microscopic Morphology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Dana Stoian
- 2nd Department of Internal Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Constantin Tudor Luca
- Department of Cardiology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Atanas G Atanasov
- The Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzębiec, Poland.,Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Vienna, Austria.,Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|