1
|
Shateri S, Khatami SH, Haghbin Toutounchi A, Rajaei S, Mahdavi M, Mahmoodi Baram S, Shahidi GA, Habibi AH, Aghamollaii V, Ghlichnia B, Safakish L, Doagoo A, Salmani F, Tafakhori A, Keramatinia A, Shahmohammadi MR, Karima S. Plasma cytokines profile in patients with Alzheimer's and Parkinson's Disease: a comparative study in terms of inflammation. Int J Neurosci 2025; 135:158-167. [PMID: 38064237 DOI: 10.1080/00207454.2023.2292951] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/05/2023] [Accepted: 12/02/2023] [Indexed: 02/04/2025]
Abstract
BACKGROUND Neurodegenerative disorders such as Alzheimer's and Parkinson's disease inflict economic and health burdens on societies. Alzheimer's disease (AD), the most prevalent form of dementia, is accompanied by progressive degradation of memory, decision-making, and judgment. Parkinson's disease (PD) is characterized by resting tremor, rigidity, bradykinesia, and loss of balance. Extensive research has pinpointed inflammation as a cause of the onset and progression of both diseases. However, it has not been confirmed which one is more formidable in terms of inflammation. METHODS To assess the extent of inflammation that is implicated in AD and PD and answer the question of which one is more inflammatory, serum levels of inflammatory biomarkers, including cytokines, chemokines, and prostaglandin E2 (PEG2), were measured in AD and PD patients as well as a healthy group. RESULTS Our results showed a significant increase in IL-1α, IL-1β, IL-4, IL-6, IL-10, IL-12p70, IP-10, MCP-1, PEG2, and TNF-α in AD and PD patients compared with the control. Interestingly, IFN-γ did not manifest any significant difference in AD or PD patients compared with the control. CONCLUSION As a hallmark of our results, it could be inferred that inflammation, as the underlying etiological cause, plays a more crucial role in PD compared with AD. Based on our results, it is proposed that anti-inflammatory remedies would be putatively more effective in PD rather than AD.
Collapse
Affiliation(s)
- Somayeh Shateri
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Alireza Haghbin Toutounchi
- Department of General Surgery, Imam Hosein Medical and Educational Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shima Rajaei
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
- Clinical Study Department, Behbalin Inc, Tehran, Iran
| | - Meisam Mahdavi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Somayeh Mahmoodi Baram
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
- Clinical Study Department, Behbalin Inc, Tehran, Iran
| | - Gholam-Ali Shahidi
- Neurology Department, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Amir Hossein Habibi
- Neurology Department, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Vajiheh Aghamollaii
- Neurology Department, Roozbeh Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Babak Ghlichnia
- Department of Neurology, School of Medicine, Iranian Center of Neurological Research, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Lily Safakish
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Alireza Doagoo
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Farzaneh Salmani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Abbas Tafakhori
- Department of Neurology, School of Medicine, Iranian Center of Neurological Research, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Aliasghar Keramatinia
- Department of Community Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Shahmohammadi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| |
Collapse
|
2
|
Mohamed SK, Karthikeyan S, Khamies E, Ahsin A, Bakhite E, S Marae I, I El-Emary T, Mague JT, I Said A, Al-Salahi R, El Bakri Y. Synthesis, structural and X-ray analysis evaluations and computational studies of newly tetrahydroisoquinoline derivatives as potent against microsomal prostaglandin E synthase 1. J Biomol Struct Dyn 2023; 42:12645-12659. [PMID: 37878040 DOI: 10.1080/07391102.2023.2272745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/10/2023] [Indexed: 10/26/2023]
Abstract
Tetrahydroisoquinolines (THIQs) are a significant class within the broad range of natural compounds known as isoquinoline alkaloids. Natural and manmade drugs based on THIQ have a variety of biological effects that protect against different infectious pathogens and neurological diseases. In this study, two new THIQ derivatives were synthesized and characterized using by X-ray crystallographic analysis. The performed Hirshfeld analysis shows the intermolecular interactions and reactive sites of compounds. The 2D fingerprints reveal dominants H···C interactions up to 8.8% in 3a while 43% H···H elemental interactions are observed in compound 3b. In studied compound 3a, the repulsion energies (k-rep) dominate the other energies where the highest amount of 63.8 kJ/mol is obtained whereas 3b has a significant contribution from E-dis to the total energy of the molecule from the energy framework study. Moreover, the density functional theory study reveals better thermodynamic and electronic stabilities. These compounds have reduced HOMO-LUMO gaps (EH-L) ranging from 3.66 to 3.60 eV, indicating their remarkable conductive and electronic properties. The significant reduction in EH-L also guarantees our synthesized compounds' soft nature and reactivity. Our studied compound's NBO charges and MEPs analysis show electron-rich sites and donor-acceptor mechanism. Our synthesized compounds have remarkable polarizability (αo) and hyperpolarizability (βo) values (446.23 - 1312.73 au), which indicates their optical and nonlinear optical properties. The density of states spectra further illustrates their notable structural-electronic properties and reduced band gaps. Based on structural activity relationship studies, we found that these tetrahydro-isoquinolines derivatives are potent against microsomal prostaglandin E synthase 1(MPGES1), the docking analysis shows that studied compounds have a good binding affinity with MPGES1, and further ADME/T analysis was carried out for both compounds. In addtion to this molecular dynamics, studies were performed to understand the binding stability of both compounds in protien complex system during 100 ns simulation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shaaban K Mohamed
- Chemistry and Environmental Division, Manchester Metropolitan University, Manchester, England
| | - Subramani Karthikeyan
- Center for Healthcare Advancement, Innovation and Research, Vellore Institute of Technology University, Chennai Campus, Chennai, India
| | - Esraa Khamies
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Atazaz Ahsin
- Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Etify Bakhite
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Islam S Marae
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Talaat I El-Emary
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Joel T Mague
- Department of Chemistry, Tulane University, New Orleans, LA, USA
| | - Awad I Said
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Rashad Al-Salahi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Youness El Bakri
- Department of Theoretical and Applied Chemistry, South Ural State University, Chelyabinsk, Russian Federation
| |
Collapse
|
3
|
Liu D, Tang F, Zhang L, Zhang JN, Zhao XL, Xu LY, Peng C, Ao H. Alpinia katsumadai Hayata Volatile Oil Is Effective in Treating 5-Fluorouracil-Induced Mucositis by Regulating Gut Microbiota and Modulating the GC/GR Pathway and the mPGES-1/PGE2/EP4 Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15156-15169. [PMID: 37800952 DOI: 10.1021/acs.jafc.3c05051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
This study was aimed to investigate the therapeutic effect and mechanism of AKHO on 5-fluorouracil (5-FU)-induced intestinal mucositis in mice. Mouse body weight, diarrhea score, and H&E staining were applied to judge the therapeutic effect of AKHO. 16S rDNA and nontargeted metabolomics have been used to study the mechanism. WB, ELISA, and immunohistochemistry were adopted to validate possible mechanisms. The results demonstrated that AKHO significantly reduced diarrhea scores and intestinal damage induced by 5-FU in mice. AKHO lowered the serum levels of LD and DAO, and upregulated the expressions of ZO-1 and occludin in the ileum. Also, AKHO upregulated the abundance of Lactobacillus in the gut and suppressed KEGG pathways such as cortisol synthesis and secretion and arachidonic acid metabolism. Further validation studies indicated that AKHO downregulated the expressions of prostaglandin E2 (PGE2), microsomal prostaglandin E synthase-1 (mPGES-1), and PGE2 receptor EP4, as well as upregulated the expression of glucocorticoid (GC) receptor (GR), leading to improved intestinal epithelial barrier function. Taken together, AKHO elicited protective effects against 5-FU-induced mucositis by regulating the expressions of tight junction proteins via modulation of GC/GR and mPGES-1/PGE2/EP4 pathway, providing novel insights into the utilization and development of this pharmaceutical/food resource.
Collapse
Affiliation(s)
- Dong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Li Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jing-Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiao-Lan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Li-Yue Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
4
|
Di Giacomo C, Malfa GA, Tomasello B, Bianchi S, Acquaviva R. Natural Compounds and Glutathione: Beyond Mere Antioxidants. Antioxidants (Basel) 2023; 12:1445. [PMID: 37507985 PMCID: PMC10376414 DOI: 10.3390/antiox12071445] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
The tripeptide glutathione plays important roles in many cell processes, including differentiation, proliferation, and apoptosis; in fact, disorders in glutathione homeostasis are involved both in the etiology and in the progression of several human diseases, including cancer. Natural compounds have been found to modulate glutathione levels and function beyond their role as mere antioxidants. For example, certain compounds can upregulate the expression of glutathione-related enzymes, increase the availability of cysteine, the limiting amino acid for glutathione synthesis, or directly interact with glutathione and modulate its function. These compounds may have therapeutic potential in a variety of disease states where glutathione dysregulation is a contributing factor. On the other hand, flavonoids' potential to deplete glutathione levels could be significant for cancer treatment. Overall, while natural compounds may have potential therapeutic and/or preventive properties and may be able to increase glutathione levels, more research is needed to fully understand their mechanisms of action and their potential benefits for the prevention and treatment of several diseases. In this review, particular emphasis will be placed on phytochemical compounds belonging to the class of polyphenols, terpenoids, and glucosinolates that have an impact on glutathione-related processes, both in physiological and pathological conditions. These classes of secondary metabolites represent the most food-derived bioactive compounds that have been intensively explored and studied in the last few decades.
Collapse
Affiliation(s)
- Claudia Di Giacomo
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Research Centre on Nutraceuticals and Health Products (CERNUT), University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Giuseppe Antonio Malfa
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Research Centre on Nutraceuticals and Health Products (CERNUT), University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Research Centre on Nutraceuticals and Health Products (CERNUT), University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Simone Bianchi
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Research Centre on Nutraceuticals and Health Products (CERNUT), University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Rosaria Acquaviva
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Research Centre on Nutraceuticals and Health Products (CERNUT), University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
5
|
Santacroce L, Colella M, Charitos IA, Di Domenico M, Palmirotta R, Jirillo E. Microbial and Host Metabolites at the Backstage of Fever: Current Knowledge about the Co-Ordinate Action of Receptors and Molecules Underlying Pathophysiology and Clinical Implications. Metabolites 2023; 13:461. [PMID: 36984901 PMCID: PMC10056708 DOI: 10.3390/metabo13030461] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Fever represents an elevation of body temperature, that exerts a protective effect against pathogens. Innate immune cells and neurons are implicated in the regulation of body temperature. Pathogen-associated molecular patterns, i.e., lipopolysaccharides from Gram-negative bacteria and peptidoglycan and lipoteichoic acid from Gram-positive bacteria are exogenous pyrogens, that bind to Toll-like receptors on immune and non-immune cells. The subsequent release of pro-inflammatory cytokines [interleukin-1 (IL-1), IL-6 and Tumor necrosis factor-alpha] and their passage through the brain trigger the febrile response. In fact, neurons of the pre-optic area produce prostaglandin E2 (PGE2), that, in turn, bind to the PGE2 receptors; thus, generating fever. Apart from classical non-steroidal anti-inflammatory drugs, i.e., aspirin and acetaminophen, various botanicals are currently used as antipyretic agents and, therefore, their mechanisms of action will be elucidated.
Collapse
Affiliation(s)
- Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (L.S.); (R.P.); (E.J.)
| | - Marica Colella
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (L.S.); (R.P.); (E.J.)
| | - Ioannis Alexandros Charitos
- CEDICLO—Interdepartmental Research Center for Pre-Latin, Latin and Oriental Rights and Culture Studies, University of Bari, 70121 Bari, Italy
| | - Marina Di Domenico
- Department of Precision Medicine, University of Campania ‘Luigi Vanvitelli’, 80138 Naples, Italy;
| | - Raffaele Palmirotta
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (L.S.); (R.P.); (E.J.)
| | - Emilio Jirillo
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy; (L.S.); (R.P.); (E.J.)
| |
Collapse
|
6
|
Kotsos D, Tziomalos K. Microsomal Prostaglandin E Synthase-1 and -2: Emerging Targets in Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2023; 24:3049. [PMID: 36769370 PMCID: PMC9918023 DOI: 10.3390/ijms24033049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects a substantial proportion of the general population and is even more prevalent in obese and diabetic patients. NAFLD, and particularly the more advanced manifestation of the disease, nonalcoholic steatohepatitis (NASH), increases the risk for both liver-related and cardiovascular morbidity. The pathogenesis of NAFLD is complex and multifactorial, with many molecular pathways implicated. Emerging data suggest that microsomal prostaglandin E synthase-1 and -2 might participate in the development and progression of NAFLD. It also appears that targeting these enzymes might represent a novel therapeutic approach for NAFLD. In the present review, we discuss the association between microsomal prostaglandin E synthase-1 and -2 and NAFLD.
Collapse
Affiliation(s)
| | - Konstantinos Tziomalos
- First Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA Hospital, 54636 Thessaloniki, Greece
| |
Collapse
|
7
|
Prostanoid Metabolites as Biomarkers in Human Disease. Metabolites 2022; 12:metabo12080721. [PMID: 36005592 PMCID: PMC9414732 DOI: 10.3390/metabo12080721] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Prostaglandins (PGD2, PGE2, PGF2α), prostacyclin (PGI2), and thromboxane A2 (TXA2) together form the prostanoid family of lipid mediators. As autacoids, these five primary prostanoids propagate intercellular signals and are involved in many physiological processes. Furthermore, alterations in their biosynthesis accompany a wide range of pathological conditions, which leads to substantially increased local levels during disease. Primary prostanoids are chemically instable and rapidly metabolized. Their metabolites are more stable, integrate the local production on a systemic level, and their analysis in various biological matrices yields valuable information under different pathological settings. Therefore, prostanoid metabolites may be used as diagnostic, predictive, or prognostic biomarkers in human disease. Although their potential as biomarkers is great and extensive research has identified major prostanoid metabolites that serve as target analytes in different biofluids, the number of studies that correlate prostanoid metabolite levels to disease outcome is still limited. We review the metabolism of primary prostanoids in humans, summarize the levels of prostanoid metabolites in healthy subjects, and highlight existing biomarker studies. Since analysis of prostanoid metabolites is challenging because of ongoing metabolism and limited half-lives, an emphasis of this review lies on the reliable measurement and interpretation of obtained levels.
Collapse
|
8
|
Abstract
PURPOSE Our understanding of thyroid-associated ophthalmopathy (TAO, A.K.A Graves' orbitopathy, thyroid eye disease) has advanced substantially, since one of us (TJS) wrote the 2010 update on TAO, appearing in this journal. METHODS PubMed was searched for relevant articles. RESULTS Recent insights have resulted from important studies conducted by many different laboratory groups around the World. A clearer understanding of autoimmune diseases in general and TAO specifically emerged from the use of improved research methodologies. Several key concepts have matured over the past decade. Among them, those arising from the refinement of mouse models of TAO, early stage investigation into restoring immune tolerance in Graves' disease, and a hard-won acknowledgement that the insulin-like growth factor-I receptor (IGF-IR) might play a critical role in the development of TAO, stand out as important. The therapeutic inhibition of IGF-IR has blossomed into an effective and safe medical treatment. Teprotumumab, a β-arrestin biased agonist monoclonal antibody inhibitor of IGF-IR has been studied in two multicenter, double-masked, placebo-controlled clinical trials demonstrated both effectiveness and a promising safety profile in moderate-to-severe, active TAO. Those studies led to the approval by the US FDA of teprotumumab, currently marketed as Tepezza for TAO. We have also learned far more about the putative role that CD34+ fibrocytes and their derivatives, CD34+ orbital fibroblasts, play in TAO. CONCLUSION The past decade has been filled with substantial scientific advances that should provide the necessary springboard for continually accelerating discovery over the next 10 years and beyond.
Collapse
Affiliation(s)
- E J Neag
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, Brehm Tower, 1000 Wall Street, Ann Arbor, MI, 48105, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
- Michigan State University College of Osteopathic Medicine, East Lansing, MI, USA
| | - T J Smith
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, Brehm Tower, 1000 Wall Street, Ann Arbor, MI, 48105, USA.
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
9
|
Leyva‐Grado V, Pugach P, Sadeghi‐Latefi N. A novel anti-inflammatory treatment for bradykinin-induced sore throat or pharyngitis. Immun Inflamm Dis 2021; 9:1321-1335. [PMID: 34153179 PMCID: PMC8589389 DOI: 10.1002/iid3.479] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Often thought of as a minor health concern, sore throat or pharyngitis is an important public health issue. It is one of the most common symptoms of upper respiratory diseases including COVID-19 and is a leading cause of physician visits and antibiotic prescriptions. However, few over-the-counter medications are proven to heal sore throat inflammation. METHODS Adenocarcinomic human alveolar basal epithelial cells (A549 cells) and three dimensional organotypic human respiratory tissues were used to study inflammation and various treatment effects on respiratory epithelia. The cells and tissues were studied both in the presence and absence of bradykinin, one of the first inflammatory mediators of pharyngitis. Inflammation was measured by analyzing the levels of prostaglandin E2 (PGE2), interleukin 8 (IL-8), and leukotriene B4 (LTB4), transepithelial electrical resistance (TEER), and lactate dehydrogenase (LDH) release. Tissue morphology was analyzed by immunohistochemistry. RESULTS In studying pharyngitis using organotypic human respiratory tissue stimulated with bradykinin, we saw an increase in PGE2 and interleukin-8 (IL-8) in response to bradykinin. Acetyl salicylic acid (ASA), a nonspecific COX inhibitor, was able to mitigate a bradykinin-induced increase in PGE2 in our studies. However, ASA was inflammatory above its therapeutic window, increasing the levels of PGE2 and IL-8 above those seen with bradykinin stimulation alone. We describe a novel, scientifically validated treatment for sore throat, that contains a low dose of ASA and other anti-inflammatory ingredients. CONCLUSION This study elucidates the complex mechanisms involved in healing pharyngitis, an inflammatory condition of the upper respiratory epithelia. An ASA-based formula (Biovanta) mitigated bradykinin-induced inflammation more strongly than ASA alone in organotypic human respiratory tissues. Surprisingly, we found that many of the most common over the counter sore throat therapies exacerbate inflammation and IL-8 in organotypic human respiratory tissues, suggesting these common treatments may increase the likelihood of further respiratory complications.
Collapse
Affiliation(s)
| | - Pavel Pugach
- Applied Biological LaboratoriesBrooklynNew YorkUSA
| | | |
Collapse
|
10
|
Budik S, Walter I, Leitner MC, Ertl R, Aurich C. Expression of Enzymes Associated with Prostaglandin Synthesis in Equine Conceptuses. Animals (Basel) 2021; 11:ani11041180. [PMID: 33924239 PMCID: PMC8074782 DOI: 10.3390/ani11041180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary The mobile preimplantative phase of equine gestation, taking place between day 9 and 16 after ovulation, is characterized by peristaltic contractions of the uterus caused by secretion of prostaglandins by the spheric equine conceptus. This mobility is necessary for maternal recognition of pregnancy in equids, taking place around day 14 after ovulation. The presented study investigated the spatial and temporal abundance of prostaglandin synthesis enzymes of the equine conceptus, elucidating a basal and an inducible system for prostaglandin E2. Prostaglandin F2α synthesis is restricted to the “periembryonic”pole area and relies on enzymatic conversion of prostaglandin E2. This scenario led to a model able to explain the embryonic forward motion driven by the peristaltic contractions of the uterus. In vitro incubation of primary trophoblast cell cultures with oxytocin showed no influence of this hormone on prostaglandin synthesis. Abstract In the horse, mobility of the conceptus is required for maternal recognition of pregnancy depending on secretion of prostaglandins by the conceptus. The aim of this study was to determine the expression and localization of key enzymes of the different pathways leading to synthesis of prostaglandin E2 and F2α in the equine conceptus during the mobility phase. Enzyme expression was analyzed via quantitative RT-PCR in total RNA samples of equine conceptuses collected on days 10 (n = 5), 12 (n = 12), 14 (n = 5) and 16 (n = 7) from healthy mares. Relative abundance of cyclooxygenase (COX)-2 mRNA was higher (p < 0.05) than of COX-1 irrespective of conceptus age and for phospholipase A2 on day 16 in comparison to all other days (p < 0.01). Abundance of mRNA of cytosolic and microsomal prostaglandin E synthase (PGES) and of carbonyl reductase (CBR) 1 was not influenced by conceptus age. Immunohistochemically, COX-1, COX-2, as well as cytosolic and microsomal PGES were present in both the ectodermal and endodermal layer of the yolk sac wall. CBR-1 was restricted to periembryonic disc area. The localisation of the key enzymes explains the mechanism of embryo mobility. In vitro incubation of primary trophoblast cell cultures with oxytocin had no effect on key enzyme synthesis.
Collapse
Affiliation(s)
- Sven Budik
- Platform for Artificial Insemination and Embryo Transfer, Department for Small Animals and Horses, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (M.-C.L.); (C.A.)
- Correspondence: ; Tel.: +43-125-077-6403
| | - Ingrid Walter
- Department of Pathobiology, Institute of Anatomy, Histology and Embryology, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria;
- VetCore Facility for Research, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria;
| | - Marie-Christine Leitner
- Platform for Artificial Insemination and Embryo Transfer, Department for Small Animals and Horses, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (M.-C.L.); (C.A.)
| | - Reinhard Ertl
- VetCore Facility for Research, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria;
| | - Christine Aurich
- Platform for Artificial Insemination and Embryo Transfer, Department for Small Animals and Horses, Vetmeduni Vienna, Veterinärplatz 1, 1210 Vienna, Austria; (M.-C.L.); (C.A.)
| |
Collapse
|
11
|
Ma LL, Liu HM, Luo CH, He YN, Wang F, Huang HZ, Han L, Yang M, Xu RC, Zhang DK. Fever and Antipyretic Supported by Traditional Chinese Medicine: A Multi-Pathway Regulation. Front Pharmacol 2021; 12:583279. [PMID: 33828481 PMCID: PMC8020597 DOI: 10.3389/fphar.2021.583279] [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: 07/14/2020] [Accepted: 01/28/2021] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease, 2019 (COVID-19), has spread rapidly around the world and become a major public health problem facing the world. Traditional Chinese medicine (TCM) has been fully committed to treat COVID-19 in China. It improved the clinical symptoms of patients and reduced the mortality rate. In light of the fever was identified as one of leading clinical features of COVID-19, this paper will first analyze the material basis of fever, including pyrogenic cytokines and a variety of the mediators of fever. Then the humoral and neural pathways of fever signal transmission will be described. The scattered evidences about fever recorded in recent years are connected in series. On this basis, the understanding of fever is further deepened from the aspects of pathology and physiology. Finally, combining with the chemical composition and pharmacological action of available TCM, we analyzed the mechanisms of TCMs to play the antipyretic effect through multiple ways. So as to further provide the basis for the research of antipyretic compound preparations of TCMs and explore the potential medicines for the prevention and treatment of COVID-19.
Collapse
Affiliation(s)
- Le-Le Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Hui-Min Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Chuan-Hong Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Ya-Nan He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Fang Wang
- State key Laboratory of Innovation Medicine and High Efficiency and Energy Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, PR China
| | - Hao-Zhou Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Li Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Ming Yang
- State key Laboratory of Innovation Medicine and High Efficiency and Energy Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, PR China
| | - Run-Chun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| | - Ding-Kun Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, PR China
| |
Collapse
|
12
|
Bedoui Y, Septembre-Malaterre A, Giry C, Jaffar-Bandjee MC, Selambarom J, Guiraud P, Gasque P. Robust COX-2-mediated prostaglandin response may drive arthralgia and bone destruction in patients with chronic inflammation post-chikungunya. PLoS Negl Trop Dis 2021; 15:e0009115. [PMID: 33596205 PMCID: PMC7920362 DOI: 10.1371/journal.pntd.0009115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/01/2021] [Accepted: 01/07/2021] [Indexed: 01/09/2023] Open
Abstract
Patients following infection by chikungunya virus (CHIKV) can suffer for months to years from arthralgia and arthritis. Interestingly, methotrexate (MTX) a major immune-regulatory drug has proved to be of clinical benefit. We have previously shown that CHIKV can persist in the joint of one patient 18 months post-infection and plausibly driving chronic joint inflammation but through ill-characterized mechanisms. We have pursued our investigations and report novel histological and in vitro data arguing for a plausible role of a COX-2-mediated inflammatory response post-CHIKV. In the joint, we found a robust COX-2 staining on endothelial cells, synovial fibroblasts and more prominently on multinucleated giant cells identified as CD11c+ osteoclasts known to be involved in bone destruction. The joint tissue was also strongly stained for CD3, CD8, CD45, CD14, CD68, CD31, CD34, MMP2, and VEGF (but not for NO synthase and two B cell markers). Dendritic cells were rarely detected. Primary human synovial fibroblasts were infected with CHIKV or stimulated either by the synthetic molecule polyriboinosinic:polyribocytidylic acid (PIC) to mimic chronic viral infection or cytokines. First, we found that PIC and CHIKV enhanced mRNA expression of COX-2. We further found that PIC but not CHIKV increased the mRNA levels of cPLA2α and of mPGES-1, two other central enzymes in PGE2 production. IFNβ upregulated cPLA2α and COX-2 transcription levels but failed to modulated mPGES-1 mRNA expression. Moreover, PIC, CHIKV and IFNβ decreased mRNA expression of the PGE2 degrading enzyme 15-PGDH. Interestingly, MTX failed to control the expression of all these enzymes. In sharp contrast, dexamethasone was able to control the capacity of pro-inflammatory cytokines, IL-1β as well as TNFα, to stimulate mRNA levels of cPLA2α, COX-2 and mPGES-1. These original data argue for a concerted action of CHIKV (including viral RNA) and cytokines plausibly released from recruited leukocytes to drive a major COX-2-mediated PGE2 proinflammatory responses to induce viral arthritis. It is important to have a better understanding of the immuno-pathogenesis of Chikungunya virus (CHIKV) and particularly focusing on the chronic phase associated to arthralgia and arthritis. Benefiting from our prospective cohort studies, we herein provide novel in vivo data identifying for the first time the implication of COX-2 and several other enzymes involved in prostaglandin biosynthesis and the persistence of the virus on the joint. Prostaglandin has major activities in inflammation and joint destruction. In vitro, we have used a model of human synovial fibroblasts to decipher the regulatory mechanisms of prostaglandin biosynthesis pathway. We have made important observations showing that the virus itself as well as major inflammatory cytokines can dramatically control the expression of all enzymes involved in the metabolism of prostaglandin. Interestingly, pharmacological investigations further revealed that dexamethasone, but not methotrexate (currently used to treat patients with chikungunya) may be of clinical values.
Collapse
Affiliation(s)
- Yosra Bedoui
- Unité mixte de recherche sur les processus infectieux en milieu insulaire tropical, INSERM U1187, CNRS 9192, IRD 249, Université de La Réunion—Plateforme Technologique CYROI Sainte-Clotilde, Île de La Réunion, France
- Laboratoire d’immunologie clinique et expérimentale de la zone de l’océan indien CHU La Réunion site Félix Guyon, Allée des Topazes, Saint Denis de La Réunion, France
| | - Axelle Septembre-Malaterre
- Unité de recherche Etudes Pharmaco-Immunologie, Université de la Réunion, CHU La Réunion site Félix Guyon, Allée des Topazes, Saint Denis de La Réunion, France
| | - Claude Giry
- Laboratoire de biologie, CNR associé des arbovirus, CHU La Réunion site Félix Guyon, Allée des Topazes, Saint Denis de La Réunion, France
| | - Marie-Christine Jaffar-Bandjee
- Laboratoire de biologie, CNR associé des arbovirus, CHU La Réunion site Félix Guyon, Allée des Topazes, Saint Denis de La Réunion, France
| | - Jimmy Selambarom
- Unité mixte de recherche sur les processus infectieux en milieu insulaire tropical, INSERM U1187, CNRS 9192, IRD 249, Université de La Réunion—Plateforme Technologique CYROI Sainte-Clotilde, Île de La Réunion, France
| | - Pascale Guiraud
- Unité mixte de recherche sur les processus infectieux en milieu insulaire tropical, INSERM U1187, CNRS 9192, IRD 249, Université de La Réunion—Plateforme Technologique CYROI Sainte-Clotilde, Île de La Réunion, France
| | - Philippe Gasque
- Unité mixte de recherche sur les processus infectieux en milieu insulaire tropical, INSERM U1187, CNRS 9192, IRD 249, Université de La Réunion—Plateforme Technologique CYROI Sainte-Clotilde, Île de La Réunion, France
- Laboratoire d’immunologie clinique et expérimentale de la zone de l’océan indien CHU La Réunion site Félix Guyon, Allée des Topazes, Saint Denis de La Réunion, France
- * E-mail: ,
| |
Collapse
|
13
|
Ji S, Guo R, Wang J, Qian L, Liu M, Xu H, Zhang J, Guan Y, Yang G, Chen L. Microsomal Prostaglandin E 2 Synthase-1 Deletion Attenuates Isoproterenol-Induced Myocardial Fibrosis in Mice. J Pharmacol Exp Ther 2020; 375:40-48. [PMID: 32759273 DOI: 10.1124/jpet.120.000023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/10/2020] [Indexed: 11/22/2022] Open
Abstract
Deletion of microsomal prostaglandin E2 synthase-1 (mPGES-1) inhibits inflammation and protects against atherosclerotic vascular diseases but displayed variable influence on pathologic cardiac remodeling. Overactivation of β-adrenergic receptors (β-ARs) causes heart dysfunction and cardiac remodeling, whereas the role of mPGES-1 in β-AR-induced cardiac remodeling is unknown. Here we addressed this question using mPGES-1 knockout mice, subjecting them to isoproterenol, a synthetic nonselective agonist for β-ARs, at 5 or 15 mg/kg per day to induce different degrees of cardiac remodeling in vivo. Cardiac structure and function were assessed by echocardiography 24 hours after the last of seven consecutive daily injections of isoproterenol, and cardiac fibrosis was examined by Masson trichrome stain in morphology and by real-time polymerase chain reaction for the expression of fibrosis-related genes. The results showed that deletion of mPGES-1 had no significant effect on isoproterenol-induced cardiac dysfunction or hypertrophy. However, the cardiac fibrosis was dramatically attenuated in the mPGES-1 knockout mice after either low-dose or high-dose isoproterenol exposure. Furthermore, in vitro study revealed that overexpression of mPGES-1 in cultured cardiac fibroblasts increased isoproterenol-induced fibrosis, whereas knocking down mPGES-1 in cardiac myocytes decreased the fibrogenesis of fibroblasts. In conclusion, mPGES-1 deletion protects against isoproterenol-induced cardiac fibrosis in mice, and targeting mPGES-1 may represent a novel strategy to attenuate pathologic cardiac fibrosis, induced by β-AR agonists. SIGNIFICANCE STATEMENT: Inhibitors of microsomal prostaglandin E2 synthase-1 (mPGES-1) are being developed as alternative analgesics that are less likely to elicit cardiovascular hazards than cyclooxygenase-2 selective nonsteroidal anti-inflammatory drugs. We have demonstrated that deletion of mPGES-1 protects inflammatory vascular diseases and promotes post-myocardial infarction survival. The role of mPGES-1 in β-adrenergic receptor-induced cardiomyopathy is unknown. Here we illustrated that deletion of mPGES-1 alleviated isoproterenol-induced cardiac fibrosis without deteriorating cardiac dysfunction. These results illustrated that targeting mPGES-1 may represent an efficacious approach to the treatment of inflammatory cardiovascular diseases.
Collapse
Affiliation(s)
- Shuang Ji
- Advanced Institute for Medical Sciences, Dalian Medical University, China (S.J., R.G., J.W., L.Q., M.L., H.X., J.Z., Y.G., L.C.) and School of Bioengineering, Dalian University of Technology, China (G.Y.)
| | - Rui Guo
- Advanced Institute for Medical Sciences, Dalian Medical University, China (S.J., R.G., J.W., L.Q., M.L., H.X., J.Z., Y.G., L.C.) and School of Bioengineering, Dalian University of Technology, China (G.Y.)
| | - Jing Wang
- Advanced Institute for Medical Sciences, Dalian Medical University, China (S.J., R.G., J.W., L.Q., M.L., H.X., J.Z., Y.G., L.C.) and School of Bioengineering, Dalian University of Technology, China (G.Y.)
| | - Lei Qian
- Advanced Institute for Medical Sciences, Dalian Medical University, China (S.J., R.G., J.W., L.Q., M.L., H.X., J.Z., Y.G., L.C.) and School of Bioengineering, Dalian University of Technology, China (G.Y.)
| | - Min Liu
- Advanced Institute for Medical Sciences, Dalian Medical University, China (S.J., R.G., J.W., L.Q., M.L., H.X., J.Z., Y.G., L.C.) and School of Bioengineering, Dalian University of Technology, China (G.Y.)
| | - Hu Xu
- Advanced Institute for Medical Sciences, Dalian Medical University, China (S.J., R.G., J.W., L.Q., M.L., H.X., J.Z., Y.G., L.C.) and School of Bioengineering, Dalian University of Technology, China (G.Y.)
| | - Jiayang Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, China (S.J., R.G., J.W., L.Q., M.L., H.X., J.Z., Y.G., L.C.) and School of Bioengineering, Dalian University of Technology, China (G.Y.)
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, China (S.J., R.G., J.W., L.Q., M.L., H.X., J.Z., Y.G., L.C.) and School of Bioengineering, Dalian University of Technology, China (G.Y.)
| | - Guangrui Yang
- Advanced Institute for Medical Sciences, Dalian Medical University, China (S.J., R.G., J.W., L.Q., M.L., H.X., J.Z., Y.G., L.C.) and School of Bioengineering, Dalian University of Technology, China (G.Y.)
| | - Lihong Chen
- Advanced Institute for Medical Sciences, Dalian Medical University, China (S.J., R.G., J.W., L.Q., M.L., H.X., J.Z., Y.G., L.C.) and School of Bioengineering, Dalian University of Technology, China (G.Y.)
| |
Collapse
|
14
|
Kulkarni P, Martson A, Vidya R, Chitnavis S, Harsulkar A. Pathophysiological landscape of osteoarthritis. Adv Clin Chem 2020; 100:37-90. [PMID: 33453867 DOI: 10.1016/bs.acc.2020.04.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A sharp rise in osteoarthritis (OA) incidence is expected as over 25% of world population ages in the coming decade. Although OA is considered a degenerative disease, mounting evidence suggests a strong connection with chronic metabolic conditions and low-grade inflammation. OA pathology is increasingly understood as a complex interplay of multiple pathological events including oxidative stress, synovitis and immune responses revealing its intricate nature. Cellular, biochemical and molecular aspects of these pathological events along with major outcomes of the relevant research studies in this area are discussed in the present review. With reference to their published and unpublished work, the authors strongly propose synovitis as a central OA pathology and the key OA pathological events are described in connection with it. Recent research outcomes also have succeeded to establish a linkage between metabolic syndrome and OA, which has been precisely included in the present review. Impact of aging process cannot be neglected in OA. Cell senescence is an important mechanism of aging through which it facilitates development of OA like other degenerative disorders, also discussed within a frame of OA. Conclusively, the reviewers urge low-grade inflammation linked to aging and derailed immune function as a pathological platform for OA development and progression. Thus, interventions targeted to prevent inflammaging hold a promising potential in effective OA management and efforts should be invested in this direction.
Collapse
Affiliation(s)
- Priya Kulkarni
- Department of Pathophysiology, Biomedicine and Translational medicine, University of Tartu, Tartu, Estonia; Department of Traumatology and Orthopaedics, Tartu University Hospital, Tartu, Estonia
| | - Aare Martson
- Department of Traumatology and Orthopaedics, Tartu University Hospital, Tartu, Estonia; Clinic of Traumatology and Orthopaedics, Tartu University Hospital, Tartu, Estonia
| | - Ragini Vidya
- Department of Pharmaceutical Biotechnology, Poona College of Pharmacy, Pune, India
| | - Shreya Chitnavis
- Department of Pharmaceutical Biotechnology, Poona College of Pharmacy, Pune, India
| | - Abhay Harsulkar
- Department of Pathophysiology, Biomedicine and Translational medicine, University of Tartu, Tartu, Estonia; Department of Pharmaceutical Biotechnology, Poona College of Pharmacy, Pune, India.
| |
Collapse
|
15
|
Dicer up-regulation by inhibition of specific proteolysis in differentiating monocytic cells. Proc Natl Acad Sci U S A 2020; 117:8573-8583. [PMID: 32220961 PMCID: PMC7165444 DOI: 10.1073/pnas.1916249117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Dicer is a ribonuclease III enzyme in biosynthesis of miRNAs, regulators of gene expression involved in macrophage differentiation. We found a specific truncation of Dicer in monocytic cells resulting from apparently constitutive cleavage by a serine protease. Inhibition of this proteolytic truncation, which occurred during macrophage differentiation in presence of TLR ligands or prostaglandin E2, up-regulates full-length Dicer and promotes miR biosynthesis. Regulation of transcription of pri-miRNA is one mode to regulate biosynthesis of mature miRNA. Inhibition of constitutive proteolysis of Dicer, as described here, provides a second layer of regulation, at the level of miRNA processing. Our data provide insights to Dicer and miRNAs in macrophage polarization/differentiation, a key process in the innate immune response. Dicer is a ribonuclease III enzyme in biosynthesis of micro-RNAs (miRNAs). Here we describe a regulation of Dicer expression in monocytic cells, based on proteolysis. In undifferentiated Mono Mac 6 (MM6) cells, full-length Dicer was undetectable; only an ∼50-kDa fragment appeared in Western blots. However, when MM6 cells were treated with zymosan or LPS during differentiation with TGF-β and 1,25diOHvitD3, full-length Dicer became abundant together with varying amounts of ∼170- and ∼50-kDa Dicer fragments. Mass spectrometry identified the Dicer fragments and showed cleavage about 450 residues upstream from the C terminus. Also, PGE2 (prostaglandin E2) added to differentiating MM6 cells up-regulated full-length Dicer, through EP2/EP4 and cAMP. The TLR stimuli strongly induced miR-146a-5p, while PGE2 increased miR-99a-5p and miR-125a-5p, both implicated in down-regulation of TNFα. The Ser protease inhibitor AEBSF (4-[2-aminoethyl] benzene sulfonyl fluoride) up-regulated full-length Dicer, both in MM6 cells and in primary human blood monocytes, indicating a specific proteolytic degradation. However, AEBSF alone did not lead to a general increase in miR expression, indicating that additional mechanisms are required to increase miRNA biosynthesis. Finally, differentiation of monocytes to macrophages with M-CSF or GM-CSF strongly up-regulated full-length Dicer. Our results suggest that differentiation regimens, both in the MM6 cell line and of peripheral blood monocytes, inhibit an apparently constitutive Dicer proteolysis, allowing for increased formation of miRNAs.
Collapse
|
16
|
Rapacz-Leonard A, Leonard M, Chmielewska-Krzesińska M, Siemieniuch M, Janowski TE. The oxytocin-prostaglandins pathways in the horse (Equus caballus) placenta during pregnancy, physiological parturition, and parturition with fetal membrane retention. Sci Rep 2020; 10:2089. [PMID: 32034259 PMCID: PMC7005845 DOI: 10.1038/s41598-020-59085-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/23/2020] [Indexed: 11/18/2022] Open
Abstract
Despite their importance in mammalian reproduction, substances in the oxytocin-prostaglandins pathways have not been investigated in the horse placenta during most of pregnancy and parturition. Therefore, we quantified placental content of oxytocin (OXT), oxytocin receptor (OXTR), and prostaglandin E2 and F2 alpha during days 90-240 of pregnancy (PREG), physiological parturition (PHYS), and parturition with fetal membrane retention (FMR) in heavy draft horses (PREG = 13, PHYS = 11, FMR = 10). We also quantified OXTR and prostaglandin endoperoxide synthase-2 (PTGS2) mRNA expression and determined the immunolocalization of OXT, OXTR, and PTGS2. For relative quantification of OXT and OXTR, we used western blotting with densitometry. To quantify the prostaglandins, we used enzyme immunoassays. For relative quantification of OXTR and PTGS2, we used RT-qPCR. For immunolocalization of OXT, OXTR, and PTGS2, we used immunohistochemistry. We found that OXT was present in cells of the allantochorion and endometrium in all groups. PTGS2 expression in the allantochorion was 14.7-fold lower in FMR than in PHYS (p = 0.007). These results suggest that OXT is synthesized in the horse placenta. As PTGS2 synthesis is induced by inflammation, they also suggest that FMR in heavy draft horses may be associated with dysregulation of inflammatory processes.
Collapse
Affiliation(s)
- Anna Rapacz-Leonard
- Department of Animal Reproduction with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland.
| | - Mark Leonard
- University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Małgorzata Chmielewska-Krzesińska
- Department of Pathophysiology, Forensic Veterinary and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Marta Siemieniuch
- Research Station in Popielno/Department of Immunology and Pathology of Reproduction, Polish Academy of Science, Olsztyn, Poland
| | - Tomasz E Janowski
- Department of Animal Reproduction with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
17
|
Guan PP, Liang YY, Cao LL, Yu X, Wang P. Cyclooxygenase-2 Induced the β-Amyloid Protein Deposition and Neuronal Apoptosis Via Upregulating the Synthesis of Prostaglandin E 2 and 15-Deoxy-Δ 12,14-prostaglandin J 2. Neurotherapeutics 2019; 16:1255-1268. [PMID: 31392591 PMCID: PMC6985346 DOI: 10.1007/s13311-019-00770-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Elevated levels of cyclooxygenase-2 (COX-2) and prostaglandins (PGs) have been shown to be involved in the pathogenesis of Alzheimer's disease. Analysis of the underlying mechanisms elucidated a function of sequential PGE2 and PGD2 synthesis in regulating β-amyloid protein (Aβ) deposition by modulating tumor necrosis factor α (TNF-α)-dependent presenilin (PS)1/2 activity in COX-2 and APP/PS1 crossed mice. Specifically, COX-2 overexpression accelerates the expression of microsomal PGE synthase-1 (mPGES-1) and lipocalin-type prostaglandin D synthase (L-PGDS), leading to the synthesis of PGE2 and 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) in 6-month-old APP/PS1 mice. Consequently, PGE2 has the ability to increase Aβ production by enhancing the expression of PS1/2 in a TNF-α-dependent manner, which accelerates the cognitive decline of COX-2/APP/PS1 mice. More interestingly, low concentrations of 15d-PGJ2 treatment facilitate the effects of PGE2 on the deposition of Aβ via TNF-α-dependent PS1/2 mechanisms. In contrast, high concentrations of 15d-PGJ2 treatment inhibit the deposition of Aβ via suppressing the expression of TNF-α-dependent PS1/2. In this regard, a high concentration of 15d-PGJ2 appears to be a therapeutic agent against Alzheimer's disease. However, the high 15d-PGJ2 concentration treatment induces neuronal apoptosis via increasing the protein levels of Bax, cleaved caspase-3, and DFF45, which further impairs the learning ability of APP/PS1 mice.
Collapse
Affiliation(s)
- Pei-Pei Guan
- College of Life and Health Sciences, Northeastern University, No. 3-11, Wenhua Road, Shenyang, 110819, China
| | - Yun-Yue Liang
- College of Life and Health Sciences, Northeastern University, No. 3-11, Wenhua Road, Shenyang, 110819, China
| | - Long-Long Cao
- College of Life and Health Sciences, Northeastern University, No. 3-11, Wenhua Road, Shenyang, 110819, China
| | - Xin Yu
- College of Life and Health Sciences, Northeastern University, No. 3-11, Wenhua Road, Shenyang, 110819, China
| | - Pu Wang
- College of Life and Health Sciences, Northeastern University, No. 3-11, Wenhua Road, Shenyang, 110819, China.
| |
Collapse
|
18
|
Li T, Liu B, Mao W, Gao R, Wu J, Deng Y, Shen Y, Liu K, Cao J. Prostaglandin E 2 promotes nitric oxide synthase 2, platelet-activating factor receptor, and matrix metalloproteinase-2 expression in Escherichia coli-challenged ex vivo endometrial explants via the prostaglandin E 2 receptor 4/protein kinase a signaling pathway. Theriogenology 2019; 134:65-73. [PMID: 31136957 DOI: 10.1016/j.theriogenology.2019.04.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/14/2019] [Accepted: 04/25/2019] [Indexed: 12/27/2022]
Abstract
Prostaglandin E2 (PGE2) is an inflammatory mediator involved in the pathogenesis of several chronic inflammatory conditions, including endometritis. Previous studies have shown that PGE2 accumulates in Escherichia coli-challenged ex vivo endometrial explants, increasing the expression of pro-inflammatory factors and aggravating tissue damage; these alterations are linked to key enzymes involved in the synthesis of PGE2, including cyclooxygenases-2 (COX-2) and microsomal PGES-1 (mPGES-1). In this study, we aimed to investigate whether administration of PGE2 modulated the activities of nitric oxide synthase 2 (NOS2), platelet-activating factor receptor (PAFR), and matrix metalloproteinase (MMP)-2 in E. coli-challenged ex vivo bovine endometrial explants. Our findings showed that COX-2 and mPGES-1 inhibitors significantly reduced NOS2, PAFR, and MMP-2 expression in the E. coli-challenged ex vivo endometrial explants. In addition, NOS2, PAFR, and MMP-2 expression levels were strongly increased in response to treatment with 15-prostaglandin dehydrogenase inhibitors in the E. coli-challenged ex vivo endometrial explants. However, these stimulatory effects could be blocked by PGE2 receptor 4 (EP4) and protein kinase A (PKA) inhibitors. Overall, these findings show that pathogenic PGE2 upregulated NOS2, PAFR, and MMP-2 expression, which may enhance inflammatory damage via the EP4/PKA signaling pathway in E. coli-challenged ex vivo endometrial explants.
Collapse
Affiliation(s)
- Tingting Li
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Bo Liu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Wei Mao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Ruifeng Gao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Jindi Wu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Yang Deng
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Yuan Shen
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Kun Liu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Jinshan Cao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Huhhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China.
| |
Collapse
|
19
|
Kumar S, Sharma SK, Kaushik G, Avti PK, Pandey SK, Sarma P, Medhi B, Khanduja KL. Therapeutic potential of arachidonyl trifluromethyl ketone, a cytosolic phospholipaseA 2 IVA specific inhibitor, in cigarette smoke condensate-induced pathological conditions in alveolar type I & II epithelial cells. Toxicol In Vitro 2019; 54:215-223. [PMID: 30253184 DOI: 10.1016/j.tiv.2018.09.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/07/2018] [Accepted: 09/19/2018] [Indexed: 11/18/2022]
Abstract
Cigarette smoke is responsible for multiple disorders and causes almost 10 million annual deaths globally but underlying mechanisms are still underexplored. Continuous exposure of Cigarette smoke condensate (CSC) leads to cytosolic phospholipase A2 (cPLA2) mediated high free radicals where cPLA2s seems to play crucial role in generated various patho-physiological conditions such as chronic inflammation, oxidative stress and cancer. In this view, we assessed the therapeutic potential of arachidonyl trifluromethyl ketone (ATK), a cPLA2 inhibitor, via pharmacological inhibition of most expressible CSC-induced cPLA2 group IVA in type-I and type-II alveolar epithelial cells. The In Vitro inhibitory effect of ATK on CSC-induced PLA2 activity and its cellular role were assessed in terms of cell viability, fluorescein diacetate (FDA) dye uptake assay for membrane integrity, reactive oxygen species (ROS)/reactive nitrogen species (RNS) levels and pro apoptotic as well as anti apoptosis markers via flow cytometry, along with extracellular signal-regulated kinases (ERK) levels using enzyme-linked immunosorbent assay (ELISA). The experimental findings demonstrated that ATK acts as potent inhibitor of cPLA2 activity and shown its effectiveness as therapeutic agent by significantly mimicking CSC-induced levels of free radicals, primary apoptosis, ratio of pro-apoptotic/apoptotic proteins and levels of ERK whereas protected cells from loss of cell viability and membrane integrity. Thus, this study is an important step towards the opening up of avenues for the applicability of the cPLA2 isoform specific inhibitors such as ATK for pre-clinical and clinical studies and could be beneficial during smoking-induced lung pathological conditions.
Collapse
Affiliation(s)
- Subodh Kumar
- Department of Biophysics, PGIMER, Chandigarh 160012, India.
| | - Sanjeev Kumar Sharma
- Department of Biophysics, PGIMER, Chandigarh 160012, India; Rajiv Gandhi Cancer Institute & Research Centre (RGCIRC), Rohini, New Delhi 110085, India
| | - Gaurav Kaushik
- Department of Biophysics, PGIMER, Chandigarh 160012, India; Surgery, School of Medicine, KU Medical Center (KUMC), Kansas City KS-66160, USA
| | | | - Satish Kumar Pandey
- Central Scientific Instruments Organisation (CSIO), Chandigarh 160030, India
| | - Phulen Sarma
- Present address: Department of Pharmacology, PGIMER, Chandigarh 160012, India
| | - Bikash Medhi
- Present address: Department of Pharmacology, PGIMER, Chandigarh 160012, India
| | | |
Collapse
|
20
|
Glutathionylation: a regulatory role of glutathione in physiological processes. Arh Hig Rada Toksikol 2018; 69:1-24. [DOI: 10.2478/aiht-2018-69-2966] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/01/2018] [Indexed: 12/18/2022] Open
Abstract
Abstract
Glutathione (γ-glutamyl-cysteinyl-glycine) is an intracellular thiol molecule and a potent antioxidant that participates in the toxic metabolism phase II biotransformation of xenobiotics. It can bind to a variety of proteins in a process known as glutathionylation. Protein glutathionylation is now recognised as one of important posttranslational regulatory mechanisms in cell and tissue physiology. Direct and indirect regulatory roles in physiological processes include glutathionylation of major transcriptional factors, eicosanoids, cytokines, and nitric oxide (NO). This review looks into these regulatory mechanisms through examples of glutathione regulation in apoptosis, vascularisation, metabolic processes, mitochondrial integrity, immune system, and neural physiology. The focus is on the physiological roles of glutathione beyond biotransformational metabolism.
Collapse
|
21
|
Marhuenda J, Medina S, Martínez-Hernández P, Arina S, Zafrilla P, Mulero J, Oger C, Galano JM, Durand T, Solana A, Ferreres F, López-García JJ, Gil-Izquierdo A. Effect of the dietary intake of melatonin- and hydroxytyrosol-rich wines by healthy female volunteers on the systemic lipidomic-related oxylipins. Food Funct 2018; 8:3745-3757. [PMID: 28956582 DOI: 10.1039/c7fo01081h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Oxylipins are lipid mediators involved in the physiopathology of all organs. Moreover, isoprostanes have been established as general and reliable in vivo oxidative stress biomarkers. Red wine has proved to exert several benefits through the maintenance of the oxidative balance of the organism. Antiradical scavenging capacity has been mainly attributed to polyphenols. However, melatonin and hydroxytyrosol should be taken into account as potent antiradical agents. The present research aimed to clarify the situation of enzymatic and oxidative injury and eicosanoid urinary excretion related to the intake of three kinds of red wines and their primary musts. Judging by the reduction in the excretion of isoprostanes, red wine consumption exhibited the highest antioxidant protection against oxidative stress, attributed to its OHTyr content (p < 0.05), and to a lesser extent to its MEL content. Similarly, the intake of red wine leads to the cardioprotective effect due to the reduction in the urinary excretion of the pro-inflammatory prostaglandin 2,3-dinor-11-β-PGF2α, besides the increase in the vasodilator prostaglandin PGE1, mediated by the melatonin (p < 0.05) and hydroxytyrosol (p < 0.05) contents. In conclusion, red wine (especially non-aged wine) exerts a higher in vivo antioxidant capacity than must or alcohol.
Collapse
Affiliation(s)
- Javier Marhuenda
- Research Group on Quality, Safety and Bioactivity of Plant Foods, Department of Food Science and Technology, CEBAS (CSIC), P.O. Box 164, 30100 Campus University Espinardo, Murcia, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Physiological and pathophysiological implications of PGE2 and the PGE2 synthases in the kidney. Prostaglandins Other Lipid Mediat 2018; 134:1-6. [DOI: 10.1016/j.prostaglandins.2017.10.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 10/09/2017] [Accepted: 10/31/2017] [Indexed: 12/16/2022]
|
23
|
Muthukaman N, Tambe M, Deshmukh S, Pisal D, Tondlekar S, Shaikh M, Sarode N, Kattige VG, Pisat M, Sawant P, Honnegowda S, Karande V, Kulkarni A, Behera D, Jadhav SB, Sangana RR, Gudi GS, Khairatkar-Joshi N, Gharat LA. Discovery of furan and dihydrofuran-fused tricyclic benzo[d]imidazole derivatives as potent and orally efficacious microsomal prostaglandin E synthase-1 (mPGES-1) inhibitors: Part-1. Bioorg Med Chem Lett 2017; 27:5131-5138. [PMID: 29100801 DOI: 10.1016/j.bmcl.2017.10.062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/18/2017] [Accepted: 10/25/2017] [Indexed: 01/28/2023]
Abstract
This letter describes the synthesis and biological evaluation of furan and dihydrofuran-fused tricyclic benzo[d]imidazole derivatives as novel mPGES-1 inhibitors, capable of inhibiting an increased PGE2 production in the disease state. Structure-activity optimization afforded many potent mPGES-1 inhibitors having <50 nM potencies in the A549 cellular assay and adequate metabolic stability in liver microsomes. Lead compounds 8l and 8m demonstrated reasonable in vitro pharmacology and pharmacokinetic properties over other compounds. In particular, 8m revealed satisfactory oral pharmacokinetics and bioavailability in multiple species like rat, guinea pig, dog and cynomolgus monkey. In addition, the representative compound 8m showed in vivo efficacy by inhibiting LPS-induced thermal hyperalgesia with an ED50 of 14.3 mg/kg in guinea pig.
Collapse
Affiliation(s)
- Nagarajan Muthukaman
- 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
| | - Sanjay Deshmukh
- 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
| | - Monali Pisat
- 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
| | - Srinivasa Honnegowda
- 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
| | - 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
|
24
|
Zhang Z, Li X, Huang H, Wang G, Qu Z, Zhang H. Cross-Coupling Effects of Silencing of Cyclooxygenase-2 (COX-2)/Aggrecanase-1 and Over-Expressed Insulin-Like Growth Factor 1 (IGF-1) in an Osteoarthritis Animal Model. Med Sci Monit 2017; 23:5302-5310. [PMID: 29112682 PMCID: PMC5687117 DOI: 10.12659/msm.907150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 10/19/2017] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND This study aimed to observe the effect of lentivirus-mediated cyclooxygenase-2 and aggrecanase-1 silencing and insulin-like growth factor-1 overexpression in human bone marrow mesenchymal stem cells after injection into model osteoarthritic knees. MATERIAL AND METHODS Using genetic recombination technique, the genes of cyclooxygenase-2, aggrecanase-1, and insulin-like growth factor-1 were recombined into the lentiviral vectors, and we transfected the human bone marrow stem cells in vitro. The BMSC transfected with lentivirus without genes served as a blank-virus group, and saline was used for another control group. One week later, the cytokines PGE2, aggrecanase-1, hIGF-1, and IL-1 were detected and compared between groups. RESULTS Compared with blank-virus group, the expression of COX-2 (85.81±5.12 ng/L) and aggrecanase1 (6.256±1.66) were decreased in the virus group (p<0.05), while the expression of hIGF-1 (17.46±1.86) was increased (p<0.05). The concentrations of PGE2 (85.81±5.12 ng/L), aggrecanase1 (51.34±5.463 ng/L), and IL-1 (82.31±4.321 ng/L) decreased (p<0.05) within the knee, but the concentration of hIGF-1 (44.33±0.7194 ng/L) increased (p<0.05). Compared with the other groups, the results of histological and immunohistochemical examinations demonstrated that the abrasion of articular cartilage was significantly improved and repaired. CONCLUSIONS Lentivirus-mediated RNAi can inhibit the expression of COX-2 mRNA and aggrecanase-1mRNA, and enhance the hIGF-1 mRNA expression, thereby influencing the concentration of cytokines in the early osteoarthritic model knee joints.
Collapse
Affiliation(s)
- Zhao Zhang
- Department of Hand and Foot Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Xiaofei Li
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Heng Huang
- Department of Hand and Foot Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Guozhong Wang
- Department of Hand and Foot Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Zhigang Qu
- Department of Hand and Foot Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Haining Zhang
- Department of Joint Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| |
Collapse
|
25
|
Yoshida K, Ito M, Hoshino Y, Matsuoka I. Effects of dexamethasone on purinergic signaling in murine mast cells: Selective suppression of P2X7 receptor expression. Biochem Biophys Res Commun 2017; 493:1587-1593. [PMID: 28988113 DOI: 10.1016/j.bbrc.2017.10.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 10/04/2017] [Indexed: 11/29/2022]
Abstract
Mast cells express many different purinergic receptors, including ionotropic P2X4 and P2X7, which recognize the accumulation of extracellular ATP released from activated and/or damaged cells. This results in the stimulation of mast cell functions. In this study, we investigated the effects of dexamethasone (Dex), an anti-inflammatory glucocorticoid widely used for the treatment of allergic disease, on purinergic receptor expression in mouse bone marrow-derived mast cells (BMMCs). Treatment of BMMCs with Dex decreased P2X7 receptor mRNA levels in a time- and concentration-dependent manner without affecting the expression of other purinergic receptor subtypes. Accordingly, fluorescence-activated cell sorting analysis revealed that Dex treatment also decreased P2X7 receptor protein levels. This effect was mimicked by prednisolone, another anti-inflammatory glucocorticoid, and was inhibited by the glucocorticoid receptor antagonist mifepristone. Functionally, treatment of BMMCs with Dex impaired the P2X7-mediated rise in intracellular Ca2+ concentration, degranulation, and ethidium uptake, a response relevant to receptor-pore formation. Finally, oral administration of Dex to C57BL/6 mice in vivo resulted in a significant decrease in P2X7 receptor expression in peritoneal mast cells. These results suggest that reduction of P2X7 receptor expression in mast cells might be one of the anti-allergic mechanisms of Dex.
Collapse
Affiliation(s)
- Kazuki Yoshida
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan
| | - Masaaki Ito
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan
| | - Yui Hoshino
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan
| | - Isao Matsuoka
- Laboratory of Pharmacology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki-shi, Gunma 370-0033, Japan.
| |
Collapse
|
26
|
Xia Z, Yan A. Computational models for the classification of mPGES-1 inhibitors with fingerprint descriptors. Mol Divers 2017; 21:661-675. [PMID: 28484935 DOI: 10.1007/s11030-017-9743-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 04/16/2017] [Indexed: 12/13/2022]
Abstract
Human microsomal prostaglandin [Formula: see text] synthase (mPGES)-1 is a promising drug target for inflammation and other diseases with inflammatory symptoms. In this work, we built classification models which were able to classify mPGES-1 inhibitors into two groups: highly active inhibitors and weakly active inhibitors. A dataset of 1910 mPGES-1 inhibitors was separated into a training set and a test set by two methods, by a Kohonen's self-organizing map or by random selection. The molecules were represented by different types of fingerprint descriptors including MACCS keys (MACCS), CDK fingerprints, Estate fingerprints, PubChem fingerprints, substructure fingerprints and 2D atom pairs fingerprint. First, we used a support vector machine (SVM) to build twelve models with six types of fingerprints and found that MACCS had some advantage over the other fingerprints in modeling. Next, we used naïve Bayes (NB), random forest (RF) and multilayer perceptron (MLP) methods to build six models with MACCS only and found that models using RF and MLP methods were better than NB. Finally, all the models with MACCS keys were used to make predictions on an external test set of 41 compounds. In summary, the models built with MACCS keys and using SVM, RF and MLP methods show good prediction performance on the test sets and the external test set. Furthermore, we made a structure-activity relationship analysis between mPGES-1 and its inhibitors based on the information gain of fingerprints and could pinpoint some key functional groups for mPGES-1 activity. It was found that highly active inhibitors usually contained an amide group, an aromatic ring or a nitrogen heterocyclic ring, and several heteroatoms substituents such as fluorine and chlorine. The carboxyl group and sulfur atom groups mainly appeared in weakly active inhibitors.
Collapse
Affiliation(s)
- Zhonghua Xia
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, P.O. Box 53, 15 BeiSanHuan East Road, Beijing, 100029, People's Republic of China
| | - Aixia Yan
- State Key Laboratory of Chemical Resource Engineering, Department of Pharmaceutical Engineering, Beijing University of Chemical Technology, P.O. Box 53, 15 BeiSanHuan East Road, Beijing, 100029, People's Republic of China.
| |
Collapse
|
27
|
Seo MJ, Oh DK. Prostaglandin synthases: Molecular characterization and involvement in prostaglandin biosynthesis. Prog Lipid Res 2017; 66:50-68. [DOI: 10.1016/j.plipres.2017.04.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 03/30/2017] [Accepted: 04/01/2017] [Indexed: 01/30/2023]
|
28
|
HARA S. Prostaglandin terminal synthases as novel therapeutic targets. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:703-723. [PMID: 29129850 PMCID: PMC5743848 DOI: 10.2183/pjab.93.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 07/21/2017] [Indexed: 06/07/2023]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) exert their anti-inflammatory and anti-tumor effects by reducing prostaglandin (PG) production via the inhibition of cyclooxygenase (COX). However, the gastrointestinal, renal and cardiovascular side effects associated with the pharmacological inhibition of the COX enzymes have focused renewed attention onto other potential targets for NSAIDs. PGH2, a COX metabolite, is converted to each PG species by species-specific PG terminal synthases. Because of their potential for more selective modulation of PG production, PG terminal synthases are now being investigated as a novel target for NSAIDs. In this review, I summarize the current understanding of PG terminal synthases, with a focus on microsomal PGE synthase-1 (mPGES-1) and PGI synthase (PGIS). mPGES-1 and PGIS cooperatively exacerbate inflammatory reactions but have opposing effects on carcinogenesis. mPGES-1 and PGIS are expected to be attractive alternatives to COX as therapeutic targets for several diseases, including inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Shuntaro HARA
- Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Tokyo, Japan
| |
Collapse
|
29
|
Wobst I, Ebert L, Birod K, Wegner MS, Hoffmann M, Thomas D, Angioni C, Parnham MJ, Steinhilber D, Tegeder I, Geisslinger G, Grösch S. R-Flurbiprofen Traps Prostaglandins within Cells by Inhibition of Multidrug Resistance-Associated Protein-4. Int J Mol Sci 2016; 18:ijms18010068. [PMID: 28042832 PMCID: PMC5297703 DOI: 10.3390/ijms18010068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 02/06/2023] Open
Abstract
R-flurbiprofen is the non-COX-inhibiting enantiomer of flurbiprofen and is not converted to S-flurbiprofen in human cells. Nevertheless, it reduces extracellular prostaglandin E2 (PGE2) in cancer or immune cell cultures and human extracellular fluid. Here, we show that R-flurbiprofen acts through a dual mechanism: (i) it inhibits the translocation of cPLA2α to the plasma membrane and thereby curtails the availability of arachidonic acid and (ii) R-flurbiprofen traps PGE2 inside of the cells by inhibiting multidrug resistance–associated protein 4 (MRP4, ABCC4), which acts as an outward transporter for prostaglandins. Consequently, the effects of R-flurbiprofen were mimicked by RNAi-mediated knockdown of MRP4. Our data show a novel mechanism by which R-flurbiprofen reduces extracellular PGs at physiological concentrations, particularly in cancers with high levels of MRP4, but the mechanism may also contribute to its anti-inflammatory and immune-modulating properties and suggests that it reduces PGs in a site- and context-dependent manner.
Collapse
Affiliation(s)
- Ivonne Wobst
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Lisa Ebert
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (L.E.); (M.J.P.)
| | - Kerstin Birod
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Marthe-Susanna Wegner
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Marika Hoffmann
- Institute of Pharmaceutical Chemistry, ZAFES, Johann Wolfgang Goethe-University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany;
| | - Dominique Thomas
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Carlo Angioni
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Michael J. Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (L.E.); (M.J.P.)
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, ZAFES, Johann Wolfgang Goethe-University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt, Germany;
| | - Irmgard Tegeder
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
| | - Gerd Geisslinger
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (L.E.); (M.J.P.)
| | - Sabine Grösch
- Pharmazentrum frankfurt, ZAFES, Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany; (I.W.); (K.B.); (M.-S.W.); (D.T.); (C.A.); (I.T.); (G.G.)
- Correspondence: ; Tel.: +49/69-6301-7820; Fax: +49/69-6301-7636
| |
Collapse
|
30
|
Ramanan M, Sinha S, Sudarshan K, Aidhen IS, Doble M. Inhibition of the enzymes in the leukotriene and prostaglandin pathways in inflammation by 3-aryl isocoumarins. Eur J Med Chem 2016; 124:428-434. [DOI: 10.1016/j.ejmech.2016.08.066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/09/2016] [Accepted: 08/29/2016] [Indexed: 12/31/2022]
|
31
|
Wrotek S, Domagalski K, Jędrzejewski T, Dec E, Kozak W. Buthionine sulfoximine, a glutathione depletor, attenuates endotoxic fever and reduces IL-1β and IL-6 level in rats. Cytokine 2016; 90:31-37. [PMID: 27764704 DOI: 10.1016/j.cyto.2016.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/12/2016] [Accepted: 10/12/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE The aim of our study was to investigate the effect of buthionine sulfoximine (BSO) - a glutathione depletor - on a course of endotoxic fever and IL-1β and IL-6 production. MATERIAL AND METHODS Male Wistar rats were subjected to intraperitoneal injection of lipopolysaccharide (LPS) from E. coli (50μg/kg, ip) to provoke fever. The level of spleen glutathione, plasma interleukin (IL)-1β, IL-6, and deep body temperature (Tb) were measured. RESULTS The LPS administration provoked fever (the average Tb was 38.14±0.05°C in NaCl/LPS-treated rats vs 37.10±0.03°C in control, not-treated rats; p<0.001). We observed that LPS injection induced a decrease in spleen glutathione level (7.67±0.92nM/g vs 13.27±0.47nM/g in not-treated rats; p<0.001). Furthermore, the injection of LPS provoked an elevation of plasma IL-1β and IL-6 concentration (from values below the lowest detectable standard in not-treated animals to 199.99±34.89pg/mL and 7500±542.21pg/mL, respectively; p<0.001). Pretreatment with BSO enhanced glutathione decrease in LPS-treated rats (5.05±0.49nM/g), and significantly affected fever (maximal Tb was 37.81±0.07°C in BSO/LPS-treated rats vs 38.76±0.11°C in NaCl/LPS-treated rats). BSO 4h after LPS injection decreased IL-1β and IL-6 gene expression (about 1.5 fold, and 2 fold, respectively). In a consequence we observed a decrease in plasma IL-6 concentration (4h after LPS injection plasma IL-6 was 4167.17±956.54pg/mL in BSO/LPS-treated rats vs 7500±542.21pg/mL in NaCl/LPS-treated rats; p<0.001), and later IL-1β (7h after LPS injection the IL-1β concentration was not detected). CONCLUSION Based on these data, we conclude that BSO, in addition to well-known application as an inhibitor of glutathione synthesis, is an antipyretic agent which reduces both IL-1β and IL-6 concentration.
Collapse
Affiliation(s)
- Sylwia Wrotek
- Department of Immunology, Nicolaus Copernicus University, Ul. Lwowska 1, 87-100 Torun, Poland.
| | - Krzysztof Domagalski
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Wilenska 4, 87-100 Torun, Poland.
| | - Tomasz Jędrzejewski
- Department of Immunology, Nicolaus Copernicus University, Ul. Lwowska 1, 87-100 Torun, Poland.
| | - Eliza Dec
- Department of Immunology, Nicolaus Copernicus University, Ul. Lwowska 1, 87-100 Torun, Poland.
| | - Wiesław Kozak
- Department of Immunology, Nicolaus Copernicus University, Ul. Lwowska 1, 87-100 Torun, Poland.
| |
Collapse
|
32
|
Arosh JA, Banu SK, McCracken JA. Novel concepts on the role of prostaglandins on luteal maintenance and maternal recognition and establishment of pregnancy in ruminants. J Dairy Sci 2016; 99:5926-5940. [PMID: 27179861 DOI: 10.3168/jds.2015-10335] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/03/2016] [Indexed: 11/19/2022]
Abstract
In ruminants, the corpus luteum (CL) of early pregnancy is resistant to luteolysis. Prostaglandin (PG)E2 is considered a luteoprotective mediator. Early studies indicate that during maternal recognition of pregnancy (MRP) in ruminants, a factor(s) from the conceptus or gravid uterus reaches the ovary locally through the utero-ovarian plexus (UOP) and protects the CL from luteolysis. The local nature of the embryonic antiluteolytic or luteoprotective effect precludes any direct effect of a protein transported or acting between the gravid uterus and CL in ruminants. During MRP, interferon tau (IFNT) secreted by the trophoblast of the conceptus inhibits endometrial pulsatile release of PGF2α and increases endometrial PGE2. Our recent studies indicate that (1) luteal PG biosynthesis is selectively directed toward PGF2α at the time of luteolysis and toward PGE2 at the time of establishment of pregnancy (ESP); (2) the ability of the CL of early pregnancy to resist luteolysis is likely due to increased intraluteal biosynthesis and signaling of PGE2; and (3) endometrial PGE2 is transported from the uterus to the CL through the UOP vascular route during ESP in sheep. Intrauterine co-administration of IFNT and prostaglandin E2 synthase 1 (PGES-1) inhibitor reestablishes endometrial PGF2α pulses and regresses the CL. In contrast, intrauterine co-administration of IFNT and PGES-1 inhibitor along with intraovarian administration of PGE2 rescues the CL. Together, the accumulating information provides compelling evidence that PGE2 produced by the CL in response to endometrial PGE2 induced by pregnancy may counteract the luteolytic effect of PGF2α as an additional luteoprotective mechanism during MRP or ESP in ruminants. Targeting PGE2 biosynthesis and signaling selectively in the endometrium or CL may provide luteoprotective therapy to improve reproductive efficiency in ruminants.
Collapse
Affiliation(s)
- Joe A Arosh
- Reproductive Endocrinology and Cell Signaling Laboratory, Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station 77483.
| | - Sakhila K Banu
- Reproductive Endocrinology and Cell Signaling Laboratory, Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station 77483
| | - John A McCracken
- Department of Animal Science, University of Connecticut, Storrs 06269
| |
Collapse
|
33
|
Chandrasekhar S, Harvey AK, Yu XP, Chambers MG, Oskins JL, Lin C, Seng TW, Thibodeaux SJ, Norman BH, Hughes NE, Schiffler MA, Fisher MJ. Identification and Characterization of Novel Microsomal Prostaglandin E Synthase-1 Inhibitors for Analgesia. J Pharmacol Exp Ther 2016; 356:635-44. [PMID: 26740668 DOI: 10.1124/jpet.115.228932] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/05/2016] [Indexed: 03/08/2025] Open
Abstract
Prostaglandin (PG) E2 plays a critical role in eliciting inflammation. Nonsteroidal anti-inflammatory drugs and selective inhibitors of cyclooxygenase, which block PGE2 production, have been used as key agents in treating inflammation and pain associated with arthritis and other conditions. However, these agents have significant side effects such as gastrointestinal bleeding and myocardial infarction, since they also block the production of prostanoids that are critical for other normal physiologic functions. Microsomal prostaglandin E2 synthase-1 is a membrane-bound terminal enzyme in the prostanoid pathway, which acts downstream of cyclooxygenase 2 and is responsible for PGE2 production during inflammation. Thus, inhibition of this enzyme would be expected to block PGE2 production without inhibiting other prostanoids and would provide analgesic efficacy without the side effects. In this report, we describe novel microsomal prostaglandin E2 synthase-1 inhibitors that are potent in blocking PGE2 production and are efficacious in a guinea pig monoiodoacetate model of arthralgia. These molecules may be useful in treating the signs and symptoms associated with arthritis.
Collapse
Affiliation(s)
| | - Anita K Harvey
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Xiao-Peng Yu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Mark G Chambers
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Jennifer L Oskins
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Chaohua Lin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Thomas W Seng
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | | | - Bryan H Norman
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Norman E Hughes
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | | | - Matthew J Fisher
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| |
Collapse
|
34
|
Koeberle A, Laufer SA, Werz O. Design and Development of Microsomal Prostaglandin E2 Synthase-1 Inhibitors: Challenges and Future Directions. J Med Chem 2016; 59:5970-86. [DOI: 10.1021/acs.jmedchem.5b01750] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Andreas Koeberle
- Chair
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, University Jena, Philosophenweg 14, 07743 Jena, Germany
| | - Stefan A. Laufer
- Department
of Pharmaceutical Chemistry, Pharmaceutical Institute, University of Tuebingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Oliver Werz
- Chair
of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, University Jena, Philosophenweg 14, 07743 Jena, Germany
| |
Collapse
|
35
|
Highly specific and sensitive immunoassay for the measurement of prostaglandin E2 in biological fluids. Bioanalysis 2015; 7:2597-607. [PMID: 26457411 DOI: 10.4155/bio.15.130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Lack of specificity of anti-PGE2 antibodies is a long-standing problem. Given quite a few analogs and low PGE2 content in biological fluids, it is quite important to simultaneously meet the demands of high specificity and sensitivity. RESULTS Highly specific anti-PGE2 antibodies were obtained by combined use of cationic carrier protein and Mannich reaction. The cross-reactivity values of the resultant polyclonal and monoclonal antibodies against eight analogs were <14 and <5%, respectively. Furthermore, we established a highly sensitive ELISA, which could be applied to direct analysis of PGE2 at the pg/ml level (LOQ = 15.6 pg/ml). CONCLUSION We provide an appropriate strategy to develop a highly-specific and sensitive immunoassay for measuring low PGE2 content in biological samples.
Collapse
|
36
|
Wrotek S, Jędrzejewski T, Nowakowska A, Kozak W. Glutathione deficiency attenuates endotoxic fever in rats. Int J Hyperthermia 2015; 31:793-9. [DOI: 10.3109/02656736.2015.1067333] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
37
|
de Hair MJH, Leclerc P, Newsum EC, Maijer KI, van de Sande MGH, Ramwadhdoebe TH, van Schaardenburg D, van Baarsen LGM, Korotkova M, Gerlag DM, Tak PP, Jakobsson PJ. Expression of Prostaglandin E2 Enzymes in the Synovium of Arthralgia Patients at Risk of Developing Rheumatoid Arthritis and in Early Arthritis Patients. PLoS One 2015. [PMID: 26225917 PMCID: PMC4520525 DOI: 10.1371/journal.pone.0133669] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Objective Arthralgia may precede the development of synovial inflammation in autoantibody-positive individuals at risk of developing rheumatoid arthritis (RA). A major pathway involved in pain is the prostaglandin (PG) E2 pathway. We investigated this pathway in the synovium of individuals with RA-specific autoantibodies and in early arthritis patients. Methods Nineteen autoantibody-positive individuals (IgM-rheumatoid factor and/or anti-cyclic citrullinated peptide antibodies) with arthralgia (n=15) and/or a positive family history of RA (n=8), who had been prospectively followed for at least 2 years, were included. In addition, we included early arthritis patients (disease-modifying antirheumatic drug naïve) who after 2 years follow up fulfilled classification criteria for RA (n=63), spondyloarthritis (SpA; n=14), or had unclassified arthritis (UA; n=27). In all subjects we assessed pain and performed synovial biopsy sampling by mini-arthroscopy at baseline. Tissue sections were examined by immunohistochemistry to detect and quantify PGE2 pathway enzymes expression levels (mPGES-1; COX-1 and -2; 15-PGDH). Results In both study groups synovial expression of PGE2 enzymes was not clearly related to pain sensation. Expression levels at baseline were not associated with the development of arthritis after follow up (6 out of 19 autoantibody-positive individuals). However, in early SpA patients the expression levels of mPGES-1 and COX-1 were significantly increased compared to RA and UA patients. Conclusion Pain in autoantibody-positive individuals without synovial inflammation who are at risk of developing RA and in early arthritis patients may be regulated by pathways other than the PGE2 pathway or originate at sites other than the synovium. In contrast, in SpA, the PGE2 pathway may be inherently linked to the pathophysiology/etiology of the disease.
Collapse
Affiliation(s)
- Maria J. H. de Hair
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Patrick Leclerc
- Rheumatology research Unit, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Elize C. Newsum
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Karen I. Maijer
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Marleen G. H. van de Sande
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Tamara H. Ramwadhdoebe
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | | | - Lisa G. M. van Baarsen
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Marina Korotkova
- Rheumatology research Unit, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Danielle M. Gerlag
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
| | - Paul-Peter Tak
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, the Netherlands
- * E-mail:
| | - Per-Johan Jakobsson
- Rheumatology research Unit, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
38
|
Koeberle A, Werz O. Perspective of microsomal prostaglandin E2 synthase-1 as drug target in inflammation-related disorders. Biochem Pharmacol 2015; 98:1-15. [PMID: 26123522 DOI: 10.1016/j.bcp.2015.06.022] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 06/23/2015] [Indexed: 02/07/2023]
Abstract
Prostaglandin (PG)E2 encompasses crucial roles in pain, fever, inflammation and diseases with inflammatory component, such as cancer, but is also essential for gastric, renal, cardiovascular and immune homeostasis. Cyclooxygenases (COX) convert arachidonic acid to the intermediate PGH2 which is isomerized to PGE2 by at least three different PGE2 synthases. Inhibitors of COX - non-steroidal anti-inflammatory drugs (NSAIDs) - are currently the only available therapeutics that target PGE2 biosynthesis. Due to adverse effects of COX inhibitors on the cardiovascular system (COX-2-selective), stomach and kidney (COX-1/2-unselective), novel pharmacological strategies are in demand. The inducible microsomal PGE2 synthase (mPGES)-1 is considered mainly responsible for the excessive PGE2 synthesis during inflammation and was suggested as promising drug target for suppressing PGE2 biosynthesis. However, 15 years after intensive research on the biology and pharmacology of mPGES-1, the therapeutic value of mPGES-1 as drug target is still vague and mPGES-1 inhibitors did not enter the market so far. This commentary will first shed light on the structure, mechanism and regulation of mPGES-1 and will then discuss its biological function and the consequence of its inhibition for the dynamic network of eicosanoids. Moreover, we (i) present current strategies for interfering with mPGES-1-mediated PGE2 synthesis, (ii) summarize bioanalytical approaches for mPGES-1 drug discovery and (iii) describe preclinical test systems for the characterization of mPGES-1 inhibitors. The pharmacological potential of selective mPGES-1 inhibitor classes as well as dual mPGES-1/5-lipoxygenase inhibitors is reviewed and pitfalls in their development, including species discrepancies and loss of in vivo activity, are discussed.
Collapse
Affiliation(s)
- Andreas Koeberle
- Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany.
| | - Oliver Werz
- Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, 07743 Jena, Germany.
| |
Collapse
|
39
|
Guillem-Llobat P, Íñiguez MA. Inhibition of lipopolysaccharide-induced gene expression by liver X receptor ligands in macrophages involves interference with early growth response factor 1. Prostaglandins Leukot Essent Fatty Acids 2015; 96:37-49. [PMID: 25736222 DOI: 10.1016/j.plefa.2015.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/09/2015] [Accepted: 02/10/2015] [Indexed: 01/08/2023]
Abstract
Liver X receptors (LXRs) are nuclear receptors that act as ligand-dependent transcription factors forming permissive heterodimers with retinoid X receptors (RXRs). In this study we aimed to assess the effect of LXR/RXR activation on the transcriptional induction of pro-inflammatory genes including cyclooxygenase-2 (COX-2) and microsomal prostaglandin E2 synthase-1 (mPGES-1) in activated macrophages. Our study shows that LXR ligands such as oxysterols, GW3965 or TO901317, as well as RXR ligands like 9cis retinoic acid or SR11237, decreased LPS-induced expression of COX-2 and mPGES-1. Consequently, LPS-dependent PGE2 production was substantially reduced in macrophages treated with LXR/RXR ligands. The inhibitory effects of LXR/RXR activation on LPS-induced expression of COX-2 and mPGES-1 in macrophages, occurred by a mechanism involving interference with transcriptional activation of these genes. LXR/RXR activation interfered with the activity of transcription factors essential in the up-regulation of the expression of pro-inflammatory genes in these cells, such as NFκB, but also Egr-1, which had not been previously associated with LXR-mediated gene repression. As this transcription factor is involved in the regulation of a variety of genes involved in inflammatory processes, LXR and RXR-mediated interference with Egr-1 signaling could represent an important event mediating the anti-inflammatory effects of these receptors in macrophages.
Collapse
Affiliation(s)
- Paloma Guillem-Llobat
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, Cantoblanco, 28049 Madrid, Spain
| | - Miguel A Íñiguez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular, Instituto de Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
40
|
Chen Y, Liu H, Xu S, Wang T, Li W. Targeting microsomal prostaglandin E2synthase-1 (mPGES-1): the development of inhibitors as an alternative to non-steroidal anti-inflammatory drugs (NSAIDs). MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00278h] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AA cascade and several key residues in the 3D structure of mPGES-1.
Collapse
Affiliation(s)
- Yuqing Chen
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | | | - Shuang Xu
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | - Tianlin Wang
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| |
Collapse
|
41
|
Prevention of oxidative stress, inflammation and mitochondrial dysfunction in the intestine by different cranberry phenolic fractions. Clin Sci (Lond) 2014; 128:197-212. [PMID: 25069567 DOI: 10.1042/cs20140210] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cranberry fruit has been reported to have high antioxidant effectiveness that is potentially linked to its richness in diversified polyphenolic content. The aim of the present study was to determine the role of cranberry polyphenolic fractions in oxidative stress (OxS), inflammation and mitochondrial functions using intestinal Caco-2/15 cells. The combination of HPLC and UltraPerformance LC®-tandem quadrupole (UPLC-TQD) techniques allowed us to characterize the profile of low, medium and high molecular mass polyphenolic compounds in cranberry extracts. The medium molecular mass fraction was enriched with flavonoids and procyanidin dimers whereas procyanidin oligomers (DP > 4) were the dominant class of polyphenols in the high molecular mass fraction. Pre-incubation of Caco-2/15 cells with these cranberry extracts prevented iron/ascorbate-mediated lipid peroxidation and counteracted lipopolysaccharide-mediated inflammation as evidenced by the decrease in pro-inflammatory cytokines (TNF-α and interleukin-6), cyclo-oxygenase-2 and prostaglandin E2. Cranberry polyphenols (CP) fractions limited both nuclear factor κB activation and Nrf2 down-regulation. Consistently, cranberry procyanidins alleviated OxS-dependent mitochondrial dysfunctions as shown by the rise in ATP production and the up-regulation of Bcl-2, as well as the decline of protein expression of cytochrome c and apoptotic-inducing factor. These mitochondrial effects were associated with a significant stimulation of peroxisome-proliferator-activated receptor γ co-activator-1-α, a central inducing factor of mitochondrial biogenesis and transcriptional co-activator of numerous downstream mediators. Finally, cranberry procyanidins forestalled the effect of iron/ascorbate on the protein expression of mitochondrial transcription factors (mtTFA, mtTFB1, mtTFB2). Our findings provide evidence for the capacity of CP to reduce intestinal OxS and inflammation while improving mitochondrial dysfunction.
Collapse
|
42
|
Abstract
The PGE2 pathway is important in inflammation-driven diseases and specific targeting of the inducible mPGES-1 is warranted due to the cardiovascular problems associated with the long-term use of COX-2 inhibitors. This review focuses on patents issued on methods of measuring mPGES-1 activity, on drugs targeting mPGES-1 and on other modulators of free extracellular PGE2 concentration. Perspectives and conclusions regarding the status of these drugs are also presented. Importantly, no selective inhibitors targeting mPGES-1 have been identified and, despite the high number of published patents, none of these drugs have yet made it to clinical trials.
Collapse
|
43
|
Ahmad W, Kumolosasi E, Jantan I, Bukhari SNA, Jasamai M. Effects of Novel Diarylpentanoid Analogues of Curcumin on Secretory Phospholipase A2, Cyclooxygenases, Lipo-oxygenase, and Microsomal Prostaglandin E Synthase-1. Chem Biol Drug Des 2014; 83:670-81. [DOI: 10.1111/cbdd.12280] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 11/23/2013] [Accepted: 01/06/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Waqas Ahmad
- Drug and Herbal Research Centre; Faculty of Pharmacy; Universiti Kebangsaan Malaysia; Jalan Raja Muda Abdul Aziz 50300 Kuala Lumpur Malaysia
| | - Endang Kumolosasi
- Drug and Herbal Research Centre; Faculty of Pharmacy; Universiti Kebangsaan Malaysia; Jalan Raja Muda Abdul Aziz 50300 Kuala Lumpur Malaysia
| | - Ibrahim Jantan
- Drug and Herbal Research Centre; Faculty of Pharmacy; Universiti Kebangsaan Malaysia; Jalan Raja Muda Abdul Aziz 50300 Kuala Lumpur Malaysia
| | - Syed N. A. Bukhari
- Drug and Herbal Research Centre; Faculty of Pharmacy; Universiti Kebangsaan Malaysia; Jalan Raja Muda Abdul Aziz 50300 Kuala Lumpur Malaysia
| | - Malina Jasamai
- Drug and Herbal Research Centre; Faculty of Pharmacy; Universiti Kebangsaan Malaysia; Jalan Raja Muda Abdul Aziz 50300 Kuala Lumpur Malaysia
| |
Collapse
|
44
|
Korotkova M, Jakobsson PJ. Persisting eicosanoid pathways in rheumatic diseases. Nat Rev Rheumatol 2014; 10:229-41. [DOI: 10.1038/nrrheum.2014.1] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
45
|
Singh Bahia M, Kumar Katare Y, Silakari O, Vyas B, Silakari P. Inhibitors of Microsomal Prostaglandin E2
Synthase-1 Enzyme as Emerging Anti-Inflammatory Candidates. Med Res Rev 2014; 34:825-55. [DOI: 10.1002/med.21306] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Malkeet Singh Bahia
- Molecular Modelling Lab (MML); Department of Pharmaceutical Sciences and Drug Research; Punjabi University; Patiala Punjab 147002 India
| | - Yogesh Kumar Katare
- Radharaman Institute of Pharmaceutical Sciences; Bhopal Madhya Pradesh 462046 India
| | - Om Silakari
- Molecular Modelling Lab (MML); Department of Pharmaceutical Sciences and Drug Research; Punjabi University; Patiala Punjab 147002 India
| | - Bhawna Vyas
- Department of Chemistry; Punjabi University; Patiala Punjab 147002 India
| | - Pragati Silakari
- Adina institute of Pharmaceutical Sciences; Sagar Madhya Pradesh (M.P.) 470001 India
| |
Collapse
|
46
|
Hanke T, Rörsch F, Thieme TM, Ferreiros N, Schneider G, Geisslinger G, Proschak E, Grösch S, Schubert-Zsilavecz M. Synthesis and pharmacological characterization of benzenesulfonamides as dual species inhibitors of human and murine mPGES-1. Bioorg Med Chem 2013; 21:7874-83. [PMID: 24183739 DOI: 10.1016/j.bmc.2013.10.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 10/01/2013] [Accepted: 10/07/2013] [Indexed: 02/04/2023]
Abstract
The microsomal prostaglandin E2 synthase 1 (mPGES-1) became a desirable target in recent years for the research of new anti-inflammatory drugs. Even though many potent inhibitors of human mPGES-1, tested in vitro assay systems, have been synthesized, they all failed in preclinical trials in rodent models of inflammation, due to the lack of activity on rodent enzyme. Within this work we want to present a new class of mPGES-1 inhibitors derived from a benzenesulfonamide scaffold with inhibitory potency on human and murine mPGES-1. Starting point with an IC50 of 13.8 μM on human mPGES-1 was compound 1 (4-{benzyl[(4-methoxyphenyl)methyl]sulfamoyl}benzoic acid; FR4), which was discovered by a virtual screening approach. Optimization during a structure-activity relationship (SAR) process leads to compound 28 (4-[(cyclohexylmethyl)[(4-phenylphenyl)methyl]sulfamoyl]benzoic acid) with an improved IC50 of 0.8 μM on human mPGES-1. For the most promising compounds a broad pharmacological characterization has been carried out to estimate their anti-inflammatory potential.
Collapse
Affiliation(s)
- Thomas Hanke
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt, Max-von-Laue-Str. 9, D-60438 Frankfurt am Main, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Korotkova M, Jakobsson PJ. Characterization of Microsomal Prostaglandin E Synthase 1 Inhibitors. Basic Clin Pharmacol Toxicol 2013; 114:64-9. [DOI: 10.1111/bcpt.12162] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 09/19/2013] [Indexed: 01/22/2023]
Affiliation(s)
- Marina Korotkova
- Rheumatology Unit; Department of Medicine; Karolinska Institutet; Stockholm Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit; Department of Medicine; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
48
|
Engström Ruud L, Wilhelms DB, Eskilsson A, Vasilache AM, Elander L, Engblom D, Blomqvist A. Acetaminophen reduces lipopolysaccharide-induced fever by inhibiting cyclooxygenase-2. Neuropharmacology 2013; 71:124-9. [DOI: 10.1016/j.neuropharm.2013.03.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 03/11/2013] [Accepted: 03/17/2013] [Indexed: 02/02/2023]
|
49
|
Leclerc P, Wähämaa H, Idborg H, Jakobsson PJ, Harris HE, Korotkova M. IL-1β/HMGB1 complexes promote The PGE2 biosynthesis pathway in synovial fibroblasts. Scand J Immunol 2013; 77:350-60. [PMID: 23488692 PMCID: PMC3670302 DOI: 10.1111/sji.12041] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/15/2013] [Indexed: 01/09/2023]
Abstract
PGE2 is a potent lipid mediator of pain and oedema found elevated in RA. Microsomal prostaglandin E synthase-1 (mPGES-1) is a terminal enzyme of the PGE2 pathway inducible by proinflammatory cytokines. mPGES-1 is markedly upregulated in RA synovial tissue despite antirheumatic treatments, suggesting that multiple inflammatory stimuli contribute to its induction. High-mobility group box chromosomal protein 1 (HMGB1) is known to induce inflammation both by direct interaction with TLR4 and by enhancement of other proinflammatory molecules signalling, through complex formation. The high expression of extracellular HMGB1 within the inflamed synovium, implies its pro-arthritogenic role in RA. We aimed to investigate the effects of IL-1β/HMGB1 complexes on mPGES-1 and other enzymes of the PGE2 pathway in synovial fibroblasts (SFs) from patients with arthritis. Furthermore, we studied the effect of COX-2 inhibition and IL-1RI antagonism on prostanoid and cytokine production by SFs. Stimulation of SFs with HMGB1 in complex with suboptimal amounts of IL-1β significantly increased mPGES-1 and COX-2 expressions as well as PGE2 production, as compared to treatment with HMGB1 or IL-1β alone. Furthermore, NS-398 reduced the production of IL-6 and IL-8, thus indicating that IL-1β/HMGB1 complexes modulate cytokine production in part through prostanoid synthesis. Treatment with IL-1RA completely abolished the induced PGE2 and cytokine production, suggesting an effect mediated through IL-1RI. IL-1β/HMGB1 complexes promote the induction of mPGES-1, COX-2 and PGE2 in SF. The amplification of the PGE2 biosynthesis pathway by HMGB1 might constitute an important pathogenic mechanism perpetuating inflammatory and destructive activities in rheumatoid arthritis.
Collapse
Affiliation(s)
- P Leclerc
- Rheumatology Research Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
50
|
Kahnt AS, Rörsch F, Diehl O, Hofmann B, Lehmann C, Steinbrink SD, Angioni C, Geisslinger G, Grösch S, Steinhilber D, Maier TJ. Cysteinyl leukotriene-receptor-1 antagonists interfere with PGE2 synthesis by inhibiting mPGES-1 activity. Biochem Pharmacol 2013; 86:286-96. [PMID: 23684692 DOI: 10.1016/j.bcp.2013.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 04/07/2013] [Accepted: 05/07/2013] [Indexed: 01/28/2023]
Abstract
Because of their favourable safety profile and beneficial anti-inflammatory properties, the CysLT1 receptor antagonists (LTRA), montelukast, zafirlukast and pranlukast are approved for the treatment of asthma and are frequently prescribed as add-on therapeutics to reduce the amount of inhaled glucocorticoids and β2-agonists. There is evidence that some of these anti-inflammatory properties might be of a secondary nature and therefore, unrelated to the CysLT1 antagonism. Here, we show that LTRA inhibit PGE2 formation in cytokine-stimulated Hela and A549 carcinoma cells and in lipopolysaccharide (LPS)-stimulated human leukocyte preparations (IC50∼20μM). Neither expression of enzymes involved in PGE2 synthesis nor arachidonic acid release and COX activities were inhibited by the compounds. In contrast, mPGES-1 activity was suppressed at low micromolar levels (IC50 between 2 and 4μM). This suppression was specific for PGE2 synthesis, since PGD2 and PGI2 levels in LPS-stimulated leukocyte preparations were not negatively affected. PGF2α levels were concomitantly inhibited, probably due to its direct synthesis from PGE2. Several major conclusions can be drawn from this study: (A) clinical trials investigating elevated doses of the compounds are helpful to confirm suppression of PGE2 synthesis in vivo; (B) studies investigating the role of CysLTs in cell culture or animal models of inflammation and cancer have to be reassessed carefully, if higher doses of LTRA were applied or serum levels in cell culture assays were low; and (C) LTRA may serve as new scaffolds for the development of potent, selective and well tolerated mPGES-1 inhibitors.
Collapse
Affiliation(s)
- Astrid Stefanie Kahnt
- Goethe-University, Institute of Pharmaceutical Chemistry, ZAFES, Max-von-Laue-Str. 9, D-60438 Frankfurt/Main, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|