1
|
Önal HT. Anti-inflammatory Photodynamic Activity of Tetraphenyl-Substituted Porphyrin in J774.2 Macrophage Cells. Photodiagnosis Photodyn Ther 2025:104597. [PMID: 40254229 DOI: 10.1016/j.pdpdt.2025.104597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 04/22/2025]
Abstract
Porphyrin derivatives have been explored as potential agents for Photodynamic Therapy (PDT). This study aimed to investigate the anti-inflammatory photodynamic activity of a symmetrical tetraphenyl-substituted porphyrin derivative in mammalian macrophage cells. In this study, the previously synthesized compound 5,10,15,20- tetra(4-tert-butylphenyl)porphyrin (POR) was evaluated. To investigate the potential effects of POR on the immune response, interleukin-6 (IL-6) and tumor necrosis factoralpha (TNF-α) levels were measured in cells stimulated with lipopolysaccharide (LPS). Cytokine analyses were performed under conditions where the compound was applied in the dark and under 5 and 10 minute light exposure. The data show that the porphyrin derivative exhibits a significant anti-inflammatory photodynamic effect in vitro when applied at subtoxic concentrations. No cytotoxicity was observed at all applied doses, both in the presence and absence of light. This is consistent with previous literature findings demonstrating the non-cytotoxic nature of porphyrin derivatives. While high-concentration POR applications create a strong anti-inflammatory response in dark conditions, a significant decrease in TNF-α and IL-6 release was recorded with light application. Furthermore, the observed effects were dose-dependent, highlighting the critical importance of optimizing the dose for potential therapeutic uses. The results indicate that the studied porphyrin derivative has photodynamic anti-inflammatory properties and suggest that this compound can be evaluated as a candidate therapeutic agent in the management of diseases where suppression of the immune response is targeted.
Collapse
Affiliation(s)
- Harika Topal Önal
- Medical Laboratory Techniques, Vocational School of Health Services, Toros University, 33140 Mersin, Turkiye.
| |
Collapse
|
2
|
Fang S, Huang X, Cai F, Qiu G, Lin F, Cai X. Design, synthesis and molecular docking of novel D-ring substituted steroidal 4,5-dihydropyrazole thiazole derivatives that act as iNOS/COX-2 inhibitors with potent anti-inflammatory activity against LPS-induced RAW264.7 macrophage cells. J Steroid Biochem Mol Biol 2024; 240:106478. [PMID: 38430971 DOI: 10.1016/j.jsbmb.2024.106478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 03/05/2024]
Abstract
Inflammation, an important biological protective response to tissue damage or microbial invasion, is considered to be an alarming signal for the progress of varied biological complications. Based on the previous reports in the literature that proved the noticeable efficacy of pyrazole and thiazole scaffold as well as nitrogen heterocyclic based compounds against acute and chronic inflammatory disease, a new set of novel D-ring substituted steroidal 4,5-dihydropyrazole thiazole derivatives were synthesized and evaluated their anti-inflammatory activities in vitro. Preliminary structure-activity relationship (SAR) analysis was conducted by their inhibitory activities against nitric oxide (NO) release in lipopolysaccharide (LPS)-induced RAW 264.7 cells, and the optimal compound 12b [3β-hydroxy-pregn-5-en-17β-yl-5'- (o- chlorophenyl)- 1'-(4''- phenyl -[1'', 3'']- thiazol-2''- yl) - 4',5'-dihydro - 1'H-pyrazol - 3'- yl] exhibited more potent anti-inflammatory activity than the positive control treatment methylprednisolone (MPS), with an IC50 value of 2.59 μM on NO production and low cytotoxicity against RAW 264.7 cells. In further mechanism study, our results showed that compound 12b significantly suppressed the production of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and inhibited the expressions of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) through blocking NF-κB p65 nuclear translocation and phosphorylation of IκBα. Compound 12b also attenuated LPS-induced activation of c-Jun amino-terminal kinase (JNK) and p38 phosphorylation in RAW 264.7 cells. Molecular docking study revealed the strong binding affinity of compound 12b to the active site of the COX-2 proteins, which confirmed that compound 12b acted as an anti-inflammatory mediator. These results indicate that steroidal derivatives bearing 4,5-dihydropyrazole thiazole structure might be considered for further research and scaffold optimization in designing anti-inflammatory drugs and compound 12b might be a promising therapeutic anti-inflammatory drug candidate.
Collapse
Affiliation(s)
- Shuopo Fang
- Department of Pharmacy, The Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xiaodan Huang
- Department of Digestive Medical Oncology, The Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Fen Cai
- Department of Nosocomial Infection Management, The Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Guodong Qiu
- Department of Pharmacy, The Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Fei Lin
- Department of Pharmacy Intravenous Admixture Services (PIVAS), The Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China.
| | - Xiaorui Cai
- Department of Pharmacy, The Affiliated Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong, China.
| |
Collapse
|
3
|
Hakli Ö, Yarali S, Öner Usta E, Ayaz F. Photodynamic anti-inflammatory activity of meso‑aryl substituted porphyrin derivative on mammalian macrophages. Photodiagnosis Photodyn Ther 2024; 45:103922. [PMID: 38081569 DOI: 10.1016/j.pdpdt.2023.103922] [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: 11/08/2023] [Revised: 11/28/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Our group focused on a meso‑aryl substituted porphyrin molecule for its photodynamic anti-inflammatory activities on the mammalian macrophages. MATERIALS AND METHODS The porphyrine derivative previously synthesized in this study was synthesized and characterized by 1H NMR. We then examined their immunomodulatory activities based on the changes in the pro-inflammatory cytokine production levels after LPS stimulation in dark and light activated conditions. RESULTS Our results suggest that porphyrin derivative had anti-inflammatory photodynamic activity in vitro at subtoxic concentrations. Our study aims to pave a way for anti-inflammatory photodynamic therapy application in the inflammatory and autoimmune disorders. Most of the studies either focus on photodynamic cytotoxicity of the porphyrin derivatives to suppress the inflammation or porphyrin derivatives' anti-inflammatory activity without the photodynamic activation. CONCLUSION Our future studies will focus on the generation and in vitro as well as in vivo characterization of the porphyrin derivatives with anti-inflammatory photodynamic therapy applications. In this way, novel drug candidates that would have lower side effects can be generated for the patients.
Collapse
Affiliation(s)
- Özgül Hakli
- Chemistry Department, Faculty of Science, Muğla Sıtkı Koçman University, Kötekli, Muğla 48000, Turkey.
| | - Sümeyye Yarali
- Chemistry Department, Faculty of Science, Muğla Sıtkı Koçman University, Kötekli, Muğla 48000, Turkey
| | - Ebru Öner Usta
- Biotechnology Department, Faculty of Science, Mersin University, Yenişehir, Mersin, Turkey
| | - Furkan Ayaz
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Biruni University, Istanbul 34010, Turkey.
| |
Collapse
|
4
|
Shen ZB, Meng HW, Meng XS, Lv ZK, Fang MY, Zhang LL, Lv ZL, Li MS, Liu AK, Han JH, Li QS, Duan YJ. Design, synthesis, and SAR study of novel flavone 1,2,4-oxadiazole derivatives with anti-inflammatory activities for the treatment of Parkinson's disease. Eur J Med Chem 2023; 255:115417. [PMID: 37137246 DOI: 10.1016/j.ejmech.2023.115417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/07/2023] [Accepted: 04/24/2023] [Indexed: 05/05/2023]
Abstract
Inflammation is one of a major feature of Parkinson's disease (PD) which poses a threat to people's health in the world. It has been reported that antioxidation and anti-inflammation have significant effects on the treatment of PD. 1,2,4-oxadiazole and flavone derivatives have remarkable antioxidant and anti-inflammatory activities. In order to find highly effective drugs for PD treatment, based on the remarkable anti-inflammatory and antioxidant activities of the 1,2,4-oxadiazole pharmacophore and the flavonoid pharmacophore, we designed and synthesized a novel series of 3-methyl-8-(3-methyl-1,2,4-oxadiazol-5-yl)-2-phenyl-4H-chromen-4-one derivatives by pharmacophore combination, and evaluated their anti-inflammatory and antioxidation activities for PD treatment. Preliminary structure-activity relationship (SAR) analysis was conducted by their inhibitory activities against reactive oxygen species (ROS) and NO release in LPS-induced BV2 Microglia cells, and the optimal compound Flo8 exhibited the most potent anti-inflammatory and antioxidant activities. Both in vivo and in vitro results showed that Flo8 inhibited neuronal apoptosis by inhibiting inflammatory and apoptotic signaling pathways. In vivo studies also showed that the compound Flo8 ameliorated motor and behavioral deficits and increased serum dopamine levels in MPTP-induced PD model mice. Taken together, this study demonstrated the compound Flo8 could be a promising agent for the treatment of PD.
Collapse
Affiliation(s)
- Zhen-Bao Shen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Hua-Wen Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xian-She Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ze-Kun Lv
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Meng-Yuan Fang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Lang-Lang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Zhi-Lin Lv
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Mu-Sen Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - An-Kang Liu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ji-Hong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Qing-Shan Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Ya-Jun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China; Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
5
|
Silva LP, Santos IP, Silva DKC, dos Reis BPZC, Meira CS, Castro MVBDS, dos Santos Filho JM, de Araujo-Neto JH, Ellena JA, da Silveira RG, Soares MBP. Molecular Hybridization Strategy on the Design, Synthesis, and Structural Characterization of Ferrocene- N-acyl Hydrazones as Immunomodulatory Agents. Molecules 2022; 27:molecules27238343. [PMID: 36500436 PMCID: PMC9737981 DOI: 10.3390/molecules27238343] [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: 09/26/2022] [Revised: 11/08/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022] Open
Abstract
Immunomodulatory agents are widely used for the treatment of immune-mediated diseases, but the range of side effects of the available drugs makes necessary the search for new immunomodulatory drugs. Here, we investigated the immunomodulatory activity of new ferrocenyl-N-acyl hydrazones derivatives (SintMed(141−156). The evaluated N-acyl hydrazones did not show cytotoxicity at the tested concentrations, presenting CC50 values greater than 50 µM. In addition, all ferrocenyl-N-acyl hydrazones modulated nitrite production in immortalized macrophages, showing inhibition values between 14.4% and 74.2%. By presenting a better activity profile, the ferrocenyl-N-acyl hydrazones SintMed149 and SintMed150 also had their cytotoxicity and anti-inflammatory effect evaluated in cultures of peritoneal macrophages. The molecules were not cytotoxic at any of the concentrations tested in peritoneal macrophages and were able to significantly reduce (p < 0.05) the production of nitrite, TNF-α, and IL-1β. Interestingly, both molecules significantly reduced the production of IL-2 and IFN-γ in cultured splenocytes activated with concanavalin A. Moreover, SintMed150 did not show signs of acute toxicity in animals treated with 50 or 100 mg/kg. Finally, we observed that ferrocenyl-N-acyl hydrazone SintMed150 at 100 mg/kg reduced the migration of neutrophils (44.6%) in an acute peritonitis model and increased animal survival by 20% in an LPS-induced endotoxic shock model. These findings suggest that such compounds have therapeutic potential to be used to treat diseases of inflammatory origin.
Collapse
Affiliation(s)
- Laís Peres Silva
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil
| | - Ivanilson Pimenta Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
| | | | | | - Cássio Santana Meira
- Department of Life Sciences, State University of Bahia (UNEB), Salvador 41150-000, BA, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
- Institute for Innovation in Advanced Health Systems (CIMATEC ISI SAS—University Center SENAI/CIMATEC), Salvador 41650-010, BA, Brazil
| | - Marcos Venícius Batista de Souza Castro
- Laboratory of Design and Synthesis Applied to Medicinal Chemistry-SintMed®, Center for Technology and Geosciences, Federal University of Pernambuco, Recife 50740-521, PE, Brazil
| | - José Maurício dos Santos Filho
- Laboratory of Design and Synthesis Applied to Medicinal Chemistry-SintMed®, Center for Technology and Geosciences, Federal University of Pernambuco, Recife 50740-521, PE, Brazil
| | - João Honorato de Araujo-Neto
- Multiuser Laboratory of Structural Crystallography, Institute of São Carlos, University of São Paulo, São Carlos 13566-590, SP, Brazil
| | - Javier Alcides Ellena
- Multiuser Laboratory of Structural Crystallography, Institute of São Carlos, University of São Paulo, São Carlos 13566-590, SP, Brazil
| | - Rafael Gomes da Silveira
- Multiuser Laboratory of Structural Crystallography, Institute of São Carlos, University of São Paulo, São Carlos 13566-590, SP, Brazil
- Department of Chemistry, Federal Institute of Goiás, Campus Ceres, Ceres 76300-000, GO, Brazil
| | - Milena Botelho Pereira Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil
- Institute for Innovation in Advanced Health Systems (CIMATEC ISI SAS—University Center SENAI/CIMATEC), Salvador 41650-010, BA, Brazil
- Correspondence:
| |
Collapse
|
6
|
Mahore A, Kamboj P, Kaleem M, Amir M. Therapeutic management of arthritis: A review on structural and target‐based approaches. Arch Pharm (Weinheim) 2022; 355:e2200182. [DOI: 10.1002/ardp.202200182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Anjali Mahore
- Department of Pharmaceutical Chemistry School of Pharmaceutical Education and Research New Delhi India
| | - Payal Kamboj
- Department of Pharmaceutical Chemistry School of Pharmaceutical Education and Research New Delhi India
| | - Mohammad Kaleem
- Department of Pharmaceutical Chemistry School of Pharmaceutical Education and Research New Delhi India
| | - Mohammad Amir
- Department of Pharmaceutical Chemistry School of Pharmaceutical Education and Research New Delhi India
| |
Collapse
|
7
|
Veloso C, Videira RA, Andrade PB, Cardoso C, Vitorino C. In vivo methodologies to assist preclinical development of topical fixed-dose combinations for pain management. Int J Pharm 2022; 616:121530. [PMID: 35121043 DOI: 10.1016/j.ijpharm.2022.121530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 11/24/2022]
Abstract
The combination in a fixed dose of two or more active pharmaceutical ingredients in the same pharmaceutical dosage form is an approach that has been used successfully in the treatment of several pathologies, including pain. In the preclinical development of a topical fixed-dose combination product with analgesic and anti-inflammatory activities for pain management, the main objective is to establish the nature of the interaction between the different active pharmaceutical ingredients while obtaining data on the medicinal product safety and efficacy. Despite the improvement of in vitro assays, animal models remain a fundamental strategy to characterise the interaction, efficacy and safety of active pharmaceutical ingredients at the physiological level, which cannot be reached by in vitro assays. Thus, the main goal of this review is to systematise the available animal models to evaluate the efficacy and safety of a new fixed-dose combination product for topical administration indicated for pain management. Particular emphasis is given to animal models that are accepted for regulatory purposes.
Collapse
Affiliation(s)
- Cláudia Veloso
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, Rua Larga, 3004-535 Coimbra, Portugal
| | - Romeu A Videira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal.
| | - Paula B Andrade
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal
| | - Catarina Cardoso
- Laboratórios Basi, Parque Industrial Manuel Lourenço Ferreira, lote 15, 3450-232 Mortágua, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, Rua Larga, 3004-535 Coimbra, Portugal.
| |
Collapse
|
8
|
Zhang Z, Cao P, Fang M, Zou T, Han J, Duan Y, Xu H, Yang X, Li QS. Design, synthesis, and SAR study of novel 4,5-dihydropyrazole-Thiazole derivatives with anti-inflammatory activities for the treatment of sepsis. Eur J Med Chem 2021; 225:113743. [PMID: 34403978 DOI: 10.1016/j.ejmech.2021.113743] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 12/24/2022]
Abstract
Systemic inflammatory response syndrome is a major feature of sepsis which is one of the major causes of death worldwide. It has been reported that 3,5-diaryl-4,5-dihydropyrazole and thiazole derivatives have many biological functions, especially in the aspect of anti-inflammation. According to the strategy of pharmacophore combination, we introduced thiazole moiety into dihydropyrazole skeleton to design and synthesize a novel series of 2-(3,5-diphenyl-4,5-dihydro-1H-pyrazol-1-yl)-4-methylthiazole derivatives, and evaluated their anti-inflammatory activities for sepsis treatment. Preliminary structure-activity relationship (SAR) analysis was conducted by their inhibitory activities against nitric oxide (NO) release in LPS-induced RAW264.7 cells, and the optimal compound E26 exhibited more potent anti-inflammatory activity than the positive control treatment indomethacin and dexamethasone. In further mechanism study, our results showed that compound E26 significantly suppressed the production of interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), NO and inhibited the expressions of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) through blocking MAPKs signaling pathway. In addition, in vivo administration of compound E26 resulted in a significant improvement of LPS-induced sepsis in C57BL/6J mice, with reducing toxicity in multiple organs. Taken together, this study demonstrated the compound E26 could be a promising agent for the treatment of sepsis.
Collapse
Affiliation(s)
- Zhen Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Peichang Cao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Mengyuan Fang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Tingfeng Zou
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Huajian Xu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| | - Qing-Shan Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.
| |
Collapse
|
9
|
Miao Y, Yang J, Yun Y, Sun J, Wang X. Synthesis and anti-rheumatoid arthritis activities of 3-(4-aminophenyl)-coumarin derivatives. J Enzyme Inhib Med Chem 2021; 36:450-461. [PMID: 33557646 PMCID: PMC7889190 DOI: 10.1080/14756366.2021.1873978] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Rheumatoid arthritis is a chronic systemic disease characterised by an unknown aetiology of inflammatory synovitis. A large number of studies have shown that synoviocytes show tumour-like dysplasia in the pathological process of RA, and the changes in the expression of related cytokines are closely related to the pathogenesis of RA. In this thesis, a series of novel 3-(4-aminophenyl) coumarins containing different substituents were synthesised to find new coumarin anti-inflammatory drugs for the treatment of rheumatoid arthritis. The results of preliminary activity screening showed that compound 5e had the strongest inhibitory activity on the proliferation of fibroid synovial cells, and it also had inhibitory effect on RA-related cytokines IL-1, IL-6, and TNF-α. The preliminary mechanism study showed that compound 5e could inhibit the activation of NF-κB and MAPKs signal pathway. The anti-inflammatory activity of compound 5ein vivo was further determined in the rat joint inflammation model.
Collapse
Affiliation(s)
- Yuhang Miao
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Yang
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yinling Yun
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Sun
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaojing Wang
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
10
|
Chen LZ, Shu HY, Wu J, Yu YL, Ma D, Huang X, Liu MM, Liu XH, Shi JB. Discovery and development of novel pyrimidine and pyrazolo/thieno-fused pyrimidine derivatives as potent and orally active inducible nitric oxide synthase dimerization inhibitor with efficacy for arthritis. Eur J Med Chem 2021; 213:113174. [PMID: 33515864 DOI: 10.1016/j.ejmech.2021.113174] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/24/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022]
Abstract
In order to discover and develop drug-like anti-inflammatory agents against arthritis, based on "Hit" we found earlier and to overcome drawbacks of toxicity, twelve series of total 89 novel pyrimidine, pyrazolo[4,3-d]pyrimidine and thieno[3,2-d]pyrimidine derivatives were designed, synthesized and screened for their anti-inflammatory activity against NO and toxicity for normal liver cells (LO2). Relationships of balance toxicity and activity have been summarized through multi-steps, and title compounds 22o, 22l were found to show lower toxicity (against LO2: IC50 = 2934, 2301 μM, respectively) and potent effect against NO release (IR = 98.3, 97.67%, at 10 μM, respectively). Furthermore, compound 22o showed potent iNOS inhibitory activity with value of IC50 is 0.96 μM and could interfere stability and formation of the active dimeric iNOS. It's anti-inflammatory activity in vivo was assessed by AIA rat model. Furthermore, the results of metabolic stability, CYP, PK study in vivo, acute toxicity study and subacute toxicity assessment indicated this compound had good drug-like properties for treatment.
Collapse
Affiliation(s)
- Liu Zeng Chen
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Hai Yang Shu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Jing Wu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Yun Long Yu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Duo Ma
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Xin Huang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Ming Ming Liu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China
| | - Xin Hua Liu
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China.
| | - Jing Bo Shi
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, PR China.
| |
Collapse
|
11
|
Zhao W, Zhang X, Zhang R, Zhang K, Li Y, Xu FJ. Self-Assembled Herbal Medicine Encapsulated by an Oxidation-Sensitive Supramolecular Hydrogel for Chronic Wound Treatment. ACS APPLIED MATERIALS & INTERFACES 2020; 12:56898-56907. [PMID: 33296174 DOI: 10.1021/acsami.0c19492] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Inflammation has been assumed to affect the pathology of wound healing and is associated with many nonhealing chronic wounds. Naturally derived herbal medicines with anti-inflammatory properties are of interest because of their effectiveness and affordability in clinical treatment. Herein, we report a supramolecular hydrogel comprising self-assembled natural herb rhein and an oxidative responsive cross-linked network based on ferrocene and β-cyclodextrin host-guest recognitions. Rhein can directly self-assemble into fibrils, exerting better anti-inflammation efficiency than its free drug form. The adaption of the supramolecular network can greatly improve the stability and retain the structural integrity of encapsulated self-assembled rhein. In addition, host-guest recognition confers dissolution of the hydrogel under oxidative stress, thereby delivering self-assembled rhein to the wound site and exerting better therapeutic efficiency. Evaluations in diabetic mice indicate that the resultant hydrogel promoted chronic wound healing by suppressing excess reactive oxygen species, facilitating the transition of the wound healing process, and restoring the normal wound-repair process. Therefore, the proposed hydrogel has a potential value as an herbal-based dressing for future clinical chronic wound management.
Collapse
Affiliation(s)
- Weiyi Zhao
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiang Zhang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Rui Zhang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Kai Zhang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yang Li
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
12
|
1,4-Disubstituted 1 H-1,2,3-Triazoles for Renal Diseases: Studies of Viability, Anti-Inflammatory, and Antioxidant Activities. Int J Mol Sci 2020; 21:ijms21113823. [PMID: 32481556 PMCID: PMC7312092 DOI: 10.3390/ijms21113823] [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] [Received: 03/29/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a hallmark of many metabolic diseases. We previously showed that ferrocene-appended 1H-1,2,3-triazole hybrids inhibit nitric oxide (NO) production in in vitro models of lipopolysaccharide-induced inflammation in the BV-2 cell. In the present study, we explored the viability, anti-inflammatory, and antioxidant potential of ferrocene-1H-1,2,3-triazole hybrids using biochemical assays in rat mesangial cells (RMCs). We found that, among all the ferrocene-1H-1,2,3-triazole hybrids, X2-X4 exhibited an antioxidant effect on mitochondrial free radicals. Among all the studied compounds, X4 demonstrated the best anti-inflammatory effect on RMCs. These results were supplemented by in silico studies including molecular docking with human cytosolic phospholipase A2 (cPLA2) and cyclooxygenase 2 (COX-2) enzymes as well as absorption, distribution, metabolism, excretion, and toxicity (ADMET) profiling. Besides, two new crystal structures of the compounds have also been reported. In addition, combining the results from the inducible nitric oxide synthase (iNOS), cPLA2, COX-2, and matrix metalloproteinase-9 (MMP-9) enzymatic activity analysis and NO production also confirmed this argument. Overall, the results of this study will be a valuable addition to the growing body of work on biological activities of triazole-based compounds.
Collapse
|