1
|
Yang Q, Liu G, Chen G, Chen G, Chen K, Fan L, Tu Y, Chen J, Shi Z, Chen C, Liu S, Deng G, Deng X, Sun C, Li X, Yang S, Zheng S, Chen B. Novel injectable adhesive hydrogel loaded with exosomes for holistic repair of hemophilic articular cartilage defect. Bioact Mater 2024; 42:85-111. [PMID: 39280582 PMCID: PMC11399810 DOI: 10.1016/j.bioactmat.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Hemophilic articular cartilage damage presents a significant challenge for surgeons, characterized by recurrent intraarticular bleeding, a severe inflammatory microenvironment, and limited self-repair capability of cartilage tissue. Currently, there is a lack of tissue engineering-based integrated therapies that address both early hemostasis, anti-inflammation, and long-lasting chondrogenesis for hemophilic articular cartilage defects. Herein, we developed an adhesive hydrogel using oxidized chondroitin sulfate and gelatin, loaded with exosomes derived from bone marrow stem cells (BMSCs) (Hydrogel-Exos). This hydrogel demonstrated favorable injectability, self-healing, biocompatibility, biodegradability, swelling, frictional and mechanical properties, providing a comprehensive approach to treating hemophilic articular cartilage defects. The adhesive hydrogel, featuring dynamic Schiff base bonds and hydrogen bonds, exhibited excellent wet tissue adhesiveness and hemostatic properties. In a pig model, the hydrogel could be smoothly injected into the knee joint cartilage defect site and gelled in situ under fluid-irrigated arthroscopic conditions. Our in vitro and in vivo experiments confirmed that the sustained release of exosomes yielded anti-inflammatory effects by modulating macrophage M2 polarization through the NF-κB pathway. This immunoregulatory effect, coupled with the extracellular matrix components provided by the adhesive hydrogel, enhanced chondrogenesis, promoted the cartilage repair and joint function restoration after hemophilic articular cartilage defects. In conclusion, our results highlight the significant application potential of Hydrogel-Exos for early hemostasis, immunoregulation, and long-term chondrogenesis in hemophilic patients with cartilage injuries. This innovative approach is well-suited for application during arthroscopic procedures, offering a promising solution for addressing the complex challenges associated with hemophilic articular cartilage damage.
Collapse
Affiliation(s)
- Qinfeng Yang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Guihua Liu
- Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou, Guangdong, 516008, China
| | - Guanghao Chen
- Department of Orthopaedics, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, 314000, China
| | - Guo Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Keyu Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Lei Fan
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yuesheng Tu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jialan Chen
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhanjun Shi
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chuan Chen
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Shubo Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Geyang Deng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xiaoqian Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, 510060, China
| | - Chunhan Sun
- Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou, Guangdong, 516008, China
| | - Xiaoyang Li
- Department of Vascular Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, 324000, China
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Shaowei Zheng
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
- State Key Laboratory of Quality Research in Chinese Medicines, Laboratory of Drug Discovery from Natural Resources and Industrialization, School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China
| | - Bin Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
2
|
Nongthombam GS, Ahmed SA, Saikia K, Gogoi S, Borah JC. Breaking boundaries in diabetic nephropathy treatment: design and synthesis of novel steroidal SGLT2 inhibitors. RSC Med Chem 2024:d4md00645c. [PMID: 39479473 PMCID: PMC11514366 DOI: 10.1039/d4md00645c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/13/2024] [Indexed: 11/02/2024] Open
Abstract
The activity of sodium glucose co-transporter 2 (SGLT2) has always been an important parameter influencing chronic kidney disease in type-2 diabetic patients. Herein, we have meticulously designed, synthesized, and evaluated several novel steroidal pyrimidine molecules that possess the capability to successfully bind to the SGLT2 protein and inhibit its activity, thereby remedying kidney-related ailments in diabetic patients. The lead steroidal pyrimidine compounds were selected after virtually screening from a library of probable N-heterocyclic steroidal scaffolds. A nano-catalyzed synthetic route was also explored for the synthesis of the steroidal pyrimidine analogs demonstrating an environmentally benign protocol. Extensive in vitro investigations encompassing SGLT2 screening assays and cell viability assessments were conducted on the synthesized compounds. Among the steroidal pyrimidine derivatives evaluated, compound 9a exhibited the highest SGLT2 inhibition activity and underwent further scrutiny. Western blot analysis was employed to determine the impact of 9a on inflammatory and fibrotic proteins, aiming to elucidate its mechanism of action. Additionally, in silico analyses were performed to illuminate the structural dynamics and molecular interaction mechanism of 9a. The overall investigation is crucial for advancing the development of the next generation of anti-diabetic drugs.
Collapse
Affiliation(s)
- Geetmani Singh Nongthombam
- Chemical Biology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology Guwahati-781035 Assam India
| | - Semim Akhtar Ahmed
- Chemical Biology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology Guwahati-781035 Assam India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 India
| | - Kangkon Saikia
- Chemical Biology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology Guwahati-781035 Assam India
| | - Sanjib Gogoi
- Applied Organic Chemistry, Chemical Sciences & Technology Division, CSIR-North East Institute of Science and Technology Jorhat 785006 India
| | - Jagat Chandra Borah
- Chemical Biology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology Guwahati-781035 Assam India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 India
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Guwahati 781101 Assam India
| |
Collapse
|
3
|
Lee SJ, Pak SW, Kim WI, Park SH, Cho YK, Ko JW, Kim TW, Kim JS, Kim JC, Lim JO, Shin IS. Silibinin Suppresses Inflammatory Responses Induced by Exposure to Asian Sand Dust. Antioxidants (Basel) 2024; 13:1187. [PMID: 39456441 PMCID: PMC11505622 DOI: 10.3390/antiox13101187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Asian sand dust (ASD), generated from the deserts of China and Mongolia, affects Korea and Japan during spring and autumn, causing harmful effects on various bio-organs, including the respiratory system, due to its irritants such as fine dust, chemicals, and toxic materials. Here, we investigated the therapeutic effects of silibinin against ASD-induced airway inflammation using mouse macrophage-like cell line RAW264.7 and a murine model. ASD was intranasally administered to mice three times a week and silibinin was administered for 6 days by oral gavage. In ASD-stimulated RAW264.7 cells, silibinin treatment decreased tumor necrosis factor-α production and reduced the expression of p-p65NF-κB, p-p38, and cyclooxygenase (COX)-2, while increasing heme oxygenase (HO)-1 expression. In ASD-exposed mice, silibinin administration reduced inflammatory cell count and cytokines in bronchoalveolar lavage fluid and decreased inflammatory cell infiltration in lung tissue. Additionally, silibinin lowered oxidative stress, as evidenced by decreased 8-hydroxy-2'-deoxyguanosin (8-OHdG) expression and increased HO-1 expression. The expression of inflammatory-related proteins, including p-p65NF-κB, COX-2, and p-p38, was markedly reduced by silibinin administration. Overall, silibinin treatment reduced the expression of p-p65NF-κB, COX-2, and p-p38 in response to ASD exposure, while increasing HO-1 expression both in vitro and in vivo. These findings suggest that silibinin mitigates pulmonary inflammation caused by ASD exposure by reducing inflammatory signaling and oxidative stress, indicating its potential as a therapeutic agent for ASD-induced pulmonary inflammation.
Collapse
Affiliation(s)
- Se-Jin Lee
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - So-Won Pak
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - Woong-Il Kim
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - Sin-Hyang Park
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - Young-Kwon Cho
- College of Health Sciences, Cheongju University, 298 Daesung-ro, Sangdang-gu, Cheongju-si 28503, Republic of Korea;
| | - Je-Won Ko
- BK21 FOUR Program, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Daejeon 34134, Republic of Korea; (J.-W.K.); (T.-W.K.)
| | - Tae-Won Kim
- BK21 FOUR Program, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Daejeon 34134, Republic of Korea; (J.-W.K.); (T.-W.K.)
| | - Joong-Sun Kim
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - Jong-Choon Kim
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - Je-Oh Lim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, 177 Geonjae-ro, Naju-si 58245, Republic of Korea
| | - In-Sik Shin
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| |
Collapse
|
4
|
Begh MZA, Khan J, Al Amin M, Sweilam SH, Dharmamoorthy G, Gupta JK, Sangeetha J, Lokeshvar R, Nafady MH, Ahmad I, Alshehri MA, Emran TB. Monoterpenoid synergy: a new frontier in biological applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03342-x. [PMID: 39105799 DOI: 10.1007/s00210-024-03342-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 07/28/2024] [Indexed: 08/07/2024]
Abstract
Monoterpenoids, compounds found in various organisms, have diverse applications in various industries. Their effectiveness is influenced by the oil's chemical composition, which in turn is influenced by plant genotype, environmental conditions, cultivation practices, and plant development stage. They are used in various industries due to their distinctive odor and taste, serving as ingredients, additives, insecticides, and repellents. These compounds have synergistic properties, resulting in superior combined effects over discrete ones, potentially beneficial for various health purposes. Many experimental studies have investigated their interactions with other ingredients and their antibacterial, insecticidal, antifungal, anticancer, anti-inflammatory, and antioxidant properties. This review discusses potential synergistic interactions between monoterpenoids and other compounds, their sources, and biological functions. It also emphasizes the urgent need for more research on their bioavailability and toxicity, underlining the importance and relevance of this comprehensive study in the current scientific landscape.
Collapse
Affiliation(s)
- Md Zamshed Alam Begh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh.
| | - Jishan Khan
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chittagong, 4318, Bangladesh
| | - Md Al Amin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| | - G Dharmamoorthy
- Department of Pharmaceutical Analysis, MB School of Pharmaceutical Sciences, Mohan Babu University (Erstwhile Sree Vidyaniketan College of Pharmacy), Tirupati, India
| | - Jeetendra Kumar Gupta
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India
| | - J Sangeetha
- Department of Pharmacognosy, Malla Reddy Institute of Pharmaceutical Sciences, Maisammaguda, Dhulapally, 500100, India
| | - R Lokeshvar
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Saveetha Nagar, Thandalam, Chennai, India
| | - Mohamed H Nafady
- Faculty of Applied Health Science Technology, Misr University for Science and Technology, Giza, 12568, Egypt
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, 61421, Saudi Arabia
| | - Mohammed Ali Alshehri
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh.
| |
Collapse
|
5
|
Quesada-Vázquez S, Eseberri I, Les F, Pérez-Matute P, Herranz-López M, Atgié C, Lopez-Yus M, Aranaz P, Oteo JA, Escoté X, Lorente-Cebrian S, Roche E, Courtois A, López V, Portillo MP, Milagro FI, Carpéné C. Polyphenols and metabolism: from present knowledge to future challenges. J Physiol Biochem 2024; 80:603-625. [PMID: 39377969 PMCID: PMC11502541 DOI: 10.1007/s13105-024-01046-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 08/31/2024] [Indexed: 10/25/2024]
Abstract
A diet rich in polyphenols and other types of phytonutrients can reduce the occurrence of chronic diseases. However, a well-established cause-and-effect association has not been clearly demonstrated and several other issues will need to be fully understood before general recommendations will be carried out In the present review, some of the future challenges that the research on phenolic compounds will have to face in the next years are discussed: toxicological aspects of polyphenols and safety risk assessment; synergistic effects between different polyphenols; metabotype-based nutritional advice based on a differential gut microbial metabolism of polyphenols (precision nutrition); combination of polyphenols with other bioactive compounds; innovative formulations to improve the bioavailability of phenolic compounds; and polyphenols in sports nutrition and recovery.Other aspects related to polyphenol research that will have a boost in the next years are: polyphenol and gut microbiota crosstalk, including prebiotic effects and biotransformation of phenolic compounds into bioactive metabolites by gut microorganisms; molecular docking, molecular dynamics simulation, and quantum and molecular mechanics studies on the protein-polyphenol complexes; and polyphenol-based coating films, nanoparticles, and hydrogels to facilitate the delivery of drugs, nucleic acids and proteins.In summary, this article provides some constructive inspirations for advancing in the research of the applications, risk assessment and metabolic effects of dietary polyphenols in humans.
Collapse
Affiliation(s)
- Sergio Quesada-Vázquez
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, 43204, Spain
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute, CIBEREHD, University of Barcelona, Spain, 08034, Barcelona, Spain
| | - Itziar Eseberri
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, 01006, Spain
| | - Francisco Les
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Zaragoza, 50830, Spain
- Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, Zaragoza, 50013, Spain
| | - Patricia Pérez-Matute
- Infectious Diseases, Microbiota and Metabolism Unit, CSIC Associated Unit. Center for Biomedical Research of La Rioja (CIBIR), Logroño, 26006, Spain
| | - María Herranz-López
- Institute of Research, Development and Innovation in Healthcare Biotechnology of Elche (IDiBE), Miguel Hernández University (UMH), Elche, 03202, Spain
| | - Claude Atgié
- Equipe ClipIn (Colloïdes pour l'Industrie et la Nutrition), Bordeaux INP, Institut CBMN, UMR 5248, Pessac, 33600, France
| | - Marta Lopez-Yus
- Adipocyte and Fat Biology Laboratory (AdipoFat), Translational Research Unit, University Hospital Miguel Servet, Zaragoza, Spain
- Instituto Aragonés de Ciencias de La Salud (IACS), Zaragoza, Spain
- Instituto de Investigación Sanitaria (IIS)-Aragón, Zaragoza, Spain
| | - Paula Aranaz
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, 31008, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - José A Oteo
- Infectious Diseases, Microbiota and Metabolism Unit, CSIC Associated Unit. Center for Biomedical Research of La Rioja (CIBIR), Logroño, 26006, Spain
- Hospital Universitario San Pedro, Logroño, 26006, Spain
| | - Xavier Escoté
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, 43204, Spain
| | - Silvia Lorente-Cebrian
- Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, Zaragoza, 50013, Spain
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Health and Sport Science, University of Zaragoza, 50009, Zaragoza, Spain
- Aragón Health Research Institute (IIS-Aragon), 50009, Zaragoza, Spain
| | - Enrique Roche
- Department of Applied Biology-Nutrition, Institute of Bioengineering, Miguel Hernández University (UMH), Elche, 03202, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, 03010, Spain
- CIBERobn Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain
| | - Arnaud Courtois
- Département des Sciences de l'Environnement, Institut des Sciences de la Vigne et du Vin, UMR OEnologie (UMR 1366, INRAE, Bordeaux INP), AXE Molécules à Intérêt Biologique, Bordeaux, 33882, France
| | - Víctor López
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Zaragoza, 50830, Spain
- Instituto Agroalimentario de Aragón-IA2, CITA-Universidad de Zaragoza, Zaragoza, 50013, Spain
| | - María Puy Portillo
- Nutrition and Obesity Group, Department of Nutrition and Food Science, University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Institute, Vitoria, 01006, Spain
- CIBERobn Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute, CIBEREHD, University of Barcelona, Spain, 08034, Barcelona, Spain
| | - Fermin I Milagro
- Department of Nutrition, Food Sciences and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, 31008, Spain.
- Navarra Institute for Health Research (IdiSNA), Pamplona, 31008, Spain.
- CIBERobn Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain.
| | - Christian Carpéné
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR1297, Toulouse, 31432, France
- Team Dinamix, Institute of Metabolic and Cardiovascular Diseases (I2MC), Paul Sabatier University, Toulouse, 31432, France
| |
Collapse
|
6
|
Nishi K, Nakatani Y, Ishida M, Kadota A, Sugahara T. Anti-Inflammatory Activity of the Combination of Nobiletin and Docosahexaenoic Acid in Lipopolysaccharide-Stimulated RAW 264.7 Cells: A Potential Synergistic Anti-Inflammatory Effect. Nutrients 2024; 16:2080. [PMID: 38999828 PMCID: PMC11243305 DOI: 10.3390/nu16132080] [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: 06/12/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
This study aimed to investigate a synergistic anti-inflammatory effect of a citrus flavonoid nobiletin and docosahexaenoic acid (DHA), one of n-3 long-chain polyunsaturated fatty acids, in combination. Simultaneous treatment with nobiletin and DHA synergistically inhibited nitric oxide production (combination index < 0.9) by mouse macrophage-like RAW 264.7 cells stimulated with lipopolysaccharide (LPS) without cytotoxicity. On the other hand, the inhibitory effect of nobiletin and DHA in combination on proinflammatory cytokine production was not synergistic. Neither nobiletin nor DHA affected the phagocytotic activity of RAW 264.7 cells stimulated with LPS. Immunoblot analysis revealed that the inhibition potency of DHA on the phosphorylation of ERK and p38 and nuclear translocation of NF-κB is markedly enhanced by simultaneously treating with nobiletin, which may lead to the synergistic anti-inflammatory effect. Overall, our findings show the potential of the synergistic anti-inflammatory effect of nobiletin and DHA in combination.
Collapse
Affiliation(s)
- Kosuke Nishi
- Food and Health Function Research Center, Ehime University, Matsuyama 790-8566, Japan; (K.N.); (M.I.)
- Department of Bioscience, Graduate School of Agriculture, Ehime University, Matsuyama 790-8566, Japan;
| | - Yuki Nakatani
- Department of Bioscience, Graduate School of Agriculture, Ehime University, Matsuyama 790-8566, Japan;
| | - Momoko Ishida
- Food and Health Function Research Center, Ehime University, Matsuyama 790-8566, Japan; (K.N.); (M.I.)
- Department of Bioscience, Graduate School of Agriculture, Ehime University, Matsuyama 790-8566, Japan;
| | | | - Takuya Sugahara
- Food and Health Function Research Center, Ehime University, Matsuyama 790-8566, Japan; (K.N.); (M.I.)
- Department of Bioscience, Graduate School of Agriculture, Ehime University, Matsuyama 790-8566, Japan;
| |
Collapse
|
7
|
Zhao T, Nong X, Zhang X, Zhou X, Yu Z, Li X, Chen G. Four new diterpenoids from the aerial parts of Leucas zeylanica (L.) R. Br. Fitoterapia 2024; 175:105948. [PMID: 38588904 DOI: 10.1016/j.fitote.2024.105948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024]
Abstract
Four new undescribed halimane- and labdane-type diterpenoids, named zeylleucapenoids E-H (1-4), along with four known analogues (5-8), were isolated from the aerial parts of Leucas zeylanica (L.) R. Br. Their structures were determined by comprehensive spectroscopic analysis and computational calculations. Compounds 1 and 2 are the highly modified halimane diterpenoids featuring a 6/6/6-fused tricyclic system with an unusual six-membered 6,11-ether ring. Compound 8 exhibits nontoxic effects for zebrafish embryo, while it displays efficient reduction against NO production in a dose-dependent manner and strongly suppresses the secretion of LPS-induced TNF-α and IL-6 cytokines in RAW264.7 macrophages. In addition, marked reductions of iNOS and COX-2 expression were observed. Molecular docking analysis indicated that 8 has high affinities with the target amino acid residues on protein-binding sites, which may be a possible mechanism contributing to the anti-inflammatory potential of this molecule.
Collapse
Affiliation(s)
- Ting Zhao
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Xuhua Nong
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Xuan Zhang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Xueming Zhou
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Zhangxin Yu
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China
| | - Xiaobao Li
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China.
| | - Guangying Chen
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou, Hainan 571158, China; Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan 571158, China.
| |
Collapse
|
8
|
Huang G, Zhang M, Feng K, Xiao J, Huang Q, Ho CT, Liu J. Natural product nobiletin-loaded Pickering emulsion stabilized by bovine serum albumin/carboxymethyl inulin complexes: preparation and digestive characteristics. Front Pharmacol 2024; 15:1375779. [PMID: 38751784 PMCID: PMC11094236 DOI: 10.3389/fphar.2024.1375779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/29/2024] [Indexed: 05/18/2024] Open
Abstract
To expand the application of nobiletin (NOB) in semi-solid functional foods, bovine serum albumin (BSA)/carboxymethyl inulin (CMI) complexes-stabilized Pickering emulsion (BCPE) (φoil = 60%, v/v) was fabricated, and the swallowing index and bioavailability of the NOB-loaded Pickering emulsion was evaluated. Confocal laser scanning microscope (CLSM) and cryo-scanning electron microscopy (cryo-SEM) images revealed that BSA/CMI complexes attached to the oil-water interface. NOB-loaded BCPE exhibited a viscoelastic and shear-thinning behavior. Fork drip test results suggested that the textural value of unloaded and NOB-loaded emulsions was International Dysphagia Diet Standardisation Initiative Level 4, which could be swallowed directly without chewing. The in vitro lipolysis model suggested that NOB had a faster digestive profile and a higher bioaccessibility in the BCPE than in the oil suspension. The in vivo rat model revealed that the oral bioavailability of NOB was increased by 2.07 folds in BCPE compared to its bioavailability in unformulated oil. Moreover, BCPE led to a higher plasma concentration of the major demethylated metabolite of NOB (4'-demethylnobiletin) than the unformulated oil. Accordingly, BCPE enhanced the oral bioavailability of NOB by improving bioaccessibility, absorption, and biotransformation.
Collapse
Affiliation(s)
- Guiying Huang
- College of Light Industry and Food Science, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, China
| | - Man Zhang
- Department of Food Science, Rutgers University, New Brunswick, NJ, United States
| | - Konglong Feng
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jie Xiao
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Qingrong Huang
- Department of Food Science, Rutgers University, New Brunswick, NJ, United States
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, United States
| | - Jun Liu
- College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
9
|
Peng X, Zhang X, Sharma G, Dai C. Thymol as a Potential Neuroprotective Agent: Mechanisms, Efficacy, and Future Prospects. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6803-6814. [PMID: 38507708 DOI: 10.1021/acs.jafc.3c06461] [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: 03/22/2024]
Abstract
Neurodegenerative diseases pose a growing global health challenge, with limited effective therapeutic options. Mitochondrial dysfunction, oxidative stress, neuroinflammation, apoptosis, and autophagy are common underlying mechanisms in these diseases. Thymol is a phenolic monoterpene compound that has gained attention for its diverse biological properties, including antioxidant, anti-inflammatory, and immunomodulatory activities. Thymol supplementation could provide potential neuroprotection and improve cognitive deficits, depressant-like effects, learning, and memory impairments in rodents. Mechanistic investigations reveal that the neuroprotective effects of thymol involve the improvement of oxidative stress, mitochondrial dysfunction, and inflammatory response. Several signaling pathways, including mitochondrial apoptotic, NF-κB, AKT, Nrf2, and CREB/BDNF pathways are also involved. In this review, the neuroprotective effects of thymol, the potential molecular mechanisms, safety, applications, and current challenges toward development as a neuroprotective agent were summarized and discussed. We hope that this review provides valuable insights for the further development of this promising natural product as a promising neuroprotective agent.
Collapse
Affiliation(s)
- Xinyan Peng
- College of Life Sciences, Yantai University, Yantai 264000, P. R. China
| | - Xiaowen Zhang
- College of Life Sciences, Yantai University, Yantai 264000, P. R. China
| | - Gaurav Sharma
- Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Chongshan Dai
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| |
Collapse
|
10
|
Liu Q, Yang GH, Wang NZ, Wang XC, Zhang ZL, Qiao LJ, Cui WJ. Dexmedetomidine suppressed the biological behavior of RAW264.7 cells treated with LPS by down-regulating HOTAIR. Heliyon 2024; 10:e27690. [PMID: 38533037 PMCID: PMC10963246 DOI: 10.1016/j.heliyon.2024.e27690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 03/05/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Background Previous studies have revealed dexmedetomidine have potential protective effects on vital organs by inhibiting the release of inflammatory cytokines. To investigate the effects of dexmedetomidine on sepsis, especially in the initial inflammatory stage of sepsis. RAW264.7 cells were used as the cell model in this study to elucidate the underlying mechanisms. Methods In this study, we conducted several assays to investigate the mechanisms of dexmedetomidine and HOTAIR in sepsis. Cell viability was assessed using the CCK-8 kit, while inflammation responses were measured using ELISA for IL-1β, IL-6, and TNF-α. Additionally, we employed qPCR, MeRIP, and RIP to further explore the underlying mechanisms. Results Our findings indicate that dexmedetomidine treatment enhanced cell viability and reduced the production of inflammatory cytokines in LPS-treated RAW264.7 cells. Furthermore, we observed that the expression of HOTAIR was increased in LPS-treated RAW264.7 cells, which was then decreased upon dexmedetomidine pre-treatment. Further investigation demonstrated that HOTAIR could counteract the beneficial effects of dexmedetomidine on cell viability and cytokine production. Interestingly, we discovered that YTHDF1 targeted HOTAIR and was upregulated in LPS-treated RAW264.7 cells, but reduced in dexmedetomidine treatment. We also found that YTHDF1 increased HOTAIR and HOTAIR m6A levels. Conclusions Collectively, our results suggest that dexmedetomidine downregulates HOTAIR and YTHDF1 expression, which in turn inhibits the biological behavior of LPS-treated RAW264.7 cells. This finding has potential implications for the prevention and treatment of sepsis-induced kidney injury.
Collapse
Affiliation(s)
- Qin Liu
- Intensive Care Unit, Shengli Oilfield Central Hospital, Dongying, Shandong, 257000, China
| | - Guang-Hu Yang
- Intensive Care Unit, Shengli Oilfield Central Hospital, Dongying, Shandong, 257000, China
| | - Nai-Zhi Wang
- Department of Respiratory and Critical Care Medicine, Jinan Central Hospital, Jinan, Shandong, 250013, China
| | - Xin-Cheng Wang
- Intensive Care Unit, Shengli Oilfield Central Hospital, Dongying, Shandong, 257000, China
| | - Zhao-Long Zhang
- Intensive Care Unit, Shengli Oilfield Central Hospital, Dongying, Shandong, 257000, China
| | - Lu-Jun Qiao
- Intensive Care Unit, Shengli Oilfield Central Hospital, Dongying, Shandong, 257000, China
| | - Wen-Juan Cui
- Intensive Care Unit, Shengli Oilfield Central Hospital, Dongying, Shandong, 257000, China
| |
Collapse
|
11
|
Zuzarte M, Sousa C, Alves-Silva J, Salgueiro L. Plant Monoterpenes and Essential Oils as Potential Anti-Ageing Agents: Insights from Preclinical Data. Biomedicines 2024; 12:365. [PMID: 38397967 PMCID: PMC10886757 DOI: 10.3390/biomedicines12020365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Ageing is a natural process characterized by a time-dependent decline of physiological integrity that compromises functionality and inevitably leads to death. This decline is also quite relevant in major human pathologies, being a primary risk factor in neurodegenerative diseases, metabolic disorders, cardiovascular diseases and musculoskeletal disorders. Bearing this in mind, it is not surprising that research aiming at improving human health during this process has burst in the last decades. Importantly, major hallmarks of the ageing process and phenotype have been identified, this knowledge being quite relevant for future studies towards the identification of putative pharmaceutical targets, enabling the development of preventive/therapeutic strategies to improve health and longevity. In this context, aromatic plants have emerged as a source of potential bioactive volatile molecules, mainly monoterpenes, with many studies referring to their anti-ageing potential. Nevertheless, an integrated review on the current knowledge is lacking, with several research approaches studying isolated ageing hallmarks or referring to an overall anti-ageing effect, without depicting possible mechanisms of action. Herein, we aim to provide an updated systematization of the bioactive potential of volatile monoterpenes on recently proposed ageing hallmarks, and highlight the main mechanisms of action already identified, as well as possible chemical entity-activity relations. By gathering and categorizing the available scattered information, we also aim to identify important research gaps that could help pave the way for future research in the field.
Collapse
Affiliation(s)
- Mónica Zuzarte
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal; (J.A.-S.); (L.S.)
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Cátia Sousa
- iNOVA4HEALTH, NOVA Medical School, Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, 1159-056 Lisboa, Portugal;
- Centro Clínico e Académico de Lisboa, 1156-056 Lisboa, Portugal
| | - Jorge Alves-Silva
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal; (J.A.-S.); (L.S.)
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3000-548 Coimbra, Portugal
| | - Lígia Salgueiro
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de S. Comba, 3000-548 Coimbra, Portugal; (J.A.-S.); (L.S.)
- Univ Coimbra, Chemical Engineering and Renewable Resources for Sustainability (CERES), Department of Chemical Engineering, 3030-790 Coimbra, Portugal
| |
Collapse
|
12
|
Surai PF, Surai A, Earle-Payne K. Silymarin and Inflammation: Food for Thoughts. Antioxidants (Basel) 2024; 13:98. [PMID: 38247522 PMCID: PMC10812610 DOI: 10.3390/antiox13010098] [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: 12/22/2023] [Revised: 01/07/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Inflammation is a vital defense mechanism, creating hostile conditions for pathogens, preventing the spread of tissue infection and repairing damaged tissues in humans and animals. However, when inflammation resolution is delayed or compromised as a result of its misregulation, the process proceeds from the acute phase to chronic inflammation, leading to the development of various chronic illnesses. It is proven that redox balance disturbances and oxidative stress are among major factors inducing NF-κB and leading to over-inflammation. Therefore, the anti-inflammatory properties of various natural antioxidants have been widely tested in various in vitro and in vivo systems. Accumulating evidence indicates that silymarin (SM) and its main constituent silibinin/silybin (SB) have great potential as an anti-inflammation agent. The main anti-inflammatory mechanism of SM/SB action is attributed to the inhibition of TLR4/NF-κB-mediated signaling pathways and the downregulated expression of pro-inflammatory mediators, including TNF-α, IL-1β, IL-6, IL-12, IL-23, CCL4, CXCL10, etc. Of note, in the same model systems, SM/SB was able to upregulate anti-inflammatory cytokines (IL-4, IL-10, IL-13, TGF-β, etc.) and lipid mediators involved in the resolution of inflammation. The inflammatory properties of SM/SB were clearly demonstrated in model systems based on immune (macrophages and monocytes) and non-immune (epithelial, skin, bone, connective tissue and cancer) cells. At the same time, the anti-inflammatory action of SM/SB was confirmed in a number of in vivo models, including toxicity models, nonalcoholic fatty liver disease, ischemia/reperfusion models, stress-induced injuries, ageing and exercising models, wound healing and many other relevant model systems. It seems likely that the anti-inflammatory activities of SM/SB are key elements on the health-promoting properties of these phytochemicals.
Collapse
Affiliation(s)
- Peter F. Surai
- Vitagene and Health Research Centre, Bristol BS4 2RS, UK
- Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria
- Faculty of Agricultural and Environmental Sciences, Szent Istvan University, H-2103 Gödöllo, Hungary
- Biochemistry and Physiology Department, Saint-Petersburg State University of Veterinary Medicine, 196084 St. Petersburg, Russia
- Faculty of Veterinary Medicine, Sumy National Agrarian University, 40021 Sumy, Ukraine
- Faculty of Technology of Grain and Grain Business, Odessa National Technological University, 65039 Odessa, Ukraine
| | | | - Katie Earle-Payne
- NHS Greater Glasgow and Clyde, Renfrewshire Health and Social Care Centre, 10 Ferry Road, Renfrew PA4 8RU, UK
| |
Collapse
|
13
|
Lingxia Z, Hong W, Man G, Xinzhou W, Lili W, Zhimin W, Liping D, Erping X. Rabdosichuanin C inhibits productions of pro-inflammatory mediators regulated by NF-κB signaling in LPS-stimulated RAW264.7 cells. J Cell Biochem 2023; 124:1667-1684. [PMID: 37850620 DOI: 10.1002/jcb.30474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 06/14/2023] [Accepted: 08/26/2023] [Indexed: 10/19/2023]
Abstract
Chronic pharyngitis (CP) is an inflammatory disease of the pharyngeal mucosa and its lymphatic tissues that is difficult to treat clinically. However, research on the exact therapeutic agents and molecular mechanisms of CP is still unclear. In this study, we investigated Rabdosichuanin C (RC) to attenuate lipopolysaccharide (LPS)-induced inflammatory damage in RAW264.7 cells by a combination of targeted virtual screening and in vitro activity assay and further clarified its molecular mechanism of action centering on the IκB/nuclear factor kappa B (NF-κB) pathway. Molecular docking and pharmacophore simulation methods were used to screen compounds with IκB inhibitory effects. Expression of genes and proteins related to the IκB/NF-κB signaling pathway by RC in LPS-induced inflammatory injury model of RAW264.7 cells was detected by PCR, enzyme-linked immunosorbent assay, and Western blot. The docking of RC with IκB protein showed good binding energy, and pharmacophore simulations further confirmed the active effect of RC in inhibiting IκB protein. RC intervention in LPS-induced RAW264.7 cells significantly reduced the expression levels of inflammatory factors tumor necrosis factor-α, interleukins-6, iNOS, and CD-86 at the messenger RNA and protein levels, downregulated IκB, p65 protein phosphorylation levels, and significantly inhibited IκB/NF-κB signaling pathway activation. Virtual screening provided us with an effective method to rapidly identify compounds RC that target inhibit the action of IκB, and the activity results showed that RC inhibits NF-κB signaling pathway activation. It is suggested that RC may play a role in the treatment of CP by inhibiting the IκB/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhang Lingxia
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Engineering Technology Research Center for Comprehensive Development and Utilization of Authentic Medicinal Materials in Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Wu Hong
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Gong Man
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Engineering Technology Research Center for Comprehensive Development and Utilization of Authentic Medicinal Materials in Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Wang Xinzhou
- Laboratory of Cell Imaging, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Wang Lili
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Wang Zhimin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dai Liping
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Engineering Technology Research Center for Comprehensive Development and Utilization of Authentic Medicinal Materials in Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xu Erping
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Engineering Technology Research Center for Comprehensive Development and Utilization of Authentic Medicinal Materials in Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
14
|
Sahoo DK, Heilmann RM, Paital B, Patel A, Yadav VK, Wong D, Jergens AE. Oxidative stress, hormones, and effects of natural antioxidants on intestinal inflammation in inflammatory bowel disease. Front Endocrinol (Lausanne) 2023; 14:1217165. [PMID: 37701897 PMCID: PMC10493311 DOI: 10.3389/fendo.2023.1217165] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/07/2023] [Indexed: 09/14/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, relapsing gastrointestinal (GI) disorder characterized by intestinal inflammation. The etiology of IBD is multifactorial and results from a complex interplay between mucosal immunity, environmental factors, and host genetics. Future therapeutics for GI disorders, including IBD, that are driven by oxidative stress require a greater understanding of the cellular and molecular mechanisms mediated by reactive oxygen species (ROS). In the GI tract, oxidative stressors include infections and pro-inflammatory responses, which boost ROS generation by promoting the production of pro-inflammatory cytokines. Nuclear factor kappa B (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2) represent two important signaling pathways in intestinal immune cells that regulate numerous physiological processes, including anti-inflammatory and antioxidant activities. Natural antioxidant compounds exhibit ROS scavenging and increase antioxidant defense capacity to inhibit pro-oxidative enzymes, which may be useful in IBD treatment. In this review, we discuss various polyphenolic substances (such as resveratrol, curcumin, quercetin, green tea flavonoids, caffeic acid phenethyl ester, luteolin, xanthohumol, genistein, alpinetin, proanthocyanidins, anthocyanins, silymarin), phenolic compounds including thymol, alkaloids such as berberine, storage polysaccharides such as tamarind xyloglucan, and other phytochemicals represented by isothiocyanate sulforaphane and food/spices (such as ginger, flaxseed oil), as well as antioxidant hormones like melatonin that target cellular signaling pathways to reduce intestinal inflammation occurring with IBD.
Collapse
Affiliation(s)
- Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Romy M. Heilmann
- Department for Small Animals, Veterinary Teaching Hospital, College of Veterinary Medicine, University of Leipzig, Leipzig, SN, Germany
| | - Biswaranjan Paital
- Redox Regulation Laboratory, Department of Zoology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, Gujarat, India
| | - Virendra Kumar Yadav
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan, Gujarat, India
| | - David Wong
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Albert E. Jergens
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
15
|
Ghiselli F, Yu LE, Piva A, Grilli E, Li Y. Evaluating protective effects of botanicals under inflammation and oxidative stress in chicken apical-out enteroids. Poult Sci 2023; 102:102821. [PMID: 37343346 PMCID: PMC10404760 DOI: 10.1016/j.psj.2023.102821] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/30/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023] Open
Abstract
Botanicals (BOTs) are well known for their anti-inflammatory and antioxidant activities. They have been widely used as feed additives to reduce inflammation and improve intestinal functions in agricultural animals. However, the effects of BOTs on chicken intestinal epithelial functions are not fully understood. The 3D apical-out chicken enteroids recapitulate the intestinal tissue, and allow convenient access to the luminal surface, thus serving as a suitable model for investigating gut functions. The aim of this study was to identify the roles of BOTs in protecting the intestinal epithelium in chicken enteroids under challenging conditions. Apical-out enteroids were isolated from the small intestines of 18 days-old chicken embryos. Lipopolysaccharide (LPS, 10 µg/mL) and menadione (400 µM) challenges were performed in the media with or without BOTs. Paracellular Fluorescein isothiocyanate-dextran 4kD (FD4) permeability, inflammatory cytokine gene expression, and reactive oxygen species (ROS) generation were analyzed post-BOTs and challenges treatments. Statistical analysis was performed using one-way ANOVA and post hoc multiple comparisons among treatments. The results showed that the LPS challenge for 24 h induced a 50% increase in FD4 permeability compared with nontreated control; thymol, thyme essential oil, and phenol-rich extract significantly (P < 0.02) reduced FD4 permeability by 25%, 41%, and 48% respectively, in comparison with LPS treatment. Moreover, the gene expression of inflammatory cytokines was upregulated, tight junction proteins and defensins were downregulated (P < 0.05) after 6 h of LPS treatment, while these BOTs treatments significantly restored the LPS-induced gene expression alterations (P < 0.05). Menadione oxidative challenge for 1 h significantly increased the ROS level compared with unchallenged control. Enteroids treated with thymol and thyme essential oils showed 30% reduced ROS levels, while the phenol-rich extract reduced them by 60%, in comparison with the challenged group (P < 0.0001). These data confirmed the role of BOTs in supporting the barrier function and reducing the disruptive effects of inflammation and oxidation in the chicken intestine.
Collapse
Affiliation(s)
- Federico Ghiselli
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Liang-En Yu
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19711, USA
| | - Andrea Piva
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy; Vetagro S.p.A. - Via Ignazio Porro, Reggio Emilia (RE), Italy
| | - Ester Grilli
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy; Vetagro Inc., Chicago, IL 60603, USA
| | - Yihang Li
- Department of Animal and Food Sciences, University of Delaware, Newark, DE 19711, USA.
| |
Collapse
|
16
|
Luo X, Xiong H, Jiang Y, Fan Y, Zuo C, Chen D, Chen L, Lin H, Gao J. Macrophage Reprogramming via Targeted ROS Scavenging and COX-2 Downregulation for Alleviating Inflammation. Bioconjug Chem 2023. [PMID: 37330989 DOI: 10.1021/acs.bioconjchem.3c00239] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Inflammation-related diseases affect large populations of people in the world and cause substantial healthcare burdens, which results in significant costs in time, material, and labor. Preventing or relieving uncontrolled inflammation is critical for the treatment of these diseases. Herein, we report a new strategy for alleviating inflammation by macrophage reprogramming via targeted reactive oxygen species (ROS) scavenging and cyclooxygenase-2 (COX-2) downregulation. As a proof of concept, we synthesize a multifunctional compound named MCI containing a mannose-based macrophage targeting moiety, an indomethacin (IMC)-based segment for inhibiting COX-2, and a caffeic acid (CAF)-based section for ROS clearance. As revealed by a series of in vitro experiments, MCI could significantly attenuate the expression of COX-2 and the level of ROS, leading to M1 to M2 macrophage reprogramming, as evidenced by the reduction and the elevation in the levels of pro-inflammatory M1 markers and anti-inflammatory M2 markers, respectively. Furthermore, in vivo experiments show MCI's promising therapeutic effects on rheumatoid arthritis (RA). Our work illustrates the success of targeted macrophage reprogramming for inflammation alleviation, which sheds light on the development of new anti-inflammatory drugs.
Collapse
Affiliation(s)
- Xiangjie Luo
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Hui Xiong
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yuhang Jiang
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yifan Fan
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Cuicui Zuo
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Dongxia Chen
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Limin Chen
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Hongyu Lin
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jinhao Gao
- Fujian Provincial Key Laboratory of Chemical Biology, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
17
|
Zhao T, Zhang X, Nong XH, Zhou XM, Chai RR, Li XB, Chen GY. Zeylleucapenoids A-D, Highly Oxygenated Diterpenoids with Anti-Inflammatory Activity from Leucas zeylanica (L.) R. Br. Molecules 2023; 28:molecules28114472. [PMID: 37298948 DOI: 10.3390/molecules28114472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Four previously undescribed highly oxygenated diterpenoids (1-4), zeylleucapenoids A-D, characterized by halimane and labdane skeletons, were isolated from the aerial parts of Leucas zeylanica. Their structures were elucidated primarily via NMR experiments. The absolute configuration of 1 was established using theoretical ECD calculations and X-ray crystallographic analysis, whereas those for 2-4 were assigned using theoretical ORD calculations. Zeylleucapenoids A-D were tested for anti-inflammatory activity against nitric oxide (NO) production in RAW264.7 macrophages, of which only 4 showed significant efficacy with an IC50 value of 38.45 μM. Further, active compound 4 was also evaluated for the inhibition of the release of pro-inflammatory cytokines TNF-α and IL-6 and was found to have a dose-dependent inhibitory effect, while it showed nontoxic activity for zebrafish embryos. A subsequent Western blotting experiment revealed that 4 inhibited the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Furthermore, molecular docking analysis indicated that the possible mechanism of action for 4 may be bind to targets via hydrogen and hydrophobic bond interactions.
Collapse
Affiliation(s)
- Ting Zhao
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Xuan Zhang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Xu-Hua Nong
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Xue-Ming Zhou
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Ru-Ru Chai
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Xiao-Bao Li
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Guang-Ying Chen
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education Hainan Normal University, Haikou 571158, China
- Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| |
Collapse
|
18
|
Huoxiang Zhengqi Oral Liquid Attenuates LPS-Induced Acute Lung Injury by Modulating Short-Chain Fatty Acid Levels and TLR4/NF- κB p65 Pathway. BIOMED RESEARCH INTERNATIONAL 2023; 2023:6183551. [PMID: 36845637 PMCID: PMC9957650 DOI: 10.1155/2023/6183551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/28/2022] [Accepted: 11/24/2022] [Indexed: 02/19/2023]
Abstract
Huoxiang Zhengqi Oral Liquid (HZOL) is a classic Chinese patent medicine used in China for more than 1,000 years in treating gastrointestinal and respiratory diseases. Clinically applied HZOL in early respiratory disease stages can reduce the proportion of lung infection patients that progress to severe acute lung injury (ALI). However, few pharmacological studies evaluated its level of protection against ALI. We explored mechanisms of HZOL against ALI by employing network pharmacology, molecular docking, and rat experiments. Firstly, network pharmacology prediction and published biological evaluation of active ingredients of HZOL suggested that HZOL exerted the protective effect in treating ALI mainly in the areas of regulation of cell adhesion, immune response, and inflammatory response and closely related to the NF-κB pathway. Secondly, molecular docking results demonstrated that imperatorin and isoimperatorin combined well with targets in the NF-κB pathway. Finally, ALI rats induced by lipopolysaccharides (LPS) were used to validate prediction after pretreatment with HZOL for 2 weeks. Results confirmed that lung and colon injury occurred in ALI rats. Furthermore, HZOL exerts anti-inflammatory effects on LPS-induced ALI and gut injury by repairing lung and colon pathology, reducing and alleviating pulmonary edema, inhibiting abnormal enhancement of thymus and spleen index, modulating hematologic indices, and increasing levels of total short-chain fatty acids (SCFAs) in the cecum. Additionally, abnormal accumulation of inflammatory cytokines IL-6, IL-1β, TNF-α, and IFN-γ in serum and bronchoalveolar lavage fluid was significantly reduced after pretreating with HZOL. Furthermore, HZOL downregulated the expression of TLR4, CD14, and MyD88 and phosphorylation of NF-κB p65 in lung tissue. Altogether, HZOL was found to exert an anti-inflammatory effect regulation by increasing levels of SCFAs, inhibiting the accumulation of inflammatory cytokines, and attenuating the activation of the TLR4/NF-κB p65 pathway. Our study provided experimental evidences for the application of HZOL in preventing and treating ALI.
Collapse
|
19
|
Natural Monoterpenes as Potential Therapeutic Agents against Atherosclerosis. Int J Mol Sci 2023; 24:ijms24032429. [PMID: 36768748 PMCID: PMC9917110 DOI: 10.3390/ijms24032429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
Traditional herbal medicines based on natural products play a pivotal role in preventing and managing atherosclerotic diseases, which are among the leading causes of death globally. Monoterpenes are a large class of naturally occurring compounds commonly found in many aromatic and medicinal plants. Emerging evidence has shown that monoterpenes have many biological properties, including cardioprotective effects. Remarkably, an increasing number of studies have demonstrated the therapeutic potential of natural monoterpenes to protect against the pathogenesis of atherosclerosis. These findings shed light on developing novel effective antiatherogenic drugs from these compounds. Herein, we provide an overview of natural monoterpenes' effects on atherogenesis and the underlying mechanisms. Monoterpenes have pleiotropic and multitargeted pharmacological properties by interacting with various cell types and intracellular molecular pathways involved in atherogenesis. These properties confer remarkable advantages in managing atherosclerosis, which has been recognized as a multifaceted vascular disease. We also discuss limitations in the potential clinical application of monoterpenes as therapeutic agents against atherosclerosis. We propose perspectives to give new insights into future preclinical research and clinical practice regarding natural monoterpenes.
Collapse
|
20
|
Sun C, Hao B, Pang D, Li Q, Li E, Yang Q, Zou Y, Liao S, Liu F. Diverse Galactooligosaccharides Differentially Reduce LPS-Induced Inflammation in Macrophages. Foods 2022; 11:foods11243973. [PMID: 36553716 PMCID: PMC9777761 DOI: 10.3390/foods11243973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/28/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
The effects of natural and synthetic galactooligosaccharides (GOS) on inflammation were explored by investigating the structure-activity relationship between the degree of GOS polymerization and in vitro anti-inflammatory activity, together with the potential underlying mechanism of their anti-inflammatory effects. The results demonstrated that GOS had strong anti-inflammatory effects in lipopolysaccharide (LPS)-induced RAW264.7 macrophages, including the inhibition of nitric oxide production and the reduced expression of pro-inflammatory mediators (interleukin-1β, interleukin-6, and tumor necrosis factor α), induced nitric oxide synthase (iNOS), cyclooxygenase 2 (COX-2), and proteins related to the Toll-like receptor 4 (TLR4)/nuclear factor (NF)-κB signaling pathway. GOS4, which has the highest degree of polymerization, exerted the strongest anti-inflammatory activity among the GOS examined. More importantly, our findings confirmed the anti-inflammatory effects of GOS on RAW264.7 macrophages via the TLR4/NF-κB pathway. Our experimental results could provide further support for the exploration of GOS in human nutrition and health.
Collapse
Affiliation(s)
- Congcong Sun
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510600, China
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China
| | - Bifang Hao
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212018, China
| | - Daorui Pang
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510600, China
| | - Qian Li
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510600, China
| | - Erna Li
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510600, China
| | - Qiong Yang
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510600, China
| | - Yuxiao Zou
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510600, China
| | - Sentai Liao
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510600, China
| | - Fan Liu
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510600, China
- Correspondence: ; Tel.:+86-20-87236897
| |
Collapse
|
21
|
Yi S, Zou L, Li Z, Sakao K, Wang Y, Hou DX. In Vitro Antioxidant Activity of Areca Nut Polyphenol Extracts on RAW264.7 Cells. Foods 2022; 11:foods11223607. [PMID: 36429198 PMCID: PMC9689504 DOI: 10.3390/foods11223607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Chewing areca nuts is a popular hobby in the Asian region, and areca nuts are rich in polyphenols, although some alkaloids are included. In this study, we explored the antioxidant activity of areca nut polyphenols (ANP) in lipopolysaccharides (LPS)-stimulated RAW264.7 cells. The results revealed that ANP reduced the level of reactive oxygen species (ROS) in LPS-stimulated RAW264.7 cells and enhanced the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1). RNA-seq analysis showed that ANP down-regulated the transcription of genes related to the cancer pathway at 160 μg/mL, and the inflammatory pathway as well as viral infection pathway at 320 μg/mL. The cellular signaling analysis further revealed that the expressions of these genes were regulated by the mitogen-activated protein kinase (MAPK) pathway, and ANP downregulated the activation of the MAPK signaling pathway stimulated by LPS. Collectively, our findings showed that ANP inhibited the MAPK pathway and activated the Nrf2/HO-1 antioxidant pathways to reduce ROS generation induced by LPS.
Collapse
Affiliation(s)
- Shuhan Yi
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima 890-0065, Japan
| | - Luyan Zou
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Zongjun Li
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Province Key Laboratory of Food Science and Biotechnology, Changsha 410128, China
- National Engineering Center of Plant Functional Components Utilization, Changsha 410128, China
| | - Kozue Sakao
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima 890-0065, Japan
- Faculty of Agriculture, Kagoshima University, Kagoshima 890-0065, Japan
| | - Yuanliang Wang
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
- Hunan Province Key Laboratory of Food Science and Biotechnology, Changsha 410128, China
- National Engineering Center of Plant Functional Components Utilization, Changsha 410128, China
- Correspondence: (Y.W.); (D.-X.H.); Tel.: +86-73184617007 (Y.W.); +81-099-285-8649 (D.-X.H.)
| | - De-Xing Hou
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima 890-0065, Japan
- Faculty of Agriculture, Kagoshima University, Kagoshima 890-0065, Japan
- Correspondence: (Y.W.); (D.-X.H.); Tel.: +86-73184617007 (Y.W.); +81-099-285-8649 (D.-X.H.)
| |
Collapse
|
22
|
Meng D, Wang Y, Liu T. Protective effects of silibinin on LPS-induced inflammation in human periodontal ligament cells. Front Chem 2022; 10:1019663. [PMID: 36300030 PMCID: PMC9591103 DOI: 10.3389/fchem.2022.1019663] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/07/2022] [Indexed: 12/22/2023] Open
Abstract
Clinically, periodontitis is a chronic nonspecific inflammation that leads to damaged teeth and their supporting gum tissues. Although many studies on periodontitis have been conducted, therapy with natural products is still rare. Silibinin has been proven to have anti-inflammatory and antioxidant activities. However, the effects of silibinin on lipopolyssacharide (LPS)-induced inflammation in periodontal ligaments (PDLs) have not yet been investigated. In this study, the PDLs were treated with silibinin (10, 20, and 40 μM) in the presence of LPS. The results showed that silibinin treatment reduced the levels of NO, PGE2, IL-6, TNF-α, MMP-1, and MMP-3 and enhanced the activities of superoxide dismutase (SOD) and glutathione (GSH). Moreover, silibinin treatment downregulated RANKL levels and upregulated OPG and ALP levels. In summary, silibinin protected PDLs against LPS-induced inflammation, oxidative stress, and osteogenic differentiation.
Collapse
Affiliation(s)
- Di Meng
- Department of Stomatology, The Central Hospital Affilliated to Shandong First Medical University, Jinan, China
| | - Yuling Wang
- Department of Stomatology, The Central Hospital Affilliated to Shandong First Medical University, Jinan, China
- Department of Stomatology, Shandong Qianfoshan Hospital, Jinan, China
| | - Tongjun Liu
- Department of Stomatology, The Central Hospital Affilliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
23
|
Liu Y, Yan H, Yu B, He J, Mao X, Yu J, Zheng P, Huang Z, Luo Y, Luo J, Wu A, Chen D. Protective Effects of Natural Antioxidants on Inflammatory Bowel Disease: Thymol and Its Pharmacological Properties. Antioxidants (Basel) 2022; 11:antiox11101947. [PMID: 36290669 PMCID: PMC9598597 DOI: 10.3390/antiox11101947] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 11/24/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a gastrointestinal disease that involves chronic mucosal or submucosal lesions that affect tissue integrity. Although IBD is not life-threatening, it sometimes causes severe complications, such as colon cancer. The exact etiology of IBD remains unclear, but several risk factors, such as pathogen infection, stress, diet, age, and genetics, have been involved in the occurrence and aggravation of IBD. Immune system malfunction with the over-production of inflammatory cytokines and associated oxidative stress are the hallmarks of IBD. Dietary intervention and medical treatment suppressing abnormal inflammation and oxidative stress are recommended as potential therapies. Thymol, a natural monoterpene phenol that is mostly found in thyme, exhibits multiple biological functions as a potential adjuvant for IBD. The purpose of this review is to summarize current findings on the protective effect of thymol on intestinal health in the context of specific animal models of IBD, describe the role of thymol in the modulation of inflammation, oxidative stress, and gut microbiota against gastrointestinal disease, and discuss the potential mechanism for its pharmacological activity.
Collapse
Affiliation(s)
| | - Hui Yan
- Correspondence: (H.Y.); (D.C.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ghiselli F, Giovagnoni G, Felici M, Tugnoli B, Piva A, Grilli E. A mixture of organic acids and thymol protects primary chicken intestinal epithelial cells from Clostridium perfringens infection in vitro. Poult Sci 2022; 101:102101. [PMID: 36088896 PMCID: PMC9464882 DOI: 10.1016/j.psj.2022.102101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/14/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022] Open
Abstract
Necrotic enteritis causes economic losses estimated to be up to 6 billion US dollars per year. Clinical and subclinical infections in poultry are also both correlated with decreased growth and feed efficiency. Moreover, in a context of increased antibiotic resistance, feed additives with enhanced antimicrobial properties are a useful and increasingly needed strategy. In this study, the protective effects of a blend of thymol and organic acids against the effects of Clostridium perfringens type A (CP) on chicken intestinal epithelial cells were investigated and compared to bacitracin, a widely used antibiotic in poultry production. Primary chicken intestinal epithelial cells were challenged with CP for a total time of 3 h to assess the beneficial effect of 2 doses of citric acid, dodecanoic acid, and thymol-containing blend, and compare them with bacitracin. During the challenge, different parameters were recorded, such as transepithelial electrical resistance, cell viability, mRNA expression, and reactive oxygen species production. CP induced inflammation with cytokine production and loss of epithelial barrier integrity. It was also able to induce reactive oxygen species production and increase the caspase expression leading to cellular death. The high dose of the blend acted similarly to bacitracin, preventing the disruptive effects of CP and inducing also an increase in zonula occludens-1 mRNA expression. The low dose only partially prevented the disruptive effects of CP but successfully reduced the associated inflammation. This study shows that the usage of thymol combined with 2 organic acids can protect primary chicken intestinal epithelial cells from CP-induced damages creating a valid candidate to substitute or adjuvate the antibiotic treatment against necrotic enteritis.
Collapse
|
25
|
Zheng Y, Chen J, Wu X, Zhang X, Hu C, Kang Y, Lin J, Li J, Huang Y, Zhang X, Li C. Enhanced Anti-Inflammatory Effects of Silibinin and Capsaicin Combination in Lipopolysaccharide-Induced RAW264.7 Cells by Inhibiting NF-κB and MAPK Activation. Front Chem 2022; 10:934541. [PMID: 35844639 PMCID: PMC9279934 DOI: 10.3389/fchem.2022.934541] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/23/2022] [Indexed: 01/22/2023] Open
Abstract
Silibinin and capsaicin both are natural product molecules with diverse biological activities. In this article, we investigated the anti-inflammatory effects of silibinin combined with capsaicin in lipopolysaccharide (LPS)-induced RAW264.7 cells. The results showed that silibinin combined with capsaicin strongly inhibited LPS-induced nitric oxide (NO), tumor necrosis factor-α (TNF-α), Interleukin-6 (IL-6), and COX-2. Moreover, silibinin combined with capsaicin potently inhibited nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways. The results of the present study indicate that silibinin combined with capsaicin effectively inhibits inflammation.
Collapse
|
26
|
Curcumin and metformin synergistically modulate peripheral and central immune mechanisms of pain. Sci Rep 2022; 12:9713. [PMID: 35690654 PMCID: PMC9188603 DOI: 10.1038/s41598-022-13647-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 05/26/2022] [Indexed: 12/14/2022] Open
Abstract
Metformin is a well-tolerated antidiabetic drug and has recently been repurposed for numerous diseases, including pain. However, a higher dose of metformin is required for effective analgesia, which can potentiate its dose-dependent gastrointestinal side effects. Curcumin is a natural polyphenol and has beneficial therapeutic effects on pain. Curcumin has been used as an analgesic adjuvant with several analgesic drugs, allowing synergistic antinociceptive effects. Nevertheless, whether curcumin can exert synergistic analgesia with metformin is still unknown. In the present study, the nature of curcumin-metformin anti-inflammatory interaction was evaluated in in vitro using lipopolysaccharide-induced RAW 264.7 macrophage and BV-2 microglia cells. In both macrophage and microglia, curcumin effectively potentiates the anti-inflammatory effects of metformin, indicating potential synergistic effects in both peripheral and central pathways of pain. The nature of the interaction between curcumin and metformin was further recapitulated using a mouse model of formalin-induced pain. Coadministration of curcumin and metformin at a 1:1 fixed ratio of their ED50 doses significantly reduced the dose required to produce a 50% effect compared to the theoretically required dose in phase II of the formalin test with a combination index value of 0.24. Besides, the synergistic interaction does not appear to involve severe CNS side effects indicated by no motor alterations, no alterations in short-term and long-term locomotive behaviors, and the general well-being of mice. Our findings suggest that curcumin exerts synergistic anti-inflammation with metformin with no potential CNS adverse effects.
Collapse
|
27
|
Anti-inflammatory effect of essential oil from Amomum Tsaoko Crevost et Lemarie. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
28
|
Xu W, Sun Y, Wang J, Wang B, Xu F, Xie Z, Wang Y. Controlled release of silibinin in GelMA hydrogels inhibits inflammation by inducing M2-type macrophage polarization and promotes vascularization in vitro. RSC Adv 2022; 12:13192-13202. [PMID: 35520139 PMCID: PMC9064440 DOI: 10.1039/d2ra00498d] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/05/2022] [Indexed: 12/28/2022] Open
Abstract
A dry socket is one of the most common complications after tooth extraction. The main etiologies are the loss of blood clots in the socket and the inflammation reaction caused by infection. Current studies on how to prevent dry sockets could not solve these two etiologies at the same time. Recent studies have demonstrated the anti-inflammation role of silibinin. In this study, silibinin was engineered into GelMA hydrogels (Sil-GelMA) with a concentration of 30 mM. The surface characteristics were observed by scanning electron microscopy and the successful loading of silibinin was detected by FTIR spectrometry. The Sil-GelMA hydrogels presented the sustained release ability of silibinin and slow degradation performance of GelMA. Furthermore, silibinin inhibited the inflammatory reaction by inducing M2-type macrophage polarization, promoting the secretion of anti-inflammatory factors (CD206, IL-10) and inhibiting the secretion of anti-inflammatory factors (IL-1β, iNOS). Silibinin also increased the secretion of vascularization-related factor VEGF and promoted vascularization in vitro. This study suggested that the Sil-GelMA hydrogels not only had an anti-inflammatory effect, but also had the potential to promote vascularization. Based on these results, the Sil-GelMA hydrogels might provide a promising prospect for prevention of dry sockets in the future.
Collapse
Affiliation(s)
- Weijian Xu
- Department of Oral and Maxillofacial Surgery, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University Hangzhou 310006 China
| | - Yingjia Sun
- Department of Oral and Maxillofacial Surgery, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University Hangzhou 310006 China
| | - Jia Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine 166 Qiutaobei Road, Shangcheng District Hangzhou Zhejiang 310016 China
| | - Baixiang Wang
- Department of Oral Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University Hangzhou 310006 China
| | - Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University Shenyang 110016 P. R. China
| | - Zhijian Xie
- Department of Oral and Maxillofacial Surgery, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University Hangzhou 310006 China
| | - Yu Wang
- Department of Oral Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University Hangzhou 310006 China
| |
Collapse
|
29
|
Bai Y, Chen J, Hu W, Wang L, Wu Y, Yu S. Silibinin Therapy Improves Cholangiocarcinoma Outcomes by Regulating ERK/Mitochondrial Pathway. Front Pharmacol 2022; 13:847905. [PMID: 35401195 PMCID: PMC8983842 DOI: 10.3389/fphar.2022.847905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Silibinin is widely utilized drug in various cancer treatments, though its application in cholangiocarcinoma has not yet been explored. For the first time, we evaluated the anticancer potential and underlying molecular mechanism of silibinin in treatment of cholangiocarcinoma treatment. Methods: HuCCT-1 and CCLP-1 cells were chosen to be an in vitro study model and were exposed to various concentrations of silibinin for indicated times. Cell viability was evaluated by the cell counting kit-8 (CCK-8) assay and half maximal inhibitory (IC50) concentrations were calculated. Cell proliferation capacity was determined through the use of colony formation and 5-Ethynyl-2′- deoxyuridine (EdU) assays. Cell apoptosis and cycle arrest were assessed by Live/Dead staining assay and flow cytometry (FCM). The protein levels of extracellular regulated protein kinases (ERK)/mitochondrial apoptotic pathway were evaluated through western blotting (WB). Mitochondrial membrane potential changes were determined via 5,5′,6,6′-Tetrachloro-1,1′,3,3′-tetraethyl-imidacarbocyanine iodide (JC-1). A cholangiocarcinoma cell line xenograft model was used to assess the anti-tumor activity of silibinin in vivo. Results: Inhibition of the ERK protein by silibinin led to a significant decrease in mitochondrial membrane potential, which, in turn, caused Cytochrome C to be released from the mitochondria. The activation of downstream apoptotic pathways led to apoptosis of cholangiocarcinoma cells. In general, silibinin inhibited the growth of cholangiocarcinoma cell line xenograft tumors. Conclusions: Silibinin is able to inhibit cholangiocarcinoma through the ERK/mitochondrial apoptotic pathway, which makes silibinin a potential anti-tumor drug candidate for cholangiocarcinoma treatment.
Collapse
Affiliation(s)
- Yang Bai
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jiaqi Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Weijian Hu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Lei Wang
- Department of Urology Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Yulian Wu
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Shi’an Yu, ; Yulian Wu,
| | - Shi’an Yu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
- *Correspondence: Shi’an Yu, ; Yulian Wu,
| |
Collapse
|
30
|
Wang W, Jiang L, Zhu Y, Mei L, Tao Y, Liu Z. Bioactivity-guided isolation of cyclooxygenase-2 inhibitors from Saussurea obvallata (DC.) Edgew. Using affinity solid phase extraction assay. JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114785. [PMID: 34718104 DOI: 10.1016/j.jep.2021.114785] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saussurea obvallata (DC.) Edgew. is a traditional Tibetan medicine used for the treatment of inflammation-related diseases, but the scientific validation was very limited. AIM OF THE STUDY This study aimed to rapid screen and targeted isolate cyclooxygenase-2 (COX-2) inhibitors from S. obvallata extract. MATERIALS AND METHODS An efficient ligand-fishing method based on affinity solid phase extraction (A-SPE) combining with HPLC was developed. The identified COX-2 inhibitors were separated using preparative liquid chromatography. In vitro COX-2 inhibition assays were employed to confirm the inhibitory activities of the isolated compounds. In addition, the effect of the isolated compounds on the production of prostaglandin E2 (PGE2) and the expression of COX-2 in LPS-induced RAW 264.7 were evaluated. RESULTS A total of four phenylpropanoids, isolariciresinol, syringaresinol, pinoresinol and balanophonin were targeted isolated as COX-2 inhibitors with IC50 values of 36.4 ± 2.6 μM, 23.1 ± 1.8 μM, 3.6 ± 0.3 μM and 12.1 ± 0.9 μM, respectively. The isolated compounds significantly inhibited LPS-induced NO production in a dose-dependent manner. And, the results of the inhibitory effect on the release of PGE2 and the expression of COX-2 in LPS-induced macrophages were consistent with A-SPE analysis. CONCLUSION The present work demonstrated that the developed A-SPE-HPLC method could successfully targeted isolated COX-2 inhibitors from S. obvallata extract. And, the isolation results indicated that the therapeutic effect of S. obvallata on inflammation-related diseases was partly based on the COX-2 active ingredients.
Collapse
Affiliation(s)
- Weidong Wang
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China; Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining, Qinghai, China; University of Chinese Academy of Science, Beijing, China
| | - Lei Jiang
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China; Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining, Qinghai, China
| | - Yunhe Zhu
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China; Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining, Qinghai, China; University of Chinese Academy of Science, Beijing, China
| | - Lijuan Mei
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China; Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining, Qinghai, China
| | - Yanduo Tao
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China; Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining, Qinghai, China.
| | - Zenggen Liu
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, China; Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining, Qinghai, China.
| |
Collapse
|
31
|
(E)-9-Octadecenoic Acid Ethyl Ester Derived from Lotus Seedpod Ameliorates Inflammatory Responses by Regulating MAPKs and NF- κB Signalling Pathways in LPS-Induced RAW264.7 Macrophages. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6731360. [PMID: 35035506 PMCID: PMC8754602 DOI: 10.1155/2022/6731360] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/15/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022]
Abstract
Inflammation is generally considered a key risk factor in the progress of several chronic diseases, such as arthritis, gastritis, and hepatitis. Natural products with anti-inflammatory ability have played a great role in the process of overcoming these inflammatory diseases. In this study, we evaluated the anti-inflammatory activities of ten natural compounds derived from lotus seedpod and discovered (E)-9-octadecenoic acid ethyl ester (E9OAEE) inhibited the production of nitric oxide (NO) optimally in lipopolysaccharide (LPS)-induced RAW264.7 macrophages. Furthermore, we explored the effects of E9OAEE on inflammatory responses and the underlying mechanisms in LPS-induced RAW264.7 macrophages. The results indicated that E9OAEE significantly suppressed the production of NO, prostaglandin E2 (PGE2), and tumour necrosis factor-α (TNFα) in a dose-dependent manner. The protein expression and mRNA levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX2) were inhibited by pretreatment of E9OAEE. Furthermore, E9OAEE restrained the phosphorylation of mitogen-activated protein kinase (MAPKs) family members, ERK, P38, and JNK stimulated by LPS-treated for 30 min and prevented the nuclear translocation of nuclear factor-kappa B (NF-κB) prompted by LPS-treated for 6 h in RAW264.7 macrophages. Taken together, we discovered an anti-inflammatory component from lotus seedpod and identified E9OAEE attenuated the inflammatory response in LPS-induced RAW264.7 macrophages probably by regulating the activation of MAPKs and NF-κB signalling pathways, which would provide some base for the development of new anti-inflammatory drugs.
Collapse
|
32
|
Liu T, Xing S, Du J, Wang M, Han J, Li Z. Synthesis and evaluation of the anti-inflammatory activity of novel 8-quinolinesulfonamide derivatives as TLR4/MD-2 inhibitors with efficacy in adjuvant-induced arthritis. Bioorg Chem 2021; 114:105037. [PMID: 34120022 DOI: 10.1016/j.bioorg.2021.105037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/24/2021] [Accepted: 05/28/2021] [Indexed: 12/19/2022]
Abstract
In this study, a series of 8-quinolinesulfonamidederivatives was synthesized, and their anti-inflammatory activity was evaluated. Among them, compound 3l was found to be the best anti-inflammatory agent, with IC50 values of 2.61 ± 0.39, 9.74 ± 0.85, and 12.71 ± 1.34 μM against NO, TNF-α and IL-1β production respectively. And 3l could significantly prevent lipopolysaccharide (LPS)-induced expression of inflammatory mediators (iNOS and COX-2). Molecule docking results showed that 3l could bind to the LPS binding site of toll-like receptor 4 (TLR4)/MD-2, and 3l was then identified as TLR4/MD-2 inhibitor by co-immunoprecipitation (co-IP) and cellular thermal shift assay (CTESA). Preliminary mechanism studies indicated that 3l could prevent TLR4 from being activated by disrupting TLR4/MD-2 heterodimerization and TLR4 homodimerization, thereby blocking the activation of the NF-κB/MAPK signaling pathway. Furthermore, observation of rat foot swelling, joint pathology and serum inflammatory cytokine levels proved that compound 3l had a significant therapeutic effect on adjuvant-induced arthritis (AIA) in rats in vivo. These results indicated that compound 3l is a potential anti-inflammatory agent, from which more effective anti-inflammatory drugs could be developed.
Collapse
Affiliation(s)
- Tongtong Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Siqi Xing
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jiyu Du
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Min Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Jianfei Han
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zeng Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|