1
|
Xia Q, Zhang J. Interaction Between Autophagy and the Inflammasome in Human Tumors: Implications for the Treatment of Human Cancers. Cell Biochem Funct 2025; 43:e70035. [PMID: 39722223 DOI: 10.1002/cbf.70035] [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: 09/27/2024] [Revised: 10/10/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
Autophagy is a physiologically regulated cellular process orchestrated by autophagy-related genes (ATGs) that, depending on the tumor type and stage, can either promote or suppress tumor growth and progression. It can also modulate cancer stem cell maintenance and immune responses. Therefore, targeted manipulation of autophagy may inhibit tumor development by overcoming tumor-promoting mechanisms. The inflammasome is another multifunctional bioprocess that induces a form of pro-inflammatory programmed cell death, called pyroptosis. Dysregulation or overactivation of the inflammasome has been implicated in tumor pathogenesis and development. Additionally, autophagy can inhibit the NLRP3 inflammasome by removing inflammatory drivers. Recent research suggests that the NLRP3 inflammasome, in turn, affects autophagy. Understanding the complex interplay between autophagy and inflammasomes could lead to more precise and effective strategies for cancer treatments. In this review, we summarize the impact of autophagy and inflammasome dysregulation on tumor progression or suppression. We then highlight their targeting for cancer treatment as monotherapy or in combination with other therapies. Furthermore, we discuss the interaction between autophagy and tumor-promoting inflammation or the NLRP3 inflammasome. Finally, based on recent findings, we review the potential of this interaction for cancer treatment.
Collapse
Affiliation(s)
- Qing Xia
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingzhou Zhang
- Peking Union Medical College, Graduate School of Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Saadati F, Modarresi Chahardehi A, Jamshidi N, Jamshidi N, Ghasemi D. Coumarin: A natural solution for alleviating inflammatory disorders. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 7:100202. [PMID: 39398983 PMCID: PMC11470182 DOI: 10.1016/j.crphar.2024.100202] [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: 04/28/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
Coumarin, a naturally occurring compound found in various plants, has a rich history of use in traditional medicine. Recent research has highlighted its anti-inflammatory properties, positioning it as a promising candidate for treating inflammatory disorders such as rheumatoid arthritis, asthma, and inflammatory bowel disease. This narrative review aims to comprehensively summarize the current knowledge regarding coumarin's pharmacological effects in alleviating inflammatory conditions by analyzing preclinical and clinical studies. The review focuses on elucidating the mechanisms through which coumarin exerts its anti-inflammatory effects, including its antioxidant activity, inhibiting pro-inflammatory cytokine production, and modulation of immune cell functions. Additionally, the paper addresses potential limitations of using coumarin, such as concerns about toxicity at high doses or with prolonged use. Before widespread clinical application, further investigation is needed to fully understand coumarin's potential benefits and risks.
Collapse
Affiliation(s)
- Farnoosh Saadati
- Department of Cellular and Molecular Biology, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | | | - Negar Jamshidi
- Kimia Andisheh Teb Medical and Molecular Research Laboratory Co., Tehran, Iran
| | - Nazanin Jamshidi
- Kimia Andisheh Teb Medical and Molecular Research Laboratory Co., Tehran, Iran
| | - Darioush Ghasemi
- Kimia Andisheh Teb Medical and Molecular Research Laboratory Co., Tehran, Iran
| |
Collapse
|
3
|
Shi LY, Wang Y, Yang YJ, Li Q, Yang ZX, Sun LH, Luo FQ, He YH, Zhang SP, Su N, Liu JQ, He Y, Guan YC, Wei ZL, Cao YX, Zhang D. NLRP4E regulates actin cap formation through SRC and CDC42 during oocyte meiosis. Cell Mol Biol Lett 2024; 29:68. [PMID: 38730334 PMCID: PMC11088158 DOI: 10.1186/s11658-024-00580-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Members of the nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain containing (NLRP) family regulate various physiological and pathological processes. However, none have been shown to regulate actin cap formation or spindle translocation during the asymmetric division of oocyte meiosis I. NLRP4E has been reported as a candidate protein in female fertility, but its function is unknown. METHODS Immunofluorescence, reverse transcription polymerase chain reaction (RT-PCR), and western blotting were employed to examine the localization and expression levels of NLRP4E and related proteins in mouse oocytes. small interfering RNA (siRNA) and antibody transfection were used to knock down NLRP4E and other proteins. Immunoprecipitation (IP)-mass spectrometry was used to identify the potential proteins interacting with NLRP4E. Coimmunoprecipitation (Co-IP) was used to verify the protein interactions. Wild type (WT) or mutant NLRP4E messenger RNA (mRNA) was injected into oocytes for rescue experiments. In vitro phosphorylation was employed to examine the activation of steroid receptor coactivator (SRC) by NLRP4E. RESULTS NLRP4E was more predominant within oocytes compared with other NLRP4 members. NLRP4E knockdown significantly inhibited actin cap formation and spindle translocation toward the cap region, resulting in the failure of polar body extrusion at the end of meiosis I. Mechanistically, GRIN1, and GANO1 activated NLRP4E by phosphorylation at Ser429 and Thr430; p-NLRP4E is translocated and is accumulated in the actin cap region during spindle translocation. Next, we found that p-NLRP4E directly phosphorylated SRC at Tyr418, while p-SRC negatively regulated p-CDC42-S71, an inactive form of CDC42 that promotes actin cap formation and spindle translocation in the GTP-bound form. CONCLUSIONS NLRP4E activated by GRIN1 and GANO1 regulates actin cap formation and spindle translocation toward the cap region through upregulation of p-SRC-Tyr418 and downregulation of p-CDC42-S71 during meiosis I.
Collapse
Affiliation(s)
- Li-Ya Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
- Reproductive Medicine Center, Shanghai East Hospital, Tongji University School of Medicine, 551 Pudong South Road, Shanghai, 200120, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Yang Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Yan-Jie Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Qian Li
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), 123 Tianfei Lane, Nanjing, 210018, China
| | - Zhi-Xia Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Li-Hua Sun
- Reproductive Medicine Center, Shanghai East Hospital, Tongji University School of Medicine, 551 Pudong South Road, Shanghai, 200120, China
| | - Fu-Qiang Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yu-Hao He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Shu-Ping Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Ning Su
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Jia-Qi Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China
| | - Ye He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yi-Chun Guan
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, 7 Rehabilitation Front Street, Zhengzhou, 450000, Henan, China.
| | - Zhao-Lian Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Yun-Xia Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Dong Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- State Key Lab of Reproductive Medicine, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
4
|
Chen H, Jian Z, Xu T, Xu L, Deng L, Shao L, Zhang L, He L, Li Y, Zhu L. Advances in the mechanism of inflammasomes activation in herpes virus infection. Front Immunol 2024; 15:1346878. [PMID: 38590522 PMCID: PMC10999540 DOI: 10.3389/fimmu.2024.1346878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/04/2024] [Indexed: 04/10/2024] Open
Abstract
Herpesviruses, prevalent DNA viruses with a double-stranded structure, establish enduring infections and play a part in various diseases. Despite their deployment of multiple tactics to evade the immune system, both localized and systemic inflammatory responses are triggered by the innate immune system's recognition of them. Recent progress has offered more profound understandings of the mechanisms behind the activation of the innate immune system by herpesviruses, specifically through inflammatory signaling. This process encompasses the initiation of an intracellular nucleoprotein complex, the inflammasome associated with inflammation.Following activation, proinflammatory cytokines such as IL-1β and IL-18 are released by the inflammasome, concurrently instigating a programmed pathway for cell death. Despite the structural resemblances between herpesviruses, the distinctive methods of inflammatory activation and the ensuing outcomes in diseases linked to the virus exhibit variations.The objective of this review is to emphasize both the similarities and differences in the mechanisms of inflammatory activation among herpesviruses, elucidating their significance in diseases resulting from these viral infections.Additionally, it identifies areas requiring further research to comprehensively grasp the impact of this crucial innate immune signaling pathway on the pathogenesis of these prevalent viruses.
Collapse
Affiliation(s)
- Hourui Chen
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhijie Jian
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Tong Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lei Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lishuang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lina Shao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Leyi Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Li He
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Youyou Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ling Zhu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
5
|
Wang J, Liu Y, Guo Y, Liu C, Yang Y, Fan X, Yang H, Liu Y, Ma T. Function and inhibition of P38 MAP kinase signaling: Targeting multiple inflammation diseases. Biochem Pharmacol 2024; 220:115973. [PMID: 38103797 DOI: 10.1016/j.bcp.2023.115973] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Inflammation is a natural host defense mechanism that protects the body from pathogenic microorganisms. A growing body of research suggests that inflammation is a key factor in triggering other diseases (lung injury, rheumatoid arthritis, etc.). However, there is no consensus on the complex mechanism of inflammatory response, which may include enzyme activation, mediator release, and tissue repair. In recent years, p38 MAPK, a member of the MAPKs family, has attracted much attention as a central target for the treatment of inflammatory diseases. However, many p38 MAPK inhibitors attempting to obtain marketing approval have failed at the clinical trial stage due to selectivity and/or toxicity issues. In this paper, we discuss the mechanism of p38 MAPK in regulating inflammatory response and its key role in major inflammatory diseases and summarize the synthetic or natural products targeting p38 MAPK to improve the inflammatory response in the last five years, which will provide ideas for the development of novel clinical anti-inflammatory drugs based on p38 MAPK inhibitors.
Collapse
Affiliation(s)
- Jiahui Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yongjian Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yushi Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Cen Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yuping Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaoxiao Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hongliu Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yonggang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Tao Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
6
|
Sharma D, Radha, Kumar M, Andrade-Cetto A, Puri S, Kumar A, Thakur M, Chandran D, Pundir A, Prakash S, Pandiselvam R, Sandhu S, Khosla A, Kumar S, Lorenzo JM. Chemical Diversity and Medicinal Potential of Vitex negundo L.: From Traditional Knowledge to Modern Clinical Trials. Chem Biodivers 2023; 20:e202301086. [PMID: 37851484 DOI: 10.1002/cbdv.202301086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/27/2023] [Accepted: 10/01/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND In Vedic context, Nirgundi (V. negundo) has been utilized for its anti-inflammatory, analgesic, and wound-healing properties. It has been employed to alleviate pain, treat skin conditions, and address various ailments. The plant's leaves, roots, and seeds have all found applications in traditional remedies. The knowledge of Nirgundi's medicinal benefits has been passed down through generations, and it continues to be a part of Ayurvedic and traditional medicine practices in India.
Collapse
Affiliation(s)
- Diksha Sharma
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, 173229, India
| | - Radha
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, 173229, India
| | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR- Central Institute for Research on Cotton Technology, Mumbai, 400019, India
- Department of Biology, East Carolina University, Greenville, 27858, USA
| | - Adolfo Andrade-Cetto
- Laboratorio de Etnofarmacología, Departamento de BiologíaCelular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Av. Universidad 3000, Circuito Exterior S/N, Coyoacán, C.U., Mexico City, 04510, Mexico
| | - Sunil Puri
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, 173229, India
| | - Amit Kumar
- GLA University, Mathura, Uttar Pradesh, 281 406, India
| | - Mamta Thakur
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, 173229, India
| | - Deepak Chandran
- Department of Animal Husbandry, Government of Kerala, Palakkad, 679335, Kerala, India
| | - Ashok Pundir
- School of Mechanical and Civil Engineering, Shoolini University of Biotechnology and Management Sciences, Solan, 173229, India
| | - Suraj Prakash
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, 173229, India
| | - Ravi Pandiselvam
- Division of Physiology, Biochemistry and Post-Harvest Technology, ICAR - Central Plantation Crops Research Institute (CPCRI), Kasaragod, 671 124, Kerala, India
| | - Surinder Sandhu
- Department of Plant Breeding and Genetics, Punjab Agricultural University, Ludhiana, 141004, India
| | - Ananya Khosla
- Stanford University, 450 Serra Mall, Stanford, California, USA, 94305
| | - Sunil Kumar
- Indian Institute of Farming Systems Research, Modipuram, 250110, India
| | - Jose M Lorenzo
- CentroTecnológico de la Carne de Galicia, rúa Galicia n○ 4, Parque Tecnológico de Galicia, San Cibrao das Viñas, 32900 Ourense, Spain
| |
Collapse
|
7
|
Ali MI, Naseer MM. Recent biological applications of heterocyclic hybrids containing s-triazine scaffold. RSC Adv 2023; 13:30462-30490. [PMID: 37854486 PMCID: PMC10580144 DOI: 10.1039/d3ra05953g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023] Open
Abstract
s-Triazine possesses an auspicious status in the field of drug discovery and development owing to its presence in many naturally occurring compounds as well as commercially available drugs like enasidenib, gedatolisib, bimiralisib, atrazine, indaziflam, and triaziflam. Easy, cost-effective, and efficient access to its derivatives in addition to their splendid biological activities such as anticancer, anti-inflammatory, antiviral, anticonvulsant, anti-tubercular, antidiabetic, antimicrobial, makes it an attractive heterocyclic nucleus in the field of medicinal chemistry. Other than the direct access of its derivatives from simple commercially available starting materials like amidine, the s-triazine derivatives have also been obtained starting from an inexpensive commercially available 2,4,6-trichloro-1,3,5-triazine (TCT) commonly known as cyanuric chloride. Owing to the high reactivity and the possibility of sequential substitution of TCT, a variety of biologically active heterocyclic scaffolds have been installed on this nucleus in order to have more potent compounds. These s-triazine-based heterocyclic hybrids have been reported to show enhanced biological activities in recent years. Therefore, it is important to summarize and highlight recent examples of these hybrids which is imperative to attract the attention of the drug development community.
Collapse
Affiliation(s)
- Muhammad Imran Ali
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan +92-5190642241 +92-5190642129
| | - Muhammad Moazzam Naseer
- Department of Chemistry, Quaid-i-Azam University Islamabad 45320 Pakistan +92-5190642241 +92-5190642129
| |
Collapse
|
8
|
Sartinah A, Nugrahani I, Ibrahim S, Anggadiredja K. Potential metabolites of Arecaceae family for the natural anti-osteoarthritis medicine: A review. Heliyon 2022; 8:e12039. [PMID: 36561673 PMCID: PMC9763769 DOI: 10.1016/j.heliyon.2022.e12039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/28/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a chronic inflammatory disorder of the joints caused by fluid and cartilage matrix component reduction. This disease results in symptoms of pain, deformity, and limitation of movement. In general, OA is treated with anti-inflammatory drugs and chondroprotection compounds, includes natural nutraceutical ingredients, which are expected to be effective and have minimal side effects. Arecaceae plants are widely spread worldwide, especially in tropical areas. The objective of this review is to collect information about the Arecaceae family as anti-OA agents, with the main study focusing on the primary and secondary metabolites of plants of the Arecaceae family, i.e., sugar palm (Arenga pinnata), nipa palm (Nypa fruticans), palmyra palm (Borassus flabellifer), date palm (Phoenix dactylifera), and betel nut (Areca catechu) have potential as anti-OA agents. The Arecaceae's metabolites that show anti-inflammatory and chondroprotective effects are galactomannan, fatty acids (linoleic and linolenic acids), flavonoids (quercetin, luteolin, isorhamnetin), phenolics (coumaric acid, ferulic acid), polyphenols (epicatechin), and steroids (stigmasterol, campesterol, spirostane). Based on the reports, the Arecaceae family plants become worthy of being explored and developed into natural anti-OA products, such as supplements or nutraceuticals.
Collapse
Affiliation(s)
- Ari Sartinah
- School of Pharmacy, Bandung Institute of Technology, Bandung 40132, Indonesia
| | - Ilma Nugrahani
- School of Pharmacy, Bandung Institute of Technology, Bandung 40132, Indonesia
- Corresponding author.
| | - Slamet Ibrahim
- Faculty of Pharmacy, Universitas Jenderal Achmad Yani, Cimahi, Indonesia
| | | |
Collapse
|
9
|
Darvish Khadem M, Tabandeh MR, Haschemi A, Kheirollah A, Shahriari A. Dimethyl itaconate reprograms neurotoxic to neuroprotective primary astrocytes through the regulation of NLRP3 inflammasome and NRF2/HO-1 pathways. Mol Cell Neurosci 2022; 122:103758. [PMID: 35868484 DOI: 10.1016/j.mcn.2022.103758] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
The activation of neurotoxic reactive astrocytes contributes to the pathogenesis of many neurodegenerative diseases. Itaconate, a product of cellular metabolism, is released from activated macrophage/microglia and has been shown to regulate inflammatory responses in several mammalian cells. This study was designed to investigate the impact of cell-permeable dimethyl itaconate (DI) on reactive astrocyte-dependent neurotoxicity. Primary murine astrocyte cells were isolated and stimulated with lipopolysaccharide (LPS) to generate reactive astrocytes. Treating these activated cells with DI was able to diminish the neurotoxic phenotype of reactive astrocytes, as we found reduced LPS-induced Nod-like receptor protein 3 (NLRP3) inflammasome activation and interleukin-1β (IL-1β) secretion. DI reduced the level of inflammasome components, attenuated inflammasome assembly and subsequently reduced caspase-1 cleavage and IL-1β levels. Additionally, DI attenuated nuclear factor-kappa B (NF-κB) phosphorylation in LPS-activated astrocytes and also protected astrocytes from LPS-induced cytotoxicity, including a lowering of Bax and caspase3. DI-treated reactive astrocytes showed an elevated GSH/GSSG ratio and improved antioxidant defense factors including catalase and superoxide dismutase, while lipid peroxidation was reduced. We found that DI activated the nuclear factor 2 (NRF2) and heme oxygenase-1 (HO-1) pathway in astrocytes and thereby potentially control redox-regulation and the inflammatory state of astrocytes. Collectively, these results indicate the neuroprotective role of DI by reprogramming astrocytes from neurotoxic A1 to neuroprotective A2 states and thereby reveal a novel potential strategy for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Mohammad Darvish Khadem
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz 61357-831351, Iran
| | - Mohammad Reza Tabandeh
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz 61357-831351, Iran; Stem Cells and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Alireza Kheirollah
- Department of Biochemistry, Medical School, Cellular & Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Shahriari
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz 61357-831351, Iran.
| |
Collapse
|
10
|
Ren Y, Feng J, Lin Y, Reinach PS, Liu Y, Xia X, Ma X, Chen W, Zheng Q. MiR-223 inhibits hyperosmolarity-induced inflammation through downregulating NLRP3 activation in human corneal epithelial cells and dry eye patients. Exp Eye Res 2022; 220:109096. [DOI: 10.1016/j.exer.2022.109096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/29/2022] [Accepted: 04/21/2022] [Indexed: 01/10/2023]
|
11
|
Kuang QX, Li QZ, Lei LR, Wang YM, Huang LJ, Dai YF, Peng W, Zhang MZ, Wang D, Gu YC, Deng Y, Guo DL. Proliferatins Suppress Lipopolysaccharide-induced Inflammation via Inhibition of the NF-κB and MAPK Signaling Pathways. Bioorg Chem 2022; 124:105810. [DOI: 10.1016/j.bioorg.2022.105810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 02/08/2023]
|
12
|
Bombaci G, Sarangdhar MA, Andina N, Tardivel A, Yu ECW, Mackie GM, Pugh M, Ozan VB, Banz Y, Spinetti T, Hirzel C, Youd E, Schefold JC, Taylor G, Gazdhar A, Bonadies N, Angelillo-Scherrer A, Schneider P, Maslowski KM, Allam R. LRR-protein RNH1 dampens the inflammasome activation and is associated with COVID-19 severity. Life Sci Alliance 2022; 5:5/6/e202101226. [PMID: 35256513 PMCID: PMC8922048 DOI: 10.26508/lsa.202101226] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/12/2022] Open
Abstract
RNH1 prevents inflammation by inhibiting inflammasome activation through controlling caspase-1 protein levels. In COVID-19 patients, RNH1 expression levels were negatively associated with disease severity and inflammation, suggesting a role for RNH1 in SARS-CoV-2–mediated inflammation and pathology. Inflammasomes are cytosolic innate immune sensors of pathogen infection and cellular damage that induce caspase-1–mediated inflammation upon activation. Although inflammation is protective, uncontrolled excessive inflammation can cause inflammatory diseases and can be detrimental, such as in coronavirus disease (COVID-19). However, the underlying mechanisms that control inflammasome activation are incompletely understood. Here we report that the leucine-rich repeat (LRR) protein ribonuclease inhibitor (RNH1), which shares homology with LRRs of NLRP (nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing) proteins, attenuates inflammasome activation. Deletion of RNH1 in macrophages increases interleukin (IL)-1β production and caspase-1 activation in response to inflammasome stimulation. Mechanistically, RNH1 decreases pro-IL-1β expression and induces proteasome-mediated caspase-1 degradation. Corroborating this, mouse models of monosodium urate (MSU)-induced peritonitis and lipopolysaccharide (LPS)-induced endotoxemia, which are dependent on caspase-1, respectively, show increased neutrophil infiltration and lethality in Rnh1−/− mice compared with wild-type mice. Furthermore, RNH1 protein levels were negatively related with disease severity and inflammation in hospitalized COVID-19 patients. We propose that RNH1 is a new inflammasome regulator with relevance to COVID-19 severity.
Collapse
Affiliation(s)
- Giuseppe Bombaci
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Mayuresh Anant Sarangdhar
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Nicola Andina
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Aubry Tardivel
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Eric Chi-Wang Yu
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Gillian M Mackie
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Matthew Pugh
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Vedat Burak Ozan
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yara Banz
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Thibaud Spinetti
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Cedric Hirzel
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Esther Youd
- School of Medicine, Dentistry and Nursing, Forensic Medicine and Science. University of Glasgow, Scotland, UK
| | - Joerg C Schefold
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Graham Taylor
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Amiq Gazdhar
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nicolas Bonadies
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Anne Angelillo-Scherrer
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Kendle M Maslowski
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Ramanjaneyulu Allam
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
Miranda RDS, Jesus BDSM, Silva Luiz SR, Viana CB, Adão Malafaia CR, Figueiredo FDS, Carvalho TDSC, Silva ML, Londero VS, Costa‐Silva TA, Lago JHG, Martins RCC. Antiinflammatory activity of natural triterpenes—An overview from 2006 to 2021. Phytother Res 2022; 36:1459-1506. [DOI: 10.1002/ptr.7359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/29/2021] [Accepted: 12/08/2021] [Indexed: 12/22/2022]
Affiliation(s)
- Rodrigo de Souza Miranda
- Institute of Natural Products Research Walter Mors Federal University of Rio de Janeiro (UFRJ) Rio de Janeiro Brazil
| | | | - Sandra Regina Silva Luiz
- Institute of Microbiology Paulo de Góes Federal University of Rio de Janeiro (IMPG‐UFRJ) Rio de Janeiro Brazil
| | - Cristina Borges Viana
- Institute of Natural Products Research Walter Mors Federal University of Rio de Janeiro (UFRJ) Rio de Janeiro Brazil
| | - Camila Rodrigues Adão Malafaia
- Laboratory of Natural Products and Biological Assays, Natural Products and Food Department, Faculty of Pharmacy Federal University of Rio de Janeiro (UFRJ) Rio de Janeiro Brazil
| | - Fabiana de Souza Figueiredo
- Institute of Natural Products Research Walter Mors Federal University of Rio de Janeiro (UFRJ) Rio de Janeiro Brazil
| | | | - Matheus Lopes Silva
- Center of Human and Natural Sciences Federal University of ABC (UFABC) Santo André Brazil
| | - Vinicius Silva Londero
- Institute of Environmental, Chemical and Pharmaceutical Sciences Federal University of São Paulo (UNIFESP) Diadema Brazil
| | | | | | - Roberto Carlos Campos Martins
- Institute of Natural Products Research Walter Mors Federal University of Rio de Janeiro (UFRJ) Rio de Janeiro Brazil
| |
Collapse
|
14
|
Sharma A, Sharma S, Kumar A, Kumar V, Sharma AK. Plant Secondary Metabolites: An Introduction of Their Chemistry and Biological Significance with Physicochemical Aspect. PLANT SECONDARY METABOLITES 2022:1-45. [DOI: 10.1007/978-981-16-4779-6_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
15
|
[Dexmedetomidine preconditioning alleviates acute lung injury induced by intestinal ischemia-reperfusion in rats by inhibiting NLRP3 inflammasome activation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1857-1863. [PMID: 35012919 PMCID: PMC8752431 DOI: 10.12122/j.issn.1673-4254.2021.12.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To investigate the protective effect of dexmedetomidine (Dex) against acute lung injury induced by intestinal ischemia-reperfusion (II/R) in rats and its effect on NLRP3 inflammasome activity. METHODS Thirty-two normal male SD rats were randomly divided into 4 groups (n=8): the sham operation group, where the superior mesenteric artery (SMA) was exposed only; II/R group, where the SMA was occluded for 1 h followed by reperfusion for 2 h; Dex+II/R group, where the rats were subjected to II/R and received intraperitoneal injection of Dex before reperfusion; and Dex group, where the rats received Dex pretreatment and sham operation. The rats in sham operation group and II/R group received intraperitoneal injection of normal saline. The wet/dry weight ratio (W/D) and myeloperoxidase (MPO) activity in the lung tissues were measured, and HE staining was used to evaluate lung pathologies and determine lung injury score of the rats. The levels of inflammatory cytokines (TNF-α, IL-18, and IL-1β) in the lung tissue were detected using ELISA, and the expressions of NLRP3, ASC, caspase-1 and p-AMPK proteins were determined with Western blotting. RESULTS Compared with the sham-operated rats, the rats with II/R injury showed obvious lung pathologies and significantly increased W/D value, MPO activity and expression of TNF-α, IL-18 and IL-1β in the lung tissue (P < 0.05) with also significantly increased expressions of NLRP3, ASC, and caspase-1 proteins (P < 0.05) but obviously lowered expression of p-AMPK protein (P < 0.05) in the lung tissues. Compared with those in II/R group, the rats in Dex+II/R group showed milder lung pathologies, significantly reduced W/D value, MPO activity and expressions of TNF-α, IL-18 and IL-1β in the lung tissue (P < 0.05), and significant lower expressions of NLRP3, ASC, and caspase-1 (P < 0.05) but higher expression of p-AMPK protein (P < 0.05). CONCLUSION Dex treatment reduces II/R-induced inflammatory response by inhibiting the activation of NLRP3 inflammasomes, thereby improving acute lung injury caused by II/R in rats.
Collapse
|
16
|
Anti-Inflammatory Mechanisms of Novel Synthetic Ruthenium Compounds. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112110092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Inflammation is the primary biological reaction to induce severe infection or injury in the immune system. Control of different inflammatory cytokines, such as nitric oxide (NO), interleukins (IL), tumor necrosis factor alpha-(TNF-α), noncytokine mediator, prostaglandin E2 (PGE2), mitogen activated protein kinases (MAPKs) and nuclear factor kappa B (NF-κB), facilitates anti-inflammatory effect of different substances. Coordination metal complexes have been applied as metallo-drugs. Several metal complexes have found to possess potent biological activities, especially anticancer, cardioprotective, chondroprotective and anti-parasitosis activities. Among the metallo drugs, ruthenium-based (Ru) complexes have paid much attention in clinical applications. Despite the kinetic nature of Ru complexes is similar to platinum in terms of cell division events, their toxic effect is lower than that of cisplatin. This paper reviews the anti-inflammatory effect of novel synthetic Ru complexes with potential molecular mechanisms that are actively involved.
Collapse
|
17
|
Zafar A, Pong Ng H, Diamond-Zaluski R, Kim GD, Ricky Chan E, Dunwoodie SL, Smith JD, Mahabeleshwar GH. CITED2 inhibits STAT1-IRF1 signaling and atherogenesis. FASEB J 2021; 35:e21833. [PMID: 34365659 DOI: 10.1096/fj.202100792r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/09/2021] [Accepted: 07/20/2021] [Indexed: 11/11/2022]
Abstract
Macrophages are the principal component of the innate immune system. They play very crucial and multifaceted roles in the pathogenesis of inflammatory vascular diseases. There is an increasing recognition that transcriptionally dynamic macrophages are the key players in the pathogenesis of inflammatory vascular diseases. In this context, the accumulation and aberrant activation of macrophages in the subendothelial layers govern atherosclerotic plaque development. Macrophage-mediated inflammation is an explicitly robust biological response that involves broad alterations in inflammatory gene expression. Thus, cell-intrinsic negative regulatory mechanisms must exist which can restrain inflammatory response in a spatiotemporal manner. In this study, we identified CBP/p300-interacting transactivator with glutamic acid/aspartic acid-rich carboxyl-terminal domain 2 (CITED2) as one such cell-intrinsic negative regulator of inflammation. Our in vivo studies show that myeloid-CITED2-deficient mice on the Apoe-/- background have larger atherosclerotic lesions on both control and high-fat/high-cholesterol diets. Our integrated transcriptomics and gene set enrichment analyses studies show that CITED2 deficiency elevates STAT1 and interferon regulatory factor 1 (IRF1) regulated pro-inflammatory gene expression in macrophages. At the molecular level, our studies identify that CITED2 deficiency elevates IFNγ-induced STAT1 transcriptional activity and STAT1 enrichment on IRF1 promoter in macrophages. More importantly, siRNA-mediated knockdown of IRF1 completely reversed elevated pro-inflammatory target gene expression in CITED2-deficient macrophages. Collectively, our study findings demonstrate that CITED2 restrains the STAT1-IRF1 signaling axis in macrophages and limits the development of atherosclerotic plaques.
Collapse
Affiliation(s)
- Atif Zafar
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Hang Pong Ng
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rachel Diamond-Zaluski
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Gun-Dong Kim
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ernest Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sally L Dunwoodie
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia.,Faculties of Medicine and Science, UNSW Sydney, Sydney, NSW, Australia
| | - Jonathan D Smith
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | | |
Collapse
|
18
|
Chen MY, Ye XJ, He XH, Ouyang DY. The Signaling Pathways Regulating NLRP3 Inflammasome Activation. Inflammation 2021; 44:1229-1245. [PMID: 34009550 DOI: 10.1007/s10753-021-01439-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/30/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023]
Abstract
The NLRP3 inflammasome is a multi-molecular complex that acts as a molecular platform to mediate caspase-1 activation, leading to IL-1β/IL-18 maturation and release in cells stimulated by various pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). This inflammasome plays an important role in the innate immunity as its activation can further promote the occurrence of inflammation, enhance the ability of host to remove pathogens, and thus facilitate the repair of injured tissues. But if the inflammasome activation is dysregulated, it will cause the development of various inflammatory diseases and metabolic disorders. Therefore, under normal conditions, the activation of inflammasome is tightly regulated by various positive and negative signaling pathways to respond to the stimuli without damaging the host itself while maintaining homeostasis. In this review, we summarize recent advances in the major signaling pathways (including TLRs, MAPK, mTOR, autophagy, PKA, AMPK, and IFNR) that regulate NLRP3 inflammasome activation, providing a brief view of the molecular network that regulates this inflammasome as a theoretical basis for therapeutic intervention of NLRP3 dysregulation-related diseases.
Collapse
Affiliation(s)
- Ming-Ye Chen
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xun-Jia Ye
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xian-Hui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Dong-Yun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
19
|
Souza COS, Ketelut-Carneiro N, Milanezi CM, Faccioli LH, Gardinassi LG, Silva JS. NLRC4 inhibits NLRP3 inflammasome and abrogates effective antifungal CD8 + T cell responses. iScience 2021; 24:102548. [PMID: 34142053 PMCID: PMC8184506 DOI: 10.1016/j.isci.2021.102548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/06/2021] [Accepted: 05/14/2021] [Indexed: 12/12/2022] Open
Abstract
The recognition of fungi by intracellular NOD-like receptors (NLRs) induces inflammasome assembly and activation. Although the NLRC4 inflammasome has been extensively studied in bacterial infections, its role during fungal infections is unclear. Paracoccidioidomycosis (PCM) is a pathogenic fungal disease caused by Paracoccidioides brasiliensis. Here, we show that NLRC4 confers susceptibility to experimental PCM by regulating NLRP3-dependent cytokine production and thus protective effector mechanisms. Early after infection, NLRC4 suppresses prostaglandin E2 production, and consequently reduces interleukin (IL)-1β release by macrophages and dendritic cells in the lungs. IL-1β is required to control fungal replication via induction of the nitric oxide synthase 2 (NOS2) pathway. At a later stage of the disease, NLRC4 impacts IL-18 release, dampening robust CD8+IFN-γ+ T cell responses and enhancing mortality of mice. These findings demonstrate that NLRC4 promotes disease by regulating the production of inflammatory cytokines and cellular responses that depend on the NLRP3 inflammasome activity. NLRC4 promotes susceptibility to a highly pathogenic fungus. NLRC4 regulates NLRP3 activity. NLRC4 inhibits early NLRP3/IL-1β/NOS2/NO axis and promotes fungal replication. NLRC4 dampens late IL-18 production, suppressing CD8+IFN-γ+ T cell responses.
Collapse
Affiliation(s)
- Camila O S Souza
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Natália Ketelut-Carneiro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Program in Innate Immunity, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Cristiane M Milanezi
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Lúcia H Faccioli
- Department of Clinical Analyses, Toxicology and Bromatological Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luiz G Gardinassi
- Department of Biosciences and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
| | - João S Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Fiocruz-Bi-Institutional Translational Medicine Platform, Ribeirão Preto, SP, Brazil
| |
Collapse
|
20
|
Structural and biochemical mechanisms of NLRP1 inhibition by DPP9. Nature 2021; 592:773-777. [PMID: 33731929 PMCID: PMC8081665 DOI: 10.1038/s41586-021-03320-w] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/01/2021] [Indexed: 11/19/2022]
Abstract
Nucleotide-binding domain, leucine-rich repeat receptors (NLRs) mediate innate immunity by forming inflammasomes. Activation of the NLR protein NLRP1 requires autocleavage within its function-to-find domain (FIIND)1–7. In resting cells, the dipeptidyl peptidases DPP8 and DPP9 interact with the FIIND of NLRP1 and suppress spontaneous NLRP1 activation8,9; however, the mechanisms through which this occurs remain unknown. Here we present structural and biochemical evidence that full-length rat NLRP1 (rNLRP1) and rat DPP9 (rDPP9) form a 2:1 complex that contains an autoinhibited rNLRP1 molecule and an active UPA–CARD fragment of rNLRP1. The ZU5 domain is required not only for autoinhibition of rNLRP1 but also for assembly of the 2:1 complex. Formation of the complex prevents UPA-mediated higher-order oligomerization of UPA–CARD fragments and strengthens ZU5-mediated NLRP1 autoinhibition. Structure-guided biochemical and functional assays show that both NLRP1 binding and enzymatic activity are required for DPP9 to suppress NLRP1 in human cells. Together, our data reveal the mechanism of DPP9-mediated inhibition of NLRP1 and shed light on the activation of the NLRP1 inflammasome. The 2:1 complex between the inflammatory mediator NLRP1 and the dipeptidyl peptidase DPP9 functions to sequester an inflammasome-forming fragment of NLRP1 and enhance NLRP1 autoinhibition.
Collapse
|
21
|
Di Q, Zhao X, Zhang R, Ma X, Liang X, Li X, Gao J, Tang H, Chen W, Xiao W. Novel clerodane-type diterpenoid Cintelactone A suppresses lipopolysaccharide -induced inflammation by promoting ubiquitination, proteasomal degradation of TRAF6. Pharmacol Res 2021; 164:105386. [PMID: 33352228 DOI: 10.1016/j.phrs.2020.105386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/29/2022]
Abstract
Cellular inflammation is the underlying cause of several diseases and development of a safe and effective anti-inflammatory drug is need-of-the hour for treatment of diseases like lung inflammation. Callicarpa integerrima Champ. is a well-known herbal medicine with hemostatic and anti-inflammatory functions. However, the exact ingredient exhibiting anti-inflammatory activity in C. integerrima Champ. is largely unknown. Here, we first isolated, purified and characterized a novel clerodane-type diterpenoid Cintelactone A (CA) from C. integerrima Champ. We demonstrated that CA could significantly inhibit lipopolysaccharide (LPS)-induced pro-inflammatory cytokines and mediators production both in mouse peritoneal macrophages and THP1 cells. Consistently, CA also relieved inflammation and reduced LPS-induced lung injury in mice. We systematically elucidated the mechanism of action as well. CA interacted with Arg78 of tumor necrosis factor receptor-associated factor 6 (TRAF6) by hydrogen bonding. It further promoted the K48-linked ubiquitination and proteasomal degradation of TRAF6, and suppressed the activation of NF-κB and MAPKs signaling pathways. Collectively, our study reveals that new clerodane-type diterpenoid CA suppresses LPS-induced inflammation by promoting TRAF6 degradation, suggesting that CA as the potential therapeutic candidate for the treatment of inflammation associated diseases.
Collapse
Affiliation(s)
- Qianqian Di
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Xibao Zhao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Ruihan Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, China
| | - Xingyu Ma
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Xinxin Liang
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, China
| | - Xiaoli Li
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, China
| | - Junbo Gao
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, China
| | - Haimei Tang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Weilin Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, 518060, China.
| | - Weilie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, China.
| |
Collapse
|
22
|
Luo Y, Xiong B, Liu H, Chen Z, Huang H, Yu C, Yang J. Koumine Suppresses IL-1β Secretion and Attenuates Inflammation Associated With Blocking ROS/NF-κB/NLRP3 Axis in Macrophages. Front Pharmacol 2021; 11:622074. [PMID: 33542692 PMCID: PMC7851739 DOI: 10.3389/fphar.2020.622074] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/15/2020] [Indexed: 12/21/2022] Open
Abstract
Koumine (KM), one of the primary constituents of Gelsemium elegans, has been used for the treatment of inflammatory diseases such as rheumatoid arthritis, but whether KM impacts the activation of the NOD-like receptor protein 3 (NLRP3) inflammasome remains unknown. This study aimed to explore the inhibitory effect of KM on NLRP3 inflammasome activation and the underlying mechanisms both in vitro using macrophages stimulated with LPS plus ATP, nigericin or monosodium urate (MSU) crystals and in vivo using an MSU-induced peritonitis model. We found that KM dose-dependently inhibited IL-1β secretion in macrophages after NLRP3 inflammasome activators stimulation. Furthermore, KM treatment efficiently attenuated the infiltration of neutrophils and suppressed IL-1β production in mice with MSU-induced peritonitis. These results indicated that KM inhibited NLRP3 inflammasome activation, and consistent with this finding, KM effectively inhibited caspase-1 activation, mature IL-1β secretion, NLRP3 formation and pro-IL-1β expression in LPS-primed macrophages treated with ATP, nigericin or MSU. The mechanistic study showed that, KM exerted a potent inhibitory effect on the NLRP3 priming step, which decreased the phosphorylation of IκBα and p65, the nuclear localization of p65, and the secretion of TNF-α and IL-6. Moreover, the assembly of NLRP3 was also interrupted by KM. KM blocked apoptosis-associated speck-like protein containing a CARD (ASC) speck formation and its oligomerization and hampered the NLRP3-ASC interaction. This suppression was attributed to the ability of KM to inhibit the production of reactive oxygen species (ROS). In support of this finding, the inhibitory effect of KM on ROS production was completely counteracted by H2O2, an ROS promoter. Our results provide the first indication that KM exerts an inhibitory effect on NLRP3 inflammasome activation associated with blocking the ROS/NF-κB/NLRP3 signal axis. KM might have potential clinical application in the treatment of NLRP3 inflammasome-related diseases.
Collapse
Affiliation(s)
- Yufei Luo
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Bojun Xiong
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Haiping Liu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Zehong Chen
- Experimental Teaching Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Huihui Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Changxi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Jian Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
23
|
Moustafa SF, Gabr NM, Zaki JT, El Awdan SA, Mina SA. The anti-inflammatory, anti-ulcer activities and phytochemical investigation of Cucumis melo L. cv. Ismailawi fruits. Nat Prod Res 2020; 35:5934-5938. [PMID: 32772557 DOI: 10.1080/14786419.2020.1803314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
This study aims to evaluate the anti-inflammatory and anti-ulcer activities of Cucumis melo L. cv. Ismailawi fruits, as well as the investigation of the phenolic content and lipoidal matter composition via high performance liquid chromatography and gas chromatography coupled to mass spectrometry respectively. Both the petroleum ether and defatted methanol extracts of the fruit pulp showed 63.13% and 54.97% decrease in oedema volume respectively after 4 h in comparison to indomethacin standard drug. Both the petroleum ether extract and ethyl acetate fractions at a dose (200 mg/kg) showed significant anti-ulcer activity decreasing both ulcer number and severity in comparison to ranitidine as standard drug. Histopathological investigation further confirmed these results. Moreover; this is the first report for the investigation of the phenolic content and the lipoidal matter of Cucumis melo L. cv. Ismailawi fruits where methyl palmatate, gallic acid and rutin represented the major detected components.
Collapse
Affiliation(s)
- Sherifa F Moustafa
- Pharmacognosy Department, Faculty of Pharmacy, 6th October University, Giza, Egypt.,Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nagwan M Gabr
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Jaky T Zaki
- Pharmacognosy Department, Faculty of Pharmacy, 6th October University, Giza, Egypt
| | - Sally A El Awdan
- Pharmacology Department, Medical Division, National Research Centre, Giza, Egypt
| | - Suzan A Mina
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| |
Collapse
|
24
|
Ming T, Yuan M, Kong Q, Huang Q, Xia Z, Wu X. Dexmedetomidine alleviates blunt chest trauma and hemorrhagic shock‑resuscitation‑induced acute lung injury through inhibiting the NLRP3 inflammasome. Mol Med Rep 2020; 22:2507-2515. [PMID: 32705267 PMCID: PMC7411430 DOI: 10.3892/mmr.2020.11335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Blunt chest trauma with hemorrhagic shock frequently induces pulmonary inflammation that leads to acute lung injury (ALI). The present study aimed to explore the protective effects of dexmedetomidine (Dex) in blunt chest trauma and hemorrhagic shock-resuscitation (THSR)-induced ALI by mediating nucleotide binding and oligomerization domain-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome formation in rats. An ALI model in rats induced by THSR was constructed and Dex was administered intraperitoneally (5 µg/kg/h) immediately after blunt chest trauma. Blood samples were collected for the determination of proinflammatory factor levels, and lung tissue specimens were harvested for wet/dry (W/D) weight ratio, hematoxylin and eosin staining, and transmission electron microscopy analyses. Additionally, malondialdehyde (MDA), superoxide dismutase (SOD), lactate dehydrogenase (LDH) and myeloperoxidase (MPO) activity were evaluated, and the expression of protein in lung tissues was examined via western blot analysis. Compared with the sham group, pathological alterations in the ALI group and the W/D ratios were significantly increased. MDA, LDH and MPO activity, and the levels of interleukin (IL)-1β, IL-18, IL-6 and tumor necrosis factor-α were significantly elevated. NLRP3, apoptosis-associated speck-like protein containing a caspase recruitment domain and caspase-1 expression was significantly increased. Conversely, Dex treatment significantly reversed these changes. The present study demonstrated that by reducing inflammatory responses, Dex exerted protective effects against THSR-ALI in rats, potentially via the inhibition of NLRP3 signaling pathways.
Collapse
Affiliation(s)
- Tingqian Ming
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Min Yuan
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qian Kong
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qin Huang
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
25
|
Huang J, Lu W, Doycheva DM, Gamdzyk M, Hu X, Liu R, Zhang JH, Tang J. IRE1α inhibition attenuates neuronal pyroptosis via miR-125/NLRP1 pathway in a neonatal hypoxic-ischemic encephalopathy rat model. J Neuroinflammation 2020; 17:152. [PMID: 32375838 PMCID: PMC7203836 DOI: 10.1186/s12974-020-01796-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inhibition of inositol-requiring enzyme-1 alpha (IRE1α), one of the sensor signaling proteins associated with endoplasmic reticulum (ER) stress, has been shown to alleviate brain injury and improve neurological behavior in a neonatal hypoxic-ischemic encephalopathy (HIE) rat model. However, there is no information about the role of IRE1α inhibitor as well as its molecular mechanisms in preventing neuronal pyroptosis induced by NLRP1 (NOD-, LRR- and pyrin domain-containing 1) inflammasome. In the present study, we hypothesized that IRE1α can degrade microRNA-125-b-2-3p (miR-125-b-2-3p) and activate NLRP1/caspased-1 pathway, and subsequently promote neuronal pyroptosis in HIE rat model. METHODS Ten-day old unsexed rat pups were subjected to hypoxia-ischemia (HI) injury, and the inhibitor of IRE1α, STF083010, was administered intranasally at 1 h after HI induction. AntimiR-125 or NLRP1 activation CRISPR was administered by intracerebroventricular (i.c.v) injection at 24 h before HI induction. Immunofluorescence staining, western blot analysis, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), brain infarct volume measurement, neurological function tests, and Fluoro-Jade C staining were performed. RESULTS Endogenous phosphorylated IRE1α (p-IRE1α), NLRP1, cleaved caspase-1, interleukin-1β (IL-1β), and interleukin-18 (IL-18) were increased and miR-125-b-2-3p was decreased in HIE rat model. STF083010 administration significantly upregulated the expression of miR-125-b-2-3p, reduced the infarct volume, improved neurobehavioral outcomes and downregulated the protein expression of NLRP1, cleaved caspase-1, IL-1β and IL-18. The protective effects of STF083010 were reversed by antimiR-125 or NLRP1 activation CRISPR. CONCLUSIONS IRE1α inhibitor, STF083010, reduced neuronal pyroptosis at least in part via miR-125/NLRP1/caspase-1 signaling pathway after HI.
Collapse
Affiliation(s)
- Juan Huang
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
| | - Weitian Lu
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
| | - Desislava Met Doycheva
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
| | - Marcin Gamdzyk
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
| | - Xiao Hu
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Rui Liu
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
- Department of Anesthesiology, Loma Linda University, Loma Linda, CA, 92350, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA.
| |
Collapse
|
26
|
Feng Z, Lu X, Gan L, Zhang Q, Lin L. Xanthones, A Promising Anti-Inflammatory Scaffold: Structure, Activity, and Drug Likeness Analysis. Molecules 2020; 25:E598. [PMID: 32019180 PMCID: PMC7037265 DOI: 10.3390/molecules25030598] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/30/2022] Open
Abstract
Inflammation is the body's self-protective response to multiple stimulus, from external harmful substances to internal danger signals released after trauma or cell dysfunction. Many diseases are considered to be related to inflammation, such as cancer, metabolic disorders, aging, and neurodegenerative diseases. Current therapeutic approaches include mainly non-steroidal anti-inflammatory drugs and glucocorticoids, which are generally of limited effectiveness and severe side-effects. Thus, it is urgent to develop novel effective anti-inflammatory therapeutic agents. Xanthones, a unique scaffold with a 9H-Xanthen-9-one core structure, widely exist in natural sources. Till now, over 250 xanthones were isolated and identified in plants from the families Gentianaceae and Hypericaceae. Many xanthones have been disclosed with anti-inflammatory properties on different models, either in vitro or in vivo. Herein, we provide a comprehensive and up-to-date review of xanthones with anti-inflammatory properties, and analyzed their drug likeness, which might be potential therapeutic agents to fight against inflammation-related diseases.
Collapse
Affiliation(s)
- Zheling Feng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China; (Z.F.); (Q.Z.)
| | - Xiuqiang Lu
- Fuqing Branch of Fujian Normal University, Fuzhou 350300, China;
| | - Lishe Gan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China;
| | - Qingwen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China; (Z.F.); (Q.Z.)
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China; (Z.F.); (Q.Z.)
| |
Collapse
|
27
|
Hidalgo-Lanussa O, Baez-Jurado E, Echeverria V, Ashraf GM, Sahebkar A, Garcia-Segura LM, Melcangi RC, Barreto GE. Lipotoxicity, neuroinflammation, glial cells and oestrogenic compounds. J Neuroendocrinol 2020; 32:e12776. [PMID: 31334878 DOI: 10.1111/jne.12776] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/16/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023]
Abstract
The high concentrations of free fatty acids as a consequence of obesity and being overweight have become risk factors for the development of different diseases, including neurodegenerative ailments. Free fatty acids are strongly related to inflammatory events, causing cellular and tissue alterations in the brain, including cell death, deficits in neurogenesis and gliogenesis, and cognitive decline. It has been reported that people with a high body mass index have a higher risk of suffering from Alzheimer's disease. Hormones such as oestradiol not only have beneficial effects on brain tissue, but also exert some adverse effects on peripheral tissues, including the ovary and breast. For this reason, some studies have evaluated the protective effect of oestrogen receptor (ER) agonists with more specific tissue activities, such as the neuroactive steroid tibolone. Activation of ERs positively affects the expression of pro-survival factors and cell signalling pathways, thus promoting cell survival. This review aims to discuss the relationship between lipotoxicity and the development of neurodegenerative diseases. We also elaborate on the cellular and molecular mechanisms involved in neuroprotection induced by oestrogens.
Collapse
Affiliation(s)
- Oscar Hidalgo-Lanussa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Eliana Baez-Jurado
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Valentina Echeverria
- Facultad de Ciencias de la Salud, Universidad San Sebastián, Concepción, Chile
- Bay Pines VA Healthcare System, Research and Development, Bay Pines, FL, USA
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, CSIC, Madrid, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Roberto C Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| |
Collapse
|
28
|
Ramírez-Serrano CE, Jiménez-Ferrer E, Herrera-Ruiz M, Zamilpa A, Vargas-Villa G, Ramírez-Carreto RJ, Chavarría A, Tortoriello J, Pedraza-Alva G, Pérez-Martínez L. A Malva parviflora´s fraction prevents the deleterious effects resulting from neuroinflammation. Biomed Pharmacother 2019; 118:109349. [DOI: 10.1016/j.biopha.2019.109349] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 07/18/2019] [Accepted: 08/07/2019] [Indexed: 01/23/2023] Open
|
29
|
Dia VP, Bradwell J, Pangloli P. Sorghum Phenolics Inhibits Inflammasomes in Lipopolysaccharide (LPS)-Primed and Adenosine Triphosphate (ATP)-Activated Macrophages. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2019; 74:307-315. [PMID: 31104201 DOI: 10.1007/s11130-019-00736-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Sorghum contains phenolic compounds with reported biological activities. The objective was to evaluate the ability of sorghum phenolic extract to inhibit inflammasomes in THP-1 human macrophages. THP-1 human macrophages was pre-treated with sorghum phenolics and the inflammasome was activated by lipopolysaccharide and adenosine triphosphate treatment. Treatment of macrophages with 50 μg sorghum extract/mL reduced IL-1β and IL-18 secretion by 59.7 and 32.0%, respectively, associated with caspase-1 activity reduction. Moreover, the production of intracellular reactive oxygen species was reduced. Our data showed the potential role of sorghum phenolics in diseases associated with aberrant inflammasomes activation.
Collapse
Affiliation(s)
- Vermont P Dia
- Department of Food Science, The University of Tennessee, Knoxville, TN, USA.
| | - Jordan Bradwell
- Department of Food Science, The University of Tennessee, Knoxville, TN, USA
| | - Philipus Pangloli
- Department of Food Science, The University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
30
|
Medrano-Jiménez E, Jiménez-Ferrer Carrillo I, Pedraza-Escalona M, Ramírez-Serrano CE, Álvarez-Arellano L, Cortés-Mendoza J, Herrera-Ruiz M, Jiménez-Ferrer E, Zamilpa A, Tortoriello J, Pedraza-Alva G, Pérez-Martínez L. Malva parviflora extract ameliorates the deleterious effects of a high fat diet on the cognitive deficit in a mouse model of Alzheimer's disease by restoring microglial function via a PPAR-γ-dependent mechanism. J Neuroinflammation 2019; 16:143. [PMID: 31291963 PMCID: PMC6617588 DOI: 10.1186/s12974-019-1515-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 06/10/2019] [Indexed: 12/12/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a neuropathology strongly associated with the activation of inflammatory pathways. Accordingly, inflammation resulting from obesity exacerbates learning and memory deficits in humans and in animal models of AD. Consequently, the long-term use of non-steroidal anti-inflammatory agents diminishes the risk for developing AD, but the side effects produced by these drugs limit their prophylactic use. Thus, plants natural products have become an excellent option for modern therapeutics. Malva parviflora is a plant well known for its anti-inflammatory properties. Methods The present study was aimed to determine the anti-inflammatory potential of M. parviflora leaf hydroalcoholic extract (MpHE) on AD pathology in lean and obese transgenic 5XFAD mice, a model of familial AD. The inflammatory response and Amyloid β (Aβ) plaque load in lean and obese 5XFAD mice untreated or treated with MpHE was evaluated by immunolocalization (Iba-1 and GFAP) and RT-qPCR (TNF) assays and thioflavin-S staining, respectively. Spatial learning memory was assessed by the Morris Water Maze behavioral test. Microglia phagocytosis capacity was analyzed in vivo and by ex vivo and in vitro assays, and its activation by morphological changes (phalloidin staining) and expression of CD86, Mgl1, and TREM-2 by RT-qPCR. The mechanism triggered by the MpHE was characterized in microglia primary cultures and ex vivo assays by immunoblot (PPAR-γ) and RT-qPCR (CD36) and in vivo by flow cytometry, using GW9662 (PPAR-γ inhibitor) and pioglitazone (PPAR-γ agonist). The presence of bioactive compounds in the MpHE was determined by HPLC. Results MpHE efficiently reduced astrogliosis, the presence of insoluble Aβ peptides in the hippocampus and spatial learning impairments, of both, lean, and obese 5XFAD mice. This was accompanied by microglial cells accumulation around Aβ plaques in the cortex and the hippocampus and decreased expression of M1 inflammatory markers. Consistent with the fact that the MpHE rescued microglia phagocytic capacity via a PPAR-γ/CD36-dependent mechanism, the MpHE possess oleanolic acid and scopoletin as active phytochemicals. Conclusions M. parviflora suppresses neuroinflammation by inhibiting microglia pro-inflammatory M1 phenotype and promoting microglia phagocytosis. Therefore, M. parviflora phytochemicals represent an alternative to prevent cognitive impairment associated with a metabolic disorder as well as an effective prophylactic candidate for AD progression. Electronic supplementary material The online version of this article (10.1186/s12974-019-1515-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elisa Medrano-Jiménez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Itzia Jiménez-Ferrer Carrillo
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Martha Pedraza-Escalona
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Cristina E Ramírez-Serrano
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Lourdes Álvarez-Arellano
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México.,Present address: CONACYT-Hospital Infantil de México Federico Gómez, CP 06720, Ciudad de México, México
| | - Javier Cortés-Mendoza
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Maribel Herrera-Ruiz
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina No. 1, CP 62790, Xochitepec, Morelos, México
| | - Enrique Jiménez-Ferrer
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina No. 1, CP 62790, Xochitepec, Morelos, México
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina No. 1, CP 62790, Xochitepec, Morelos, México
| | - Jaime Tortoriello
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina No. 1, CP 62790, Xochitepec, Morelos, México
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), A.P. 510-3, CP, 62210, Cuernavaca, Morelos, México.
| |
Collapse
|
31
|
Pedraza-Alva G, Ramírez-Serrano CE, Pedraza F, Flores-Vallejo RDC, Villarreal ML, Pérez-Martínez L. From traditional remedies to cutting-edge medicine: Using ancient mesoamerican knowledge to address complex disorders relevant to psychoneuroimmunology. Brain Behav Immun 2019; 79:3-5. [PMID: 30980951 DOI: 10.1016/j.bbi.2019.04.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 10/27/2022] Open
Affiliation(s)
- Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos 62210, Mexico
| | - Cristina E Ramírez-Serrano
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos 62210, Mexico
| | - Fernando Pedraza
- Department of Evolutionary Biology and Environmental Studies, University of Zurich, Zurich 8057, Switzerland
| | - Rosario Del Carmen Flores-Vallejo
- Laboratorio de Investigación de Plantas Medicinales, Centro de Investigación en Biotecnología (CEIB), Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca, Morelos 62209, Mexico
| | - María Luisa Villarreal
- Laboratorio de Investigación de Plantas Medicinales, Centro de Investigación en Biotecnología (CEIB), Universidad Autónoma del Estado de Morelos (UAEM), Cuernavaca, Morelos 62209, Mexico
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos 62210, Mexico.
| |
Collapse
|
32
|
Ezzat SM, Raslan M, Salama MM, Menze ET, El Hawary SS. In vivo anti-inflammatory activity and UPLC-MS/MS profiling of the peels and pulps of Cucumis melo var. cantalupensis and Cucumis melo var. reticulatus. JOURNAL OF ETHNOPHARMACOLOGY 2019; 237:245-254. [PMID: 30857984 DOI: 10.1016/j.jep.2019.03.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 03/05/2019] [Accepted: 03/05/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cucumis melo var. cantalupensis and Cucumis melo var. reticulatus are the most famous varieties of netted muskmelon or cantaloupe in Egypt. Cantaloupe has a great reputation as an anti-inflammatory drug for hot inflammation of liver, cough, eczema, and kidney disorders such as ulcers in the urinary tract, and our objective was to confirm this use scientifically. MATERIALS AND METHODS Inflammation was induced in adult male Sprague Dawley rats by subcutaneous injection of 0.05 ml of carrageenan (1% solution in saline) into the plantar surface of the right hind paw 30 min after oral pretreatment of the rats with 95% ethanolic extracts of Cucumis melo var. cantalupensis peels (CCP) and pulps (CCU) and Cucumis melo var. reticulatus peels (CRP) and pulps (CRU) at doses of 25 and 50 mg/kg. Indomethacin (10 mg/kg) was used as a standard drug. The effect of the tested samples was measured on the oedema volume, as well as PGE-2, TNF-α, IL-6 and IL-1β levels. Metabolic profiling of the extracts was performed using UPLC-MS/MS analysis. RESULTS Pretreatment of rats with the ethanol extract of the pulps and peels of the two varieties at doses of 25 and 50 mg/kg significantly inhibited the carrageenan-induced increase in the oedema volume of the rat paws after 3 h, except for the low dose of the French cantaloupe pulp. CRP at 50 mg/kg caused the most significant reductions in both TNF-α (P < 0.05) and IL-1β (P < 0.001) levels, while CCP caused the most significant reductions in PGE-2 and IL-6 (P < 0.05) levels. Increases in PGE-2, TNF-α, IL-6 and IL-1β levels were also significantly prevented by indomethacin (10 mg/kg). UPLC-MS/MS facilitated the identification of 44 phenolic compounds, including phenolic acids and flavonoids. CONCLUSION This is the first report of the chemical and biological study of the peels of Cucumis melo var. cantalupensis and Cucumis melo var. reticulatus.
Collapse
Affiliation(s)
- Shahira M Ezzat
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, October University for Modern Science and Arts (MSA), 6th October, 12566, Egypt
| | - Mai Raslan
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, 62511, Egypt.
| | - Maha M Salama
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt; Department of Pharmacognosy, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Seham S El Hawary
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
33
|
Leu WJ, Chen JC, Guh JH. Extract From Plectranthus amboinicus Inhibit Maturation and Release of Interleukin 1β Through Inhibition of NF-κB Nuclear Translocation and NLRP3 Inflammasome Activation. Front Pharmacol 2019; 10:573. [PMID: 31191313 PMCID: PMC6546882 DOI: 10.3389/fphar.2019.00573] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/06/2019] [Indexed: 01/10/2023] Open
Abstract
Uncontrolled inflammation may produce massive inflammatory cytokines, in which interleukin 1β (IL-1β) plays a key role, resulting in tissue damage and serious disorders. The activation of NLRP3 inflammasome is one of the major mechanisms in maturation and release of IL-1β. Plectranthus amboinicus is a perennial herb. Several pharmacological activities of natural components and crude extracts from P. amboinicus have been reported including anti-inflammation; however, the underlying mechanism is not clear. Phorbol-12-myristate 13-acetate-differentiated THP-1 monocytic leukemia cells were used as a reliable model in this study to examine the effect on inflammasome signaling pathway by PA-F4, an extract from Plectranthus amboinicus. PA-F4 inhibited ATP-induced release of caspase-1, IL-1β, and IL-18 from lipopolysaccharides (LPS)-primed cells. PA-F4 induced a concentration-dependent inhibition of both ASC dimerization and oligomerization in cells under LPS priming plus ATP stimulation. Co-immunoprecipitation of NLRP3 and ASC demonstrated that PA-F4 significantly blunted the interaction between NLRP3 and ASC. Furthermore, PA-F4 completely abolished ATP-induced K+ efflux reaction in LPS-primed cells. Taken together, PA-F4 displayed an inhibitory activity on NLRP3 inflammasome activation. Moreover, PA-F4 also inhibited LPS-induced p65 NF-κB activation, suggesting an inhibitory activity on LPS priming step. Further identification showed that rosmarinic acid, cirsimaritin, salvigenin, and carvacrol, four constituents in PA-F4, inhibited LPS-induced IL-6 release. In contrast, rosmarinic acid, cirsimaritin and carvacrol but not salvigenin inhibited ATP-induced caspase-1 release from LPS-primed cells. In conclusion, PA-F4 displayed an inhibitory activity on activation of NLRP3 inflammasome. PA-F4 inhibited LPS priming step through block of p65 NF-κB activation. It also inhibited ATP-induced signaling pathways in LPS-primed cells including the inhibition of both ASC dimerization and oligomerization, K+ efflux reaction, and the release reaction of caspase-1, IL-1β, and IL-18. Rosmarinic acid, cirsimaritin, salvigenin, and carvacrol could partly explain PA-F4-mediated inhibitory activity on blocking the activation of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Wohn-Jenn Leu
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | | | - Jih-Hwa Guh
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
34
|
SERPINB1-mediated checkpoint of inflammatory caspase activation. Nat Immunol 2019; 20:276-287. [PMID: 30692621 DOI: 10.1038/s41590-018-0303-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/14/2018] [Indexed: 02/07/2023]
Abstract
Inflammatory caspases (caspase-1, caspase-4, caspase-5 and caspase-11 (caspase-1/-4/-5/-11)) mediate host defense against microbial infections, processing pro-inflammatory cytokines and triggering pyroptosis. However, precise checkpoints are required to prevent their unsolicited activation. Here we report that serpin family B member 1 (SERPINB1) limited the activity of those caspases by suppressing their caspase-recruitment domain (CARD) oligomerization and enzymatic activation. While the reactive center loop of SERPINB1 inhibits neutrophil serine proteases, its carboxy-terminal CARD-binding motif restrained the activation of pro-caspase-1/-4/-5/-11. Consequently, knockdown or deletion of SERPINB1 prompted spontaneous activation of caspase-1/-4/-5/-11, release of the cytokine IL-1β and pyroptosis, inducing elevated inflammation after non-hygienic co-housing with pet-store mice and enhanced sensitivity to lipopolysaccharide- or Acinetobacter baumannii-induced endotoxemia. Our results reveal that SERPINB1 acts as a vital gatekeeper of inflammation by restraining neutrophil serine proteases and inflammatory caspases in a genetically and functionally separable manner.
Collapse
|
35
|
Mitochondrial Neuroglobin Is Necessary for Protection Induced by Conditioned Medium from Human Adipose-Derived Mesenchymal Stem Cells in Astrocytic Cells Subjected to Scratch and Metabolic Injury. Mol Neurobiol 2018; 56:5167-5187. [PMID: 30536184 DOI: 10.1007/s12035-018-1442-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022]
Abstract
Astrocytes are specialized cells capable of regulating inflammatory responses in neurodegenerative diseases or traumatic brain injury. In addition to playing an important role in neuroinflammation, these cells regulate essential functions for the preservation of brain tissue. Therefore, the search for therapeutic alternatives to preserve these cells and maintain their functions contributes in some way to counteract the progress of the injury and maintain neuronal survival in various brain pathologies. Among these strategies, the conditioned medium from human adipose-derived mesenchymal stem cells (CM-hMSCA) has been reported with a potential beneficial effect against several neuropathologies. In this study, we evaluated the potential effect of CM-hMSCA in a model of human astrocytes (T98G cells) subjected to scratch injury. Our findings demonstrated that CM-hMSCA regulates the cytokines IL-2, IL-6, IL-8, IL-10, GM-CSF, and TNF-α, downregulates calcium at the cytoplasmic level, and regulates mitochondrial dynamics and the respiratory chain. These actions are accompanied by modulation of the expression of different proteins involved in signaling pathways such as AKT/pAKT and ERK1/2/pERK, and may mediate the localization of neuroglobin (Ngb) at the cellular level. We also confirmed that Ngb mediated the protective effects of CM-hMSCA through regulation of proteins involved in survival pathways and oxidative stress. In conclusion, regulation of brain inflammation combined with the recovery of fundamental cellular aspects in the face of injury makes CM-hMSCA a promising candidate for the protection of astrocytes in brain pathologies.
Collapse
|
36
|
Ramachandran RA, Lupfer C, Zaki H. The Inflammasome: Regulation of Nitric Oxide and Antimicrobial Host Defence. Adv Microb Physiol 2018; 72:65-115. [PMID: 29778217 DOI: 10.1016/bs.ampbs.2018.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) is a gaseous signalling molecule that plays diverse physiological functions including antimicrobial host defence. During microbial infection, NO is synthesized by inducible NO synthase (iNOS), which is expressed by host immune cells through the recognition of microbial pattern molecules. Therefore, sensing pathogens or their pattern molecules by pattern recognition receptors (PRRs), which are located at the cell surface, endosomal and phagosomal compartment, or in the cytosol, is key in inducing iNOS and eliciting antimicrobial host defence. A group of cytosolic PRRs is involved in inducing NO and other antimicrobial molecules by forming a molecular complex called the inflammasome. Assembled inflammasomes activate inflammatory caspases, such as caspase-1 and caspase-11, which in turn process proinflammatory cytokines IL-1β and IL-18 into their mature forms and induce pyroptotic cell death. IL-1β and IL-18 play a central role in immunity against microbial infection through activation and recruitment of immune cells, induction of inflammatory molecules, and regulation of antimicrobial mediators including NO. Interestingly, NO can also regulate inflammasome activity in an autocrine and paracrine manner. Here, we discuss molecular mechanisms of inflammasome formation and the inflammasome-mediated regulation of host defence responses during microbial infections.
Collapse
Affiliation(s)
| | | | - Hasan Zaki
- UT Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
37
|
Suzuki S, Suzuki T, Mimuro H, Mizushima T, Sasakawa C. Shigella hijacks the glomulin-cIAPs-inflammasome axis to promote inflammation. EMBO Rep 2017; 19:89-101. [PMID: 29191979 DOI: 10.15252/embr.201643841] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 10/25/2017] [Accepted: 11/06/2017] [Indexed: 11/09/2022] Open
Abstract
Shigella deploys a unique mechanism to manipulate macrophage pyroptosis by delivering the IpaH7.8 E3 ubiquitin ligase via its type III secretion system. IpaH7.8 ubiquitinates glomulin (GLMN) and elicits its degradation, thereby inducing inflammasome activation and pyroptotic cell death of macrophages. Here, we show that GLMN specifically binds cellular inhibitor of apoptosis proteins 1 and 2 (cIAP1 and cIAP2), members of the inhibitor of apoptosis (IAP) family of RING-E3 ligases, which results in reduced E3 ligase activity, and consequently inflammasome-mediated death of macrophages. Importantly, reducing the levels of GLMN in macrophages via IpaH7.8, or siRNA-mediated knockdown, enhances inflammasome activation in response to infection by Shigella, Salmonella, or Pseudomonas, stimulation with NLRP3 inflammasome activators (including SiO2, alum, or MSU), or stimulation of the AIM2 inflammasome by poly dA:dT GLMN binds specifically to the RING domain of both cIAPs, which inhibits their self-ubiquitination activity. These findings suggest that GLMN is a negative regulator of cIAP-mediated inflammasome activation, and highlight a unique Shigella stratagem to kill macrophages, promoting severe inflammation.
Collapse
Affiliation(s)
- Shiho Suzuki
- Division of Bacterial Infection Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan .,Department of Bacterial Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshihiko Suzuki
- Department of Bacterial Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hitomi Mimuro
- Division of Bacteriology, Department of Infectious Diseases Control, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Infection Microbiology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tsunehiro Mizushima
- Picobiology Institute, Graduate School of Life Science, University of Hyogo, Hyogo, Japan
| | - Chihiro Sasakawa
- Division of Bacterial Infection Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan .,Medical Mycology Research Center, Chiba University, Chiba, Japan.,Nippon Institute for Biological Science, Tokyo, Japan
| |
Collapse
|
38
|
Pretorius E, Mbotwe S, Bester J, Robinson CJ, Kell DB. Acute induction of anomalous and amyloidogenic blood clotting by molecular amplification of highly substoichiometric levels of bacterial lipopolysaccharide. J R Soc Interface 2017; 13:rsif.2016.0539. [PMID: 27605168 PMCID: PMC5046953 DOI: 10.1098/rsif.2016.0539] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/16/2016] [Indexed: 02/06/2023] Open
Abstract
It is well known that a variety of inflammatory diseases are accompanied by hypercoagulability, and a number of more-or-less longer-term signalling pathways have been shown to be involved. In recent work, we have suggested a direct and primary role for bacterial lipopolysaccharide (LPS) in this hypercoagulability, but it seems never to have been tested directly. Here, we show that the addition of tiny concentrations (0.2 ng l−1) of bacterial LPS to both whole blood and platelet-poor plasma of normal, healthy donors leads to marked changes in the nature of the fibrin fibres so formed, as observed by ultrastructural and fluorescence microscopy (the latter implying that the fibrin is actually in an amyloid β-sheet-rich form that on stoichiometric grounds must occur autocatalytically). They resemble those seen in a number of inflammatory (and also amyloid) diseases, consistent with an involvement of LPS in their aetiology. These changes are mirrored by changes in their viscoelastic properties as measured by thromboelastography. As the terminal stages of coagulation involve the polymerization of fibrinogen into fibrin fibres, we tested whether LPS would bind to fibrinogen directly. We demonstrated this using isothermal calorimetry. Finally, we show that these changes in fibre structure are mirrored when the experiment is done simply with purified fibrinogen and thrombin (±0.2 ng l−1 LPS). This ratio of concentrations of LPS : fibrinogen in vivo represents a molecular amplification by the LPS of more than 108-fold, a number that is probably unparalleled in biology. The observation of a direct effect of such highly substoichiometric amounts of LPS on both fibrinogen and coagulation can account for the role of very small numbers of dormant bacteria in disease progression in a great many inflammatory conditions, and opens up this process to further mechanistic analysis and possible treatment.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| | - Sthembile Mbotwe
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| | - Janette Bester
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| | - Christopher J Robinson
- Faculty of Life Sciences, The University of Manchester, 131, Princess Street, Manchester M1 7DN, Lancs, UK The Manchester Institute of Biotechnology, The University of Manchester, 131, Princess Street, Manchester M1 7DN, Lancs, UK Centre for Synthetic Biology of Fine and Speciality Chemicals, The University of Manchester, 131, Princess Street, Manchester M1 7DN, Lancs, UK
| | - Douglas B Kell
- School of Chemistry, The University of Manchester, 131, Princess Street, Manchester M1 7DN, Lancs, UK The Manchester Institute of Biotechnology, The University of Manchester, 131, Princess Street, Manchester M1 7DN, Lancs, UK Centre for Synthetic Biology of Fine and Speciality Chemicals, The University of Manchester, 131, Princess Street, Manchester M1 7DN, Lancs, UK
| |
Collapse
|
39
|
Hidalgo-Lanussa O, Ávila-Rodriguez M, Baez-Jurado E, Zamudio J, Echeverria V, Garcia-Segura LM, Barreto GE. Tibolone Reduces Oxidative Damage and Inflammation in Microglia Stimulated with Palmitic Acid through Mechanisms Involving Estrogen Receptor Beta. Mol Neurobiol 2017; 55:5462-5477. [DOI: 10.1007/s12035-017-0777-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 09/15/2017] [Indexed: 12/12/2022]
|
40
|
Álvarez-Arellano L, Pedraza-Escalona M, Blanco-Ayala T, Camacho-Concha N, Cortés-Mendoza J, Pérez-Martínez L, Pedraza-Alva G. Autophagy impairment by caspase-1-dependent inflammation mediates memory loss in response to β-Amyloid peptide accumulation. J Neurosci Res 2017; 96:234-246. [PMID: 28801921 DOI: 10.1002/jnr.24130] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 07/01/2017] [Accepted: 07/10/2017] [Indexed: 12/20/2022]
Abstract
β-Amyloid peptide accumulation in the cortex and in the hippocampus results in neurodegeneration and memory loss. Recently, it became evident that the inflammatory response triggered by β-Amyloid peptides promotes neuronal cell death and degeneration. In addition to inflammation, β-Amyloid peptides also induce alterations in neuronal autophagy, eventually leading to neuronal cell death. Thus, here we evaluated whether the inflammatory response induced by the β-Amyloid peptides impairs memory via disrupting the autophagic flux. We show that male mice overexpressing β-Amyloid peptides (5XFAD) but lacking caspase-1, presented reduced β-Amyloid plaques in the cortex and in the hippocampus; restored brain autophagic flux and improved learning and memory capacity. At the molecular level, inhibition of the inflammatory response in the 5XFAD mice restored LC3-II levels and prevented the accumulation of oligomeric p62 and ubiquitylated proteins. Furthermore, caspase-1 deficiency reinstates activation of the AMPK/Raptor pathway while down-regulating AKT/mTOR pathway. Consistent with this, we found an inverse correlation between the increase of autophagolysosomes in the cortex of 5XFAD mice lacking caspase-1 and the presence of mitochondria with altered morphology. Together our results indicate that β-Amyloid peptide-induced caspase-1 activation, disrupts autophagy in the cortex and in the hippocampus resulting in neurodegeneration and memory loss.
Collapse
Affiliation(s)
- Lourdes Álvarez-Arellano
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., México
| | - Martha Pedraza-Escalona
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., México
| | - Tonali Blanco-Ayala
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., México
| | - Nohemí Camacho-Concha
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., México
| | - Javier Cortés-Mendoza
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., México
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., México
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., México
| |
Collapse
|
41
|
Thyroid hormone suppresses ischemia-reperfusion-induced liver NLRP3 inflammasome activation: Role of AMP-activated protein kinase. Immunol Lett 2017; 184:92-97. [DOI: 10.1016/j.imlet.2017.01.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/28/2016] [Accepted: 01/12/2017] [Indexed: 01/21/2023]
|
42
|
Chen RJ, Lee YH, Yeh YL, Wang YJ, Wang BJ. The Roles of Autophagy and the Inflammasome during Environmental Stress-Triggered Skin Inflammation. Int J Mol Sci 2016; 17:E2063. [PMID: 27941683 PMCID: PMC5187863 DOI: 10.3390/ijms17122063] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/29/2016] [Accepted: 12/05/2016] [Indexed: 02/07/2023] Open
Abstract
Inflammatory skin diseases are the most common problem in dermatology. The induction of skin inflammation by environmental stressors such as ultraviolet radiation (UVR), hexavalent chromium (Cr(VI)) and TiO₂/ZnO/Ag nanoparticles (NPs) has been demonstrated previously. Recent studies have indicated that the inflammasome is often wrongly activated by these environmental irritants, thus inducing massive inflammation and resulting in the development of inflammatory diseases. The regulation of the inflammasome with respect to skin inflammation is complex and is still not completely understood. Autophagy, an intracellular degradation system that is associated with the maintenance of cellular homeostasis, plays a key role in inflammasome inactivation. As a housekeeping pathway, cells utilize autophagy to maintain the homeostasis of the organ structure and function when exposed to environmental stressors. However, only a few studies have examined the effect of autophagy and/or the inflammasome on skin pathogenesis. Here we review recent findings regarding the involvement of autophagy and inflammasome activation during skin inflammation. We posit that autophagy induction is a novel mechanism inter-modulating environmental stressor-induced skin inflammation. We also attempt to highlight the role of the inflammasome and the possible underlying mechanisms and pathways reflecting the pathogenesis of skin inflammation induced by UVR, Cr(VI) and TiO₂/ZnO/Ag NPs. A more profound understanding about the crosstalk between autophagy and the inflammasome will contribute to the development of prevention and intervention strategies against human skin disease.
Collapse
Affiliation(s)
- Rong-Jane Chen
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| | - Yu-Hsuan Lee
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| | - Ya-Ling Yeh
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
- Department of Biomedical Informatics, Asia University, Taichung 41354, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan.
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Bour-Jr Wang
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
- Department of Occupational and Environmental Medicine, National Cheng Kung University Hospital, Tainan 70428, Taiwan.
- Department of Cosmetic Science and Institute of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan.
| |
Collapse
|
43
|
Jin HS, Kim TS, Jo EK. Emerging roles of orphan nuclear receptors in regulation of innate immunity. Arch Pharm Res 2016; 39:1491-1502. [PMID: 27699647 DOI: 10.1007/s12272-016-0841-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/23/2016] [Indexed: 01/25/2023]
Abstract
Innate immunity constitutes the first line of defense against pathogenic and dangerous insults. However, it is a double-edged sword, as it functions in both clearance of infection and inflammatory damage. It is therefore important that innate immune responses are tightly controlled to prevent harmful excessive inflammation. Nuclear receptors (NRs) are a family of transcription factors that play critical roles in various physiological responses. Orphan NRs are a subset of NRs for which the ligands and functions are unclear. Accumulating evidence has revealed that orphan NRs play essential roles in innate immune responses to prevent pathogenic inflammatory responses and to enhance antimicrobial host defenses. In this review, we describe current knowledge on the roles and mechanisms of orphan NRs in the regulation of innate immune responses. Discovery of new functions of orphan NRs would facilitate development of novel preventive and therapeutic strategies against human inflammatory diseases.
Collapse
Affiliation(s)
- Hyo Sun Jin
- Department of Microbiology, Department of Medical Science, Chungnam National University School of Medicine, 6 Munhwa-dong, Jungku, Daejeon, 301-747, South Korea
| | - Tae Sung Kim
- Department of Microbiology, Department of Medical Science, Chungnam National University School of Medicine, 6 Munhwa-dong, Jungku, Daejeon, 301-747, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Department of Medical Science, Chungnam National University School of Medicine, 6 Munhwa-dong, Jungku, Daejeon, 301-747, South Korea.
| |
Collapse
|
44
|
Azab A, Nassar A, Azab AN. Anti-Inflammatory Activity of Natural Products. Molecules 2016; 21:molecules21101321. [PMID: 27706084 PMCID: PMC6274146 DOI: 10.3390/molecules21101321] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/27/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022] Open
Abstract
This article presents highlights of the published literature regarding the anti-inflammatory activities of natural products. Many review articles were published in this regard, however, most of them have presented this important issue from a regional, limited perspective. This paper summarizes the vast range of review and research articles that have reported on the anti-inflammatory effects of extracts and/or pure compounds derived from natural products. Moreover, this review pinpoints some interesting traditionally used medicinal plants that were not investigated yet.
Collapse
Affiliation(s)
- Abdullatif Azab
- Institute of Applied Research, The Galilee Society, P.O. Box 437, 20200 Shefa-Amr, Israel.
| | - Ahmad Nassar
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, 84105 Beer-Sheva, Israel.
| | - Abed N Azab
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, 84105 Beer-Sheva, Israel.
- Department of Nursing, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, 84105 Beer-Sheva, Israel.
| |
Collapse
|
45
|
Nurmi K, Kareinen I, Virkanen J, Rajamäki K, Kouri VP, Vaali K, Levonen AL, Fyhrquist N, Matikainen S, Kovanen PT, Eklund KK. Hemin and Cobalt Protoporphyrin Inhibit NLRP3 Inflammasome Activation by Enhancing Autophagy: A Novel Mechanism of Inflammasome Regulation. J Innate Immun 2016; 9:65-82. [PMID: 27655219 DOI: 10.1159/000448894] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 08/04/2016] [Indexed: 01/06/2023] Open
Abstract
Inflammasomes are intracellular protein platforms, which, upon activation, produce the highly proinflammatory cytokines interleukin (IL)-1β and IL-18. Heme, hemin and their degradation products possess significant immunomodulatory functions. Here, we studied whether hemin regulates inflammasome function in macrophages. Both hemin and its derivative, cobalt protoporphyrin (CoPP), significantly reduced IL-1β secretion by cultured human primary macrophages, the human monocytic leukemia cell line and also mouse bone marrow-derived and peritoneal macrophages. Intraperitoneal administration of CoPP to mice prior to urate crystal-induced peritonitis alleviated IL-1β secretion to the peritoneal cavity. In cultured macrophages, hemin and CoPP inhibited NLRP3 inflammasome assembly by reducing the amount of intracellular apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC). The reduction of ASC was associated with enhanced autophagosome formation and autophagic flux. Inhibition of autophagy prevented the CoPP-induced depletion of ASC, implying that the depletion was caused by increased autophagy. Our data indicate that hemin functions as an endogenous negative regulator of the NLRP3 inflammasome. The inhibition is mediated via enhanced autophagy that results in increased degradation of ASC. This regulatory mechanism may provide a novel approach for the treatment of inflammasome-related diseases.
Collapse
|
46
|
Recent Advances of the NLRP3 Inflammasome in Central Nervous System Disorders. J Immunol Res 2016; 2016:9238290. [PMID: 27652274 PMCID: PMC5019917 DOI: 10.1155/2016/9238290] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/02/2016] [Indexed: 12/22/2022] Open
Abstract
Inflammasomes are multiprotein complexes that trigger the activation of caspases-1 and subsequently the maturation of proinflammatory cytokines interleukin-1β and interleukin-18. These cytokines play a critical role in mediating inflammation and innate immunity response. Among various inflammasome complexes, the NLRP3 inflammasome is the best characterized, which has been demonstrated as a crucial role in various diseases. Here, we review recently described mechanisms that are involved in the activation and regulation of NLRP3 inflammasome. In addition, we summarize the recent researches on the role of NLRP3 inflammasome in central nervous system (CNS) diseases, including traumatic brain injury, ischemic stroke and hemorrhagic stroke, brain tumor, neurodegenerative diseases, and other CNS diseases. In conclusion, the NLRP3 inflammasome may be a promising therapeutic target for these CNS diseases.
Collapse
|
47
|
Chang ML, Liang KH, Ku CL, Lo CC, Cheng YT, Hsu CM, Yeh CT, Chiu CT. Resistin reinforces interferon λ-3 to eliminate hepatitis C virus with fine-tuning from RETN single-nucleotide polymorphisms. Sci Rep 2016; 6:30799. [PMID: 27477870 PMCID: PMC4967850 DOI: 10.1038/srep30799] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 07/11/2016] [Indexed: 12/16/2022] Open
Abstract
The effect of resistin (RETN) on the response to anti-HCV therapy remains unclear. A prospective cohort study was performed using 655 consecutive HCV patients, of whom 513 had completed a course of interferon-based therapy. Multivariate and GEE analyses revealed four RETN single-nucleotide polymorphisms (SNPs), rs34861192, rs3219175, rs3745367 and rs1423096, to be synergistically associated with resistin levels. After adjusting for co-factors such as interferon λ-3 (IFNL3)-rs12979860, the resistin level and the hyper-resistinemic genotype at the 4 RETN SNPs were positively and negatively associated with a sustained virological response (SVR), respectively. RETN-rs3745367 was in linkage disequilibrium with IFNL3-rs12979860. Compared to non-SVR patients, SVR patients had higher levels of pre-therapy resistin, primarily originating from intrahepatic lymphocytes, stellate cells, Kupffer cells, hepatic progenitor cells and hepatocytes. This difference diminished over the course of therapy, as only SVR patients exhibited a 24-week post-therapy decrease in resistin. Both resistin and IFNL3 mRNAs were upregulated, but only resistin mRNA was upregulated by recombinant resistin in peripheral blood mononuclear cells with and without hyper-resistinemic genotypes of the 4 RETN SNPs, respectively. Fine-tuned by RETN SNPs, intrahepatic, multi-cellular resistin reinforced IFNL3 in eliminating HCV via immunomodulation to counteract pro-inflammation. These results encourage the development of novel resistin-targeted anti-viral agents.
Collapse
Affiliation(s)
- Ming-Ling Chang
- Liver Research Center, Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kung-Hao Liang
- Liver Research Center, Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Cheng-Lung Ku
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Chi Lo
- Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Ting Cheng
- Liver Research Center, Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chen-Ming Hsu
- Liver Research Center, Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chau-Ting Yeh
- Liver Research Center, Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Cheng-Tang Chiu
- Liver Research Center, Division of Hepatology, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
48
|
Katsnelson MA, Lozada-Soto KM, Russo HM, Miller BA, Dubyak GR. NLRP3 inflammasome signaling is activated by low-level lysosome disruption but inhibited by extensive lysosome disruption: roles for K+ efflux and Ca2+ influx. Am J Physiol Cell Physiol 2016; 311:C83-C100. [PMID: 27170638 DOI: 10.1152/ajpcell.00298.2015] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 05/05/2016] [Indexed: 12/28/2022]
Abstract
Nucleotide-binding domain, leucine-rich-repeat-containing family, pyrin domain-containing 3 (NLRP3) is a cytosolic protein that nucleates assembly of inflammasome signaling platforms, which facilitate caspase-1-mediated IL-1β release and other inflammatory responses in myeloid leukocytes. NLRP3 inflammasomes are assembled in response to multiple pathogen- or environmental stress-induced changes in basic cell physiology, including the destabilization of lysosome integrity and activation of K(+)-permeable channels/transporters in the plasma membrane (PM). However, the quantitative relationships between lysosome membrane permeabilization (LMP), induction of increased PM K(+) permeability, and activation of NLRP3 signaling are incompletely characterized. We used Leu-Leu-O-methyl ester (LLME), a soluble lysosomotropic agent, to quantitatively track the kinetics and extent of LMP in relation to NLRP3 inflammasome signaling responses (ASC oligomerization, caspase-1 activation, IL-1β release) and PM cation fluxes in murine bone marrow-derived dendritic cells (BMDCs). Treatment of BMDCs with submillimolar (≤1 mM) LLME induced slower and partial increases in LMP that correlated with robust NLRP3 inflammasome activation and K(+) efflux. In contrast, supramillimolar (≥2 mM) LLME elicited extremely rapid and complete collapse of lysosome integrity that was correlated with suppression of inflammasome signaling. Supramillimolar LLME also induced dominant negative effects on inflammasome activation by the canonical NLRP3 agonist nigericin; this inhibition correlated with an increase in NLRP3 ubiquitination. LMP elicited rapid BMDC death by both inflammasome-dependent pyroptosis and inflammasome-independent necrosis. LMP also triggered Ca(2+) influx, which attenuated LLME-stimulated NLRP3 inflammasome signaling but potentiated LLME-induced necrosis. Taken together, these studies reveal a previously unappreciated signaling network that defines the coupling between LMP, changes in PM cation fluxes, cell death, and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Michael A Katsnelson
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Kristen M Lozada-Soto
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Hana M Russo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Barbara A Miller
- Department of Pediatrics, Penn State Hershey Children's Hospital, Hershey, Pennsylvania
| | - George R Dubyak
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio;
| |
Collapse
|
49
|
Brownstein MJ. NLRC4 inflammasomes: ‘their appearance and their work was a wheel in the middle of a wheel’. Oral Dis 2016; 22:85-6. [DOI: 10.1111/odi.12406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
50
|
|