1
|
Akter R, Noor F, Tonmoy HS, Ahmed A. Potential of SIRT6 modulators in targeting molecular pathways involved in cardiovascular diseases and their treatment-A comprehensive review. Biochem Pharmacol 2025; 233:116787. [PMID: 39894306 DOI: 10.1016/j.bcp.2025.116787] [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: 09/20/2024] [Revised: 01/09/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality and morbidity, accounting for major public health concerns worldwide. CVD poses an immense burden on the global healthcare system and economy. Ischemic heart disease, stroke, heart failure, atherosclerosis, and hypertension are the major diseases belonging to CVDs and ischemic heart diseases and stroke contribute to most CVD-induced deaths. Previously published review articles focused on the role of SIRT6 in CVDs but did not focus on the important role of SIRT6 in modulating the signaling pathways involved in CVDs and targeting them to treat CVDs. Thus, this review aims to identify and delineate the major signaling pathways that are involved in CVDs and whether SIRT6 can modulate those pathways to improve and treat CVDs. Alongside possible applications of small molecule modulators of SIRT6 in cardiovascular disease treatment have been comprehensively analyzed.
Collapse
Affiliation(s)
- Raushanara Akter
- School of Pharmacy, KHA 224 Bir Uttam Rafiqul Islam Avenue, Merul Badda, BRAC University, Dhaka 1212, Bangladesh.
| | - Fouzia Noor
- School of Pharmacy, KHA 224 Bir Uttam Rafiqul Islam Avenue, Merul Badda, BRAC University, Dhaka 1212, Bangladesh
| | - Hasan Shahriyer Tonmoy
- School of Pharmacy, KHA 224 Bir Uttam Rafiqul Islam Avenue, Merul Badda, BRAC University, Dhaka 1212, Bangladesh
| | - Ashfaq Ahmed
- School of Pharmacy, KHA 224 Bir Uttam Rafiqul Islam Avenue, Merul Badda, BRAC University, Dhaka 1212, Bangladesh
| |
Collapse
|
2
|
Ren F, Li Y, Luo H, Gao S, Jiang S, Yang J, Rao C, Chen Y, Peng C. Extraction, detection, bioactivity, and product development of luteolin: A review. Heliyon 2024; 10:e41068. [PMID: 39759280 PMCID: PMC11700251 DOI: 10.1016/j.heliyon.2024.e41068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Luteolin is a kind of natural flavonoid, widely existing in a variety of plants, has been revealed to have a wide range of biological activities. In recent years, the research results of luteolin are abundant. Here we review the latest research results of luteolin in order to provide new ideas for further research and development of luteolin. In this paper, the focus of the search was published between 2010 and 2024 on the extraction and determination of luteolin, biological activities, and the development and application of luteolin products. A comprehensive search using the keyword "luteolin" was conducted in the PubMed, Web of Science and WIPO databases. Through the collection of related literature, this paper summarized a variety of extraction techniques of luteolin, including immersion extraction, solvent extraction, ultrasonic-assisted extraction, supercritical fluid extraction and so on. The determination methods include: thin layer chromatography (TLC), high performance liquid chromatography (HPLC), capillary electrophoresis (CE), electrochemical method (ED) and so on. In addition, the biological activities of luteolin, including antioxidant, anti-inflammatory, anti-tumor, antibacterial, analgesic and so on, were described. And luteolin as the main component of the product is being gradually developed, and has been used in the field of food, medicine and cosmetics. This paper provides a reference for further study of luteolin.
Collapse
Affiliation(s)
- Fajian Ren
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ying Li
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Hanyuan Luo
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Song Gao
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Shanshan Jiang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Jian Yang
- Chuan-chu UNITED INTERNATIONAL Engineering Co., LTD, Chengdu, China
| | - Chaolong Rao
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yan Chen
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Cheng Peng
- State Key Laboratory of Traditional Chinese Medicine Resources in Southwest China, Chengdu, 611137, China
| |
Collapse
|
3
|
Zheng H, Li T, Hu Z, Zheng Q, Wang J. The potential of flavonoids to mitigate cellular senescence in cardiovascular disease. Biogerontology 2024; 25:985-1010. [PMID: 39325277 DOI: 10.1007/s10522-024-10141-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
Aging is one of the most significant factors affecting cardiovascular health, with cellular senescence being a central hallmark. Senescent cells (SCs) secrete a specific set of signaling molecules known as the senescence-associated secretory phenotype (SASP). The SASP has a remarkable impact on age-associated diseases, particularly cardiovascular diseases (CVD). Targeting SCs through anti-aging therapies represents a novel strategy to effectively retard senescence and attenuate disease progression. Accumulating evidence demonstrates that the flavonoids, widely presented in fruits and vegetables worldwide, can delay or treat CVD via selectively eliminating SCs (senolytics) and modulating SASPs (senomorphics). Nevertheless, only sporadic research has illustrated the application of flavonoids in targeting SCs for CVD, which requires further exploration. This review recapitulates the hallmarks and key molecular mechanisms involved in cellular senescence, then summarizes senescence of different types of cardiac cells and describes the mechanisms by which cellular senescence affects CVD development. The discussion culminates with the potential use of flavonoids via exerting their biological effects on cellular senescence to reduce CVD incidence. This summary will provide valuable insights for cardiovascular drug design, development and clinical applications leveraging flavonoids.
Collapse
Affiliation(s)
- Huimin Zheng
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, Jiangsu, People's Republic of China
| | - Tiantian Li
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, Jiangsu, People's Republic of China
| | - Ziyun Hu
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, Jiangsu, People's Republic of China
| | - Qi Zheng
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, Jiangsu, People's Republic of China
| | - Junsong Wang
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, 210094, Jiangsu, People's Republic of China.
| |
Collapse
|
4
|
Zumerle S, Sarill M, Saponaro M, Colucci M, Contu L, Lazzarini E, Sartori R, Pezzini C, Rinaldi A, Scanu A, Sgrignani J, Locatelli P, Sabbadin M, Valdata A, Brina D, Giacomini I, Rizzo B, Pierantoni A, Sharifi S, Bressan S, Altomare C, Goshovska Y, Giraudo C, Luisetto R, Iaccarino L, Torcasio C, Mosole S, Pasquini E, Rinaldi A, Pellegrini L, Peron G, Fassan M, Masiero S, Giori AM, Dall'Acqua S, Auwerx J, Cippà P, Cavalli A, Bolis M, Sandri M, Barile L, Montopoli M, Alimonti A. Targeting senescence induced by age or chemotherapy with a polyphenol-rich natural extract improves longevity and healthspan in mice. NATURE AGING 2024; 4:1231-1248. [PMID: 38951692 PMCID: PMC11408255 DOI: 10.1038/s43587-024-00663-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 05/31/2024] [Indexed: 07/03/2024]
Abstract
Accumulating senescent cells within tissues contribute to the progression of aging and age-related diseases. Botanical extracts, rich in phytoconstituents, present a useful resource for discovering therapies that could target senescence and thus improve healthspan. Here, we show that daily oral administration of a standardized extract of Salvia haenkei (Haenkenium (HK)) extended lifespan and healthspan of naturally aged mice. HK treatment inhibited age-induced inflammation, fibrosis and senescence markers across several tissues, as well as increased muscle strength and fur thickness compared with age-matched controls. We also found that HK treatment reduced acutely induced senescence by the chemotherapeutic agent doxorubicin, using p16LUC reporter mice. We profiled the constituent components of HK by mass spectrometry, and identified luteolin-the most concentrated flavonoid in HK-as a senomorphic compound. Mechanistically, by performing surface plasmon resonance and in situ proximity ligation assay, we found that luteolin disrupted the p16-CDK6 interaction. This work demonstrates that administration of HK promotes longevity in mice, possibly by modulating cellular senescence and by disrupting the p16-CDK6 interaction.
Collapse
Affiliation(s)
- Sara Zumerle
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Miles Sarill
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Miriam Saponaro
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
- Department of Urology and Pediatric Urology, University Medical Center Bonn (UKB), Bonn, Germany
| | - Manuel Colucci
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera italiana, Lugano, Switzerland
- Faculty of Biology and Medicine, University of Lausanne UNIL, Lausanne, Switzerland
| | - Liliana Contu
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
| | - Edoardo Lazzarini
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Euler Institute, Università della Svizzera Italiana, Lugano, Switzerland
| | - Roberta Sartori
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Camilla Pezzini
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Anna Rinaldi
- Department of Medicine, Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Anna Scanu
- Department of Neuroscience, Rehabilitation Unit, University of Padova, Padova, Italy
| | - Jacopo Sgrignani
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Patrizia Locatelli
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Marianna Sabbadin
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Aurora Valdata
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera italiana, Lugano, Switzerland
- Department of Health Sciences and Technology (D-HEST), ETH Zurich, Zurich, Switzerland
| | - Daniela Brina
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera italiana, Lugano, Switzerland
| | - Isabella Giacomini
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Beatrice Rizzo
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Alessandra Pierantoni
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- Institute for Research on Cancer and Aging, Nice, France
| | - Saman Sharifi
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Medicine, University of Padova, Padova, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Silvia Bressan
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera italiana, Lugano, Switzerland
| | - Claudia Altomare
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Euler Institute, Università della Svizzera Italiana, Lugano, Switzerland
| | - Yulia Goshovska
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Euler Institute, Università della Svizzera Italiana, Lugano, Switzerland
| | - Chiara Giraudo
- Department of Medicine, University of Padova, Padova, Italy
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health - DCTV, University of Padova, Padova, Italy
| | - Roberto Luisetto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Luca Iaccarino
- Department of Medicine, University of Padova, Padova, Italy
| | - Cristina Torcasio
- Department of Medicine, Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Simone Mosole
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera italiana, Lugano, Switzerland
| | - Emiliano Pasquini
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera italiana, Lugano, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera italiana, Lugano, Switzerland
| | - Laura Pellegrini
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera italiana, Lugano, Switzerland
| | - Gregorio Peron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Matteo Fassan
- Department of Medicine, University of Padova, Padova, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Stefano Masiero
- Department of Neuroscience, Rehabilitation Unit, University of Padova, Padova, Italy
| | | | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Pietro Cippà
- Università della Svizzera italiana, Lugano, Switzerland
- Department of Medicine, Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Andrea Cavalli
- Università della Svizzera italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Marco Bolis
- Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Università della Svizzera italiana, Lugano, Switzerland
| | - Marco Sandri
- Veneto Institute of Molecular Medicine, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Euler Institute, Università della Svizzera Italiana, Lugano, Switzerland
| | - Monica Montopoli
- Veneto Institute of Molecular Medicine, Padova, Italy.
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.
| | - Andrea Alimonti
- Veneto Institute of Molecular Medicine, Padova, Italy.
- Department of Medicine, University of Padova, Padova, Italy.
- Institute of Oncology Research (IOR), Bellinzona, Switzerland.
- Università della Svizzera italiana, Lugano, Switzerland.
- Department of Health Sciences and Technology (D-HEST), ETH Zurich, Zurich, Switzerland.
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
| |
Collapse
|
5
|
Tu H, Gao Q, Zhou Y, Peng L, Wu D, Zhang D, Yang J. The role of sirtuins in intervertebral disc degeneration: Mechanisms and therapeutic potential. J Cell Physiol 2024; 239:e31328. [PMID: 38922861 DOI: 10.1002/jcp.31328] [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: 12/17/2023] [Revised: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 06/28/2024]
Abstract
Intervertebral disc degeneration (IDD) is one of the main causes of low back pain, which affects the patients' quality of life and health and imposes a significant socioeconomic burden. Despite great efforts made by researchers to understand the pathogenesis of IDD, effective strategies for preventing and treating this disease remain very limited. Sirtuins are a highly conserved family of (NAD+)-dependent deacetylases in mammals that are involved in a variety of metabolic processes in vivo. In recent years, sirtuins have attracted much attention owing to their regulatory roles in IDD on physiological activities such as inflammation, apoptosis, autophagy, aging, oxidative stress, and mitochondrial function. At the same time, many studies have explored the therapeutic effects of sirtuins-targeting activators or micro-RNA in IDD. This review summarizes the molecular pathways of sirtuins involved in IDD, and summarizes the therapeutic role of activators or micro-RNA targeting Sirtuins in IDD, as well as the current limitations and challenges, with a view to provide possible solutions for the treatment of IDD.
Collapse
Affiliation(s)
- Heng Tu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Qian Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yumeng Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Li Peng
- Key Laboratory of Bio-Resource & Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Dan Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Demao Zhang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Wu Z, Shi R, Yan S, Zhang S, Lu B, Huang Z, Ji L. Integrating network pharmacology, experimental validation and molecular docking to reveal the alleviation of Yinhuang granule on idiopathic pulmonary fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155368. [PMID: 38498951 DOI: 10.1016/j.phymed.2024.155368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/21/2023] [Accepted: 01/15/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic disease characterized by the abnormal proliferation of fibroblast and excessive deposition of extracellular matrix (ECM), accompanied by inflammation and ultimately respiratory failure. Yinhuang granule (YHG), with clinical properties of clearing heat, detoxifying and anti-inflammation, is commonly used to heal upper respiratory diseases in China for decades. PURPOSE To explore the improvement of YHG on bleomycin (BLM)-induced IPF in mice and its possible engaged mechanism. METHODS The mortality rate was recorded, lung function was determined and hematoxylin-eosin (H&E) staining was carried out to explore the alleviation of YHG on BLM-caused IPF in mice. Hydroxyproline, collagen I and collagen III contents were detected, and Sirius red and Masson staining were conducted to evaluate YHG's alleviation on lung fibrosis. The underlying mechanism was predicted by network pharmacology, and confirmed by Real-time polymerase chain reaction (RT-PCR), Western-blot (WB) and enzyme linked immunosorbent assay (ELISA). The binding affinity between related key proteins and active compounds in YHG was calculated by using molecular docking, and further validated by cellular thermal shift assay (CESTA). RESULTS YHG (400, 800 mg/kg) weakened lung damage and pulmonary fibrosis in mice induced by BLM. Network pharmacology and experimental validation displayed that inflammation and angiogenesis participated in the YHG-provided improvement on IPF, and key involved molecules included tumor necrosis factor-α (TNFα), vascular endothelial growth factor-A (VEGFA), interleukine-6 (IL-6), etc. The data of molecular docking presented that some main active compounds from YHG had a high binding affinity with TNFR1 or VEGFR2, and some of them were further validated by CESTA. CONCLUSION YHG effectively improved the BLM-induced IPF in mice via reducing inflammation and angiogenesis.
Collapse
Affiliation(s)
- Zeqi Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ruijia Shi
- School of Basic Medical Science of Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shihao Yan
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai,200123, China
| | - Shaobo Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhenlin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
7
|
Nadalin P, Kim JK, Park SU. Recent insights into luteolin and its biological and pharmacological activities. EXCLI JOURNAL 2024; 23:787-794. [PMID: 39165588 PMCID: PMC11333740 DOI: 10.17179/excli2024-7168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/23/2024] [Indexed: 08/22/2024]
Affiliation(s)
- Priscilla Nadalin
- Department of Crop Science, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
| | - Jae Kwang Kim
- Division of Life Sciences and Convergence Research Center for Insect Vectors, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea
| | - Sang Un Park
- Department of Crop Science, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Korea
- Department of Smart Agricultural Systems, Graduate School, Chungnam National University, Daejeon 34134, Republic of Korea
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Daejeon 34134, Republic of Korea
| |
Collapse
|
8
|
Tao Y, Yu X, Li X, Xu Y, Wang H, Zhang L, Lin R, Wang Y, Fan P. M6A methylation-regulated autophagy may be a new therapeutic target for intervertebral disc degeneration. Cell Biol Int 2024; 48:389-403. [PMID: 38317355 DOI: 10.1002/cbin.12135] [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: 09/07/2023] [Accepted: 01/01/2024] [Indexed: 02/07/2024]
Abstract
Degeneration of intervertebral discs is considered one of the most important causes of low back pain and disability. The intervertebral disc (IVD) is characterized by its susceptibility to various stressors that accelerate the senescence and apoptosis of nucleus pulposus cells, resulting in the loss of these cells and dysfunction of the intervertebral disc. Therefore, how to reduce the loss of nucleus pulposus cells under stress environment is the main problem in treating intervertebral disc degeneration. Autophagy is a kind of programmed cell death, which can provide energy by recycling substances in cells. It is considered to be an effective method to reduce the senescence and apoptosis of nucleus pulposus cells under stress. However, further research is needed on the mechanisms by which autophagy of nucleus pulposus cells is regulated under stress environments. M6A methylation, as the most extensive RNA modification in eukaryotic cells, participates in various cellular biological functions and is believed to be related to the regulation of autophagy under stress environments, may play a significant role in nucleus pulposus responding to stress. This article first summarizes the effects of various stressors on the death and autophagy of nucleus pulposus cells. Then, it summarizes the regulatory mechanism of m6A methylation on autophagy-related genes under stress and the role of these autophagy genes in nucleus pulposus cells. Finally, it proposes that the methylation modification of autophagy-related genes regulated by m6A may become a new treatment approach for intervertebral disc degeneration, providing new insights and ideas for the clinical treatment of intervertebral disc degeneration.
Collapse
Affiliation(s)
- Yuao Tao
- Department of Spine Center, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Xiaoyu Yu
- Department of Gynaecology and Obstetrics, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaolong Li
- Department of Spine Center, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yuzhu Xu
- Department of Spine Center, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Hui Wang
- Department of Spine Center, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Lele Zhang
- Department of Spine Center, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Rubing Lin
- Department of Orthopedics, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Yuntao Wang
- Department of Spine Center, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Pan Fan
- Department of Spine Center, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| |
Collapse
|
9
|
Shen J, Lan Y, Ji Z, Liu H. Sirtuins in intervertebral disc degeneration: current understanding. Mol Med 2024; 30:44. [PMID: 38553713 PMCID: PMC10981339 DOI: 10.1186/s10020-024-00811-0] [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: 01/09/2024] [Accepted: 03/20/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is one of the etiologic factors of degenerative spinal diseases, which can lead to a variety of pathological spinal conditions such as disc herniation, spinal stenosis, and scoliosis. IVDD is a leading cause of lower back pain, the prevalence of which increases with age. Recently, Sirtuins/SIRTs and their related activators have received attention for their activity in the treatment of IVDD. In this paper, a comprehensive systematic review of the literature on the role of SIRTs and their activators on IVDD in recent years is presented. The molecular pathways involved in the regulation of IVDD by SIRTs are summarized, and the effects of SIRTs on senescence, inflammatory responses, oxidative stress, and mitochondrial dysfunction in myeloid cells are discussed with a view to suggesting possible solutions for the current treatment of IVDD. PURPOSE This paper focuses on the molecular mechanisms by which SIRTs and their activators act on IVDD. METHODS A literature search was conducted in Pubmed and Web of Science databases over a 13-year period from 2011 to 2024 for the terms "SIRT", "Sirtuin", "IVDD", "IDD", "IVD", "NP", "Intervertebral disc degeneration", "Intervertebral disc" and "Nucleus pulposus". RESULTS According to the results, SIRTs and a large number of activators showed positive effects against IVDD.SIRTs modulate autophagy, myeloid apoptosis, oxidative stress and extracellular matrix degradation. In addition, they attenuate inflammatory factor-induced disc damage and maintain homeostasis during disc degeneration. Several clinical studies have reported the protective effects of some SIRTs activators (e.g., resveratrol, melatonin, honokiol, and 1,4-dihydropyridine) against IVDD. CONCLUSION The fact that SIRTs and their activators play a hundred different roles in IVDD helps to better understand their potential to develop further treatments for IVDD. NOVELTY This review summarizes current information on the mechanisms of action of SIRTs in IVDD and the challenges and limitations of translating their basic research into therapy.
Collapse
Affiliation(s)
- Jianlin Shen
- Department of Orthopaedics, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China
- Central Laboratory, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China
| | - Yujian Lan
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ziyu Ji
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Huan Liu
- Department of Orthopaedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- The Third People's Hospital of Longmatan District, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
10
|
Lombardo GE, Russo C, Maugeri A, Navarra M. Sirtuins as Players in the Signal Transduction of Citrus Flavonoids. Int J Mol Sci 2024; 25:1956. [PMID: 38396635 PMCID: PMC10889095 DOI: 10.3390/ijms25041956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Sirtuins (SIRTs) belong to the family of nicotine adenine dinucleotide (NAD+)-dependent class III histone deacetylases, which come into play in the regulation of epigenetic processes through the deacetylation of histones and other substrates. The human genome encodes for seven homologs (SIRT1-7), which are localized into the nucleus, cytoplasm, and mitochondria, with different enzymatic activities and regulatory mechanisms. Indeed, SIRTs are involved in different physio-pathological processes responsible for the onset of several human illnesses, such as cardiovascular and neurodegenerative diseases, obesity and diabetes, age-related disorders, and cancer. Nowadays, it is well-known that Citrus fruits, typical of the Mediterranean diet, are an important source of bioactive compounds, such as polyphenols. Among these, flavonoids are recognized as potential agents endowed with a wide range of beneficial properties, including antioxidant, anti-inflammatory, hypolipidemic, and antitumoral ones. On these bases, we offer a comprehensive overview on biological effects exerted by Citrus flavonoids via targeting SIRTs, which acted as modulator of several signaling pathways. According to the reported studies, Citrus flavonoids appear to be promising SIRT modulators in many different pathologies, a role which might be potentially evaluated in future therapies, along with encouraging the study of those SIRT members which still lack proper evidence on their support.
Collapse
Affiliation(s)
- Giovanni Enrico Lombardo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (G.E.L.); (C.R.); (M.N.)
| | - Caterina Russo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (G.E.L.); (C.R.); (M.N.)
| | - Alessandro Maugeri
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (G.E.L.); (C.R.); (M.N.)
| |
Collapse
|
11
|
Fu W, Xu L, Chen Y, Zhang Z, Chen S, Li Q, You X. Luteolin induces ferroptosis in prostate cancer cells by promoting TFEB nuclear translocation and increasing ferritinophagy. Prostate 2024; 84:223-236. [PMID: 37904332 DOI: 10.1002/pros.24642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 08/22/2023] [Accepted: 10/17/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND As the second most common cancer in men and the leading cause of cancer-related death, prostate cancer (PCa) could potentially be treated by inducing ferroptosis. In this study, we aimed to investigate whether luteolin could induce ferroptosis in PCa cells through the transcription Factor EB (TFEB). METHODS Different concentrations of luteolin were applied to treat normal prostate epithelial cells RWPE-1 and PCa cell lines DU145, PC-3, VCaP, and LNcaP. Ferrostatin-1 (Fer-1), Necrostain-1 (Nec-1), 3-methyladenine (3-MA), chloroquine (CQ), and the apoptosis inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone (Z-VAD-FMK) were added to treat DU145 and PC-3 cells. Additionally, we knocked down TFEB and performed in vitro cell experiments. Finally, tumor-forming experiments in nude mice were conducted to verify luteolin mechanism in PCa after knocking down TFEB. RESULTS There was no significant difference in RWPE-1 at 12, 24, and 48 h after treatment with 60 μM luteolin. However, a significant difference was observed between DU145 and PC-3 cells. Luteolin exhibited a promoting effect on PCa cell death. After treatment with luteolin, cell viability, and Ki67 expression were decreased, and AnV-PI-positive dead cells were increased. Fer-1, Nec-1, 3-MA, and Z-VAD-FMK reversed luteolin effects on DU145 and PC-3 cell viability, proliferation, and AnV-PI-positive dead cells. Among them, Fer-1 and 3-MA were more effective. Luteolin-induced increased autophagy and ferroptosis in DU145 and PC-3 cells. Moreover, luteolin promoted ferroptosis by inducing increased autophagy in DU145 and PC-3 cells. However, knockdown of TFEB reversed the ability of luteolin to induce lysosome degradation of ferritin. In addition, luteolin promoted PCa ferroptosis by inducing ferritinophagy in vivo. CONCLUSIONS Luteolin-induced ferroptosis in PCa cells by promoting TFEB nuclear translocation and increasing ferritinophagy.
Collapse
Affiliation(s)
- Wei Fu
- Department of Andrology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lei Xu
- Department of Andrology, The First Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, China
| | - Yingwen Chen
- Department of Andrology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zezheng Zhang
- Department of Andrology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Shuchao Chen
- Department of Andrology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Qixin Li
- Department of Andrology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xujun You
- Department of Andrology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
12
|
Kang L, Zhang H, Jia C, Zhang R, Shen C. Epigenetic modifications of inflammation in intervertebral disc degeneration. Ageing Res Rev 2023; 87:101902. [PMID: 36871778 DOI: 10.1016/j.arr.2023.101902] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/17/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Intervertebral disc degeneration (IDD) is a common cause of joint-related chronic disability in elderly individuals worldwide. It seriously impacts the quality of life and inflicts a substantial social and economic burden. The pathological mechanisms underlying IDD have not been fully revealed, leading to less satisfactory clinical treatment outcomes. More studies are urgently needed to reveal its precise pathological mechanisms. Numerous studies have revealed that inflammation is closely related to various pathological processes of IDD, including the continuous loss of extracellular matrix, cell apoptosis, and senescence, indicating the important role of inflammation in the pathological mechanism of IDD. Epigenetic modifications affect the functions and characteristics of genes mainly through DNA methylation, histone modification, non-coding RNA regulation, and other mechanisms, thus having a major effect on the survival state of the body. Recently, the role of epigenetic modifications in inflammation during IDD has been attracting research interest. In this review, we summarize the roles of different types of epigenetic modifications in inflammation during IDD in recent years, to improve our understanding of the etiology of IDD and to transform basic research strategy into a clinically effective treatment for joint-related chronic disability in elderly individuals.
Collapse
Affiliation(s)
- Liang Kang
- Department of Orthopedics & Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Huaqing Zhang
- Department of Orthopedics & Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Chongyu Jia
- Department of Orthopedics & Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Renjie Zhang
- Department of Orthopedics & Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Cailiang Shen
- Department of Orthopedics & Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| |
Collapse
|
13
|
Qian Y, Yin J, Ni J, Chen X, Shen Y. A Network Pharmacology Method Combined with Molecular Docking Verification to Explore the Therapeutic Mechanisms Underlying Simiao Pill Herbal Medicine against Hyperuricemia. BIOMED RESEARCH INTERNATIONAL 2023; 2023:2507683. [PMID: 36817858 PMCID: PMC9935928 DOI: 10.1155/2023/2507683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 02/11/2023]
Abstract
Objective Hyperuricemia (HUA) is a common metabolic disease caused by disordered purine metabolism. We aim to reveal the mechanisms underlying the anti-HUA function of Simiao pill and provide therapeutic targets. Methods Simiao pill-related targets were obtained using Herbal Ingredients' Targets (HIT), Traditional Chinese Medicine Systems Pharmacology (TCMSP), and Traditional Chinese Medicine Integrated Database (TCMID). HUA-associated targets were retrieved from GeneCards, DisGeNET, and Therapeutic Targets Database (TTD). Protein-protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database, ggraph and igraph R packages. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using ClusterProfiler. The top 10 core targets were identified through cytoHubba. Molecular docking was conducted using PyMOL and AutoDock high-performance liquid chromatograph (HPLC) analysis was performed to identify effective compounds of Simiao pill. Results Simiao pill-HUA target network contained 80 targets. The key targets were mainly involved in inflammatory responses. Insulin (INS), tumor necrosis factor (TNF), interleukin-6 (IL6), interleukin 1 beta (IL1B), vascular endothelial growth factor A (VEGFA), leptin (LEP), signal transducer and activator of transcription 3 (STAT3), C-C motif chemokine ligand 2 (CCL2), interleukin-10 (IL10), and toll like receptor 4 (TLR4) were the top 10 targets in the PPI network. GO analysis demonstrated the main implication of the targets in molecular responses, production, and metabolism. KEGG analysis revealed that Simiao pill might mitigate HUA through advanced glycation end-product- (AGE-) receptor for AGE- (RAGE-) and hypoxia-inducible factor-1- (HIF-1-) associated pathways. IL1B, IL6, IL10, TLR4, and TNF were finally determined as the promising targets of Simiao pill treating HUA. Through molecular docking and HPLC analysis, luteolin, quercetin, rutaecarpine, baicalin, and atractylenolide I were the main active compounds. Conclusions Simiao pill can mitigate HUA by restraining inflammation, mediating AGE-RAGE- and HIF-1-related pathways, and targeting IL1B, IL6, IL10, TLR4, and TNF.
Collapse
Affiliation(s)
- Yue Qian
- Rehabilitation Center, Hangzhou Wuyunshan Hospital (Hangzhou Institute of Health Promotion), Hangzhou 310000, China
| | - Jiazhen Yin
- Department of Nephrology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou 310000, China
| | - Juemin Ni
- Rehabilitation Center, Hangzhou Wuyunshan Hospital (Hangzhou Institute of Health Promotion), Hangzhou 310000, China
| | - Xiaona Chen
- Rehabilitation Center, Hangzhou Wuyunshan Hospital (Hangzhou Institute of Health Promotion), Hangzhou 310000, China
| | - Yan Shen
- Department of Nursing, Hangzhou Wuyunshan Hospital (Hangzhou Institute of Health Promotion), Hangzhou 310000, China
| |
Collapse
|
14
|
In Vitro Effect of Flavonoids on Basophils Degranulation and Intestinal Epithelial Barrier Damage Induced by ω-5 Gliadin-Derived Peptide. Foods 2022; 11:foods11233857. [PMID: 36496664 PMCID: PMC9741160 DOI: 10.3390/foods11233857] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/23/2022] [Accepted: 11/26/2022] [Indexed: 12/02/2022] Open
Abstract
Flavonoids have antioxidant, anti-inflammatory and immunomodulatory properties, and may alleviate food allergic reactions and intestinal inflammation induced by ω-5 gliadin, a main allergen of wheat food allergy in children. In this study, a human basophil KU812 cell degranulation model and a Caco-2 monolayer cell model were constructed in vitro to evaluate the effects of four flavonoids on the allergenicity of ω-5 gliadin peptides and ω-5 gliadin peptide-induced barrier damage in Caco-2 intestinal epithelial monolayers. The results show that baicalein, luteolin, isorhamnetin and naringenin can significantly inhibit the degranulation of KU812 cells stimulated by ω-5 gliadin-derived peptide P4 and the release of IL-6 and TNF-α. In addition, the four flavonoids significantly inhibited the ω-5 gliadin-derived peptide P4 to induce the release of IL-6, IL-8 in Caco-2 cells, inhibited the release of zonulin, and significantly increase the expression of tight junction proteins Occludin and ZO-1 in the Caco-2 cell monolayer. In conclusion, baicalein, luteolin, isorhamnetin and naringenin inhibit degranulation stimulated by wheat allergen and enhance intestinal barrier functions, which supports the potential pharmaceutical application of the four flavonoids treatment for wheat food allergy.
Collapse
|
15
|
Muruganathan N, Dhanapal AR, Baskar V, Muthuramalingam P, Selvaraj D, Aara H, Shiek Abdullah MZ, Sivanesan I. Recent Updates on Source, Biosynthesis, and Therapeutic Potential of Natural Flavonoid Luteolin: A Review. Metabolites 2022; 12:1145. [PMID: 36422285 PMCID: PMC9696498 DOI: 10.3390/metabo12111145] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 08/27/2023] Open
Abstract
Nature gives immense resources that are beneficial to humankind. The natural compounds present in plants provide primary nutritional values to our diet. Apart from food, plants also provide chemical compounds with therapeutic values. The importance of these plant secondary metabolites is increasing due to more studies revealing their beneficial properties in treating and managing various diseases and their symptoms. Among them, flavonoids are crucial secondary metabolite compounds present in most plants. Of the reported 8000 flavonoid compounds, luteolin is an essential dietary compound. This review discusses the source of the essential flavonoid luteolin in various plants and its biosynthesis. Furthermore, the potential health benefits of luteolins such as anti-cancer, anti-microbial, anti-inflammatory, antioxidant, and anti-diabetic effects and their mechanisms are discussed in detail. The activity of luteolin and its derivatives are diverse, as they help to prevent and control many diseases and their life-threatening effects. This review will enhance the knowledge and recent findings regarding luteolin and its therapeutic effects, which are certainly useful in potentially utilizing this natural metabolite.
Collapse
Affiliation(s)
- Nandakumar Muruganathan
- Department of Plant Pathology and Microbiology, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Anand Raj Dhanapal
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
- Centre for Plant Tissue Culture & Central Instrumentation Laboratory, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Venkidasamy Baskar
- Department of Oral & Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Pandiyan Muthuramalingam
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52725, Republic of Korea
| | - Dhivya Selvaraj
- Department of Computer Science and Engineering CSE-AI, Amrita School of Engineering, Chennai 601103, Tamil Nadu, India
| | - Husne Aara
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | | | - Iyyakkannu Sivanesan
- Department of Bioresources and Food Science, Institute of Natural Science and Agriculture, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|