1
|
El-Marasy SA, Mostafa RE, Mabrok HB, Khattab MS, Awdan SAE. Protective effect of irbesartan against hepatic ischemia-reperfusion injury in rats: role of ERK, STAT3, and PPAR-γ inflammatory pathways in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1681-1693. [PMID: 39167169 PMCID: PMC11825560 DOI: 10.1007/s00210-024-03301-6] [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: 03/25/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024]
Abstract
This study aimed to elucidate the possible hepatocellular protective role of irbesartan during hepatic ischemia-reperfusion injury (HIRI) and the probable underlying mechanisms. Wistar rats were allocated into four groups: sham; HIRI (control); irbesartan (50 mg/kg) + HIRI; irbesartan (100 mg/kg) + HIRI; irbesartan + GW9662 (1 mg/kg, i.p.) + HIRI. Rats pretreated orally with irbesartan or vehicle for 14 days underwent 45-min hepatic ischemia followed by 60-min reperfusion. Irbesartan preconditioning diminished alanine transaminase (ALT) and aspartate transaminase (AST) serum levels, and reduced extracellular signal-regulated kinase (ERK) and signal transducer and activator of transcription 3 (STAT3). Irbesartan decreased proapoptotic BAX (bcl-2-like protein 4), increased anti-apoptotic B-cell lymphoma 2 (BCL2) hepatic content, and thereby reduced BAX/BCL2 ratio. Moreover, irbesartan preconditioning reduced autophagy-related proteins Beclin1 and LC3 II, and elevated p62 (protein responsible for autophagosome degradation). It elevated proliferator-activated receptor γ (PPAR-γ), and reduced tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) hepatic gene expression. Also, hepatic protein expressions of nuclear factor kappa-B p65 (NF-κB p65) and caspase-3 were lessoned by irbesartan pretreatment in HIRI rats. However, GW9662 abrogated irbesartan's effect on HIRI. The protective effect of irbesartan on HIRI may be mediated by alleviation of ERK, STAT3, and PPAR-γ inflammatory pathways, exerting anti-apoptotic and anti-autophagic effects in HIRI in rats.
Collapse
Affiliation(s)
- Salma A El-Marasy
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt.
| | - Rasha E Mostafa
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Hoda B Mabrok
- Nutrition and Food Science Department, Food Industries and Nutrition Research Institute, National Research Centre, Giza, Egypt
| | - Marwa S Khattab
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Sally A El Awdan
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| |
Collapse
|
2
|
Pandey P, Lakhanpal S, Mahmood D, Kang HN, Kim B, Kang S, Choi J, Moon S, Pandey S, Ballal S, Kumar S, Khan F, Kim B. Bergenin, a bioactive flavonoid: advancements in the prospects of anticancer mechanism, pharmacokinetics and nanoformulations. Front Pharmacol 2025; 15:1481587. [PMID: 39834829 PMCID: PMC11743450 DOI: 10.3389/fphar.2024.1481587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
The natural world is a vast reservoir of exceptionally varied and inventive chemical compositions. Natural products are used as initial compounds to create combinatorial libraries by targeted modifications and then by analyzing their structure-activity connections. This stage is regarded as a crucial milestone in drug discovery and development. Bergenin, a naturally occurring secondary metabolite, has been extracted from several plant components. It is a constituent found in herbal and Ayurvedic preparations. It demonstrates antiviral, antifungal, antitussive, antiplasmodial, anti-inflammatory, antihepatotoxic, antiarrhythmic, antitumor, antiulcerogenic, antidiabetic, and wound healing activities. Bergenin efficiently inhibited the proliferation of human cancer cells by stimulating the production of intracellular reactive oxygen species (ROS), causing DNA damage and leading to cell cycle arrest in the G1/G2 phases by blocking cell signaling pathways. More comprehensive reviews are needed on the anticancer properties of bergenin. Therefore, our review aimed to update the multifaceted benefits of bergenin to the future scientists and researchers, which can be leveraged to formulate safer and novel cancer therapies, while also establishing a robust framework for future investigations into bergenin in cancer treatment. More preclinical and clinical investigations are needed to validate the candidature of bergenin as a potent anticancer agent.
Collapse
Affiliation(s)
- Pratibha Pandey
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, India
- Chitkara Centre for Research and Development, Chitkara University Himanchal Pradesh, Baddi, India
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Danish Mahmood
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia
| | - Han Na Kang
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Byunggyu Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jinwon Choi
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seungjoon Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Shivam Pandey
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, India
| | - Sanjay Kumar
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Rajasthan, India
| | - Fahad Khan
- Center for Global Health Research Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Peng J, Liu S, Liu F, Qin L, Wang Q, He Y, Zhang D, He W, Ke X, Wu F, Wang X. Substitutability evaluation and resource utilization of non-medicinal parts of Ardisia crenata Sims, a traditional ethnic medicine. PHYSIOLOGIA PLANTARUM 2025; 177:e70068. [PMID: 39936472 DOI: 10.1111/ppl.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/16/2024] [Accepted: 12/04/2024] [Indexed: 02/13/2025]
Abstract
The severe scarcity of wild resources of Ardisia crenata Sims (AS) has greatly limited its current clinical and pharmaceutical applications. We aimed to demonstrate the substitutability of stems and leaves for AS roots. In this study, 28 local samples from the Guizhou Province of China were selected for a comprehensive comparison of quality markers, chemical composition and efficacy. HPLC analysis showed that both stems and leaves contained bergenin, the main active ingredient of AS. Metabolomics studies showed that the chemical composition of the three parts was 99% similar. Network pharmacology analysis showed that similar signalling targets including STAT3 and MAPK1, AKT1 and EP300, SRC and TP53, were targeted by three parts. Animal experiments further demonstrated that extracts from the three different parts of the plant relieved the pathological symptoms of acute pharyngitis, and decreased serum inflammatory factors, showing similar efficacy. In sum, these findings strongly proved that stems and leaves can be used as supplements or substitutes for roots, providing a powerful strategy to solve the problem of AS resource shortage and greatly promoting the optimal utilization of AS. The study also anticipated some additional benefits, including the expansion of natural sources of bergenin and the discovery of potential cancer-related targets of AS.
Collapse
Affiliation(s)
- Jie Peng
- Zunyi Medical University, Zunyi, China
| | - Sha Liu
- Zunyi Medical University, Zunyi, China
| | - Fusong Liu
- Zunyi Medical University, Zunyi, China
- State Key Laboratory of Aridland Crop Science / Gansu Agricultural University, Lanzhou, China
| | - Lin Qin
- Zunyi Medical University, Zunyi, China
| | - Qian Wang
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, School of stomatology, Zunyi Medical University, Zunyi, China
| | - Yuqi He
- Zunyi Medical University, Zunyi, China
| | - Delin Zhang
- Clinical Pharmacy, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | | | - Xinyue Ke
- Zunyi Medical University, Zunyi, China
| | - Faming Wu
- Zunyi Medical University, Zunyi, China
| | | |
Collapse
|
4
|
da Rocha GHO, Müller C, Przybylski-Wartner S, Schaller H, Riemschneider S, Lehmann J. AhR-Induced Anti-Inflammatory Effects on a Caco-2/THP-1 Co-Culture Model of Intestinal Inflammation Are Mediated by PPARγ. Int J Mol Sci 2024; 25:13072. [PMID: 39684781 DOI: 10.3390/ijms252313072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The aryl hydrocarbon receptor (AhR) and the peroxisome proliferator-activated receptor γ (PPARγ) are ligand-activated transcription factors that have in recent years been investigated for their anti-inflammatory properties for treatment of inflammatory bowel diseases (IBDs). These are globally prevalent chronic maladies of the gut that lack cost-efficient therapeutical options capable of inducing long-term remission. In the present study, we used an in vitro Transwell® co-culture model composed of Caco-2 epithelial cells in the apical compartment and lipopolysaccharide-treated (LPS) THP-1 macrophages in the basolateral compartment. Secretion of cytokines, disruption of epithelial integrity, and expression of surface markers and junctional proteins were assessed in order to investigate interactions between AhR and PPARγ on the ligand-elicited effects on the control of inflammation. The results revealed that the potent AhR ligand 6-formylindolo[3,2-b]carbazole (FICZ) attenuated LPS-induced IL-6 release by macrophages, which then stabilized Caco-2 monolayer permeability by decreasing claudin-2 expression. These effects were disrupted by GW9662 and to some extent by CH223191, inhibitors of PPARγ and AhR, respectively. Our main findings evidence PPARγ might be a downstream regulator of AhR activation essential for its ligand-based anti-inflammatory effects, suggesting it might be employed as either an auxiliary target or as a biomarker of therapeutical efficacy on AhR-based IBD pharmacotherapy.
Collapse
Affiliation(s)
| | - Claudia Müller
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany
| | - Susanne Przybylski-Wartner
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany
| | - Heidrun Schaller
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany
| | - Sina Riemschneider
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany
| | - Jörg Lehmann
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases [CIMD], 04103 Leipzig, Germany
| |
Collapse
|
5
|
Rezaiezadeh H, Langarizadeh MA, Tavakoli MR, Sabokro M, Banazadeh M, Kohlmeier KA, Shabani M. Therapeutic potential of Bergenin in the management of neurological-based diseases and disorders. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8349-8366. [PMID: 38850305 DOI: 10.1007/s00210-024-03197-2] [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: 02/16/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Originally sourced from plants, Bergenin has been used as a medicinal compound in traditional medicine for centuries, and anecdotal reports suggest a wide range of therapeutic uses. Naturally-occurring and lab-synthesized Bergenin, as well as some of its related compounds, have been shown in in vivo and in vitro studies to alter activity of several enzymes and proteins critical in cellular functioning, including reelin, GSK-3β, Lingo-1, Ten-4, GP-43, Aβ 1-42, P-tau, SOD1,2, GPx, Glx1, NQO1, HO1, PPAR-ɣ, BDNF, VEGF, and STAT6. Additionally, Bergenin alters levels of several cytokines, such as IL-6, IL-1β, TNF-α, and TGF-β. Behavioral and cellular effects of Bergenin have been shown to involve PI3K/Akt, NF-κB, PKC, Nrf2, and Sirt1/FOXO3a pathways. These pathways, enzymes, and proteins have been shown to be important in normal neurological functioning, and/or dysfunctions in these pathways and proteins have been shown to be important in several neuro-based disorders or diseases, which suggests that Bergenin could be therapeutic in management of neuropsychiatric conditions or neurological disorders. In preclinical studies, Bergenin has been shown to be useful for the management of Alzheimer's disease, Parkinson's disease, anxiety, depression, addiction, epilepsy, insomnia, stroke, and potentially, state control. Our review aims to summarize current evidence supporting the conclusion that Bergenin could play a role in treating various neuro-based disorders and that future studies should be conducted to evaluate the mechanisms by which Bergenin could exert its therapeutic effects.
Collapse
Affiliation(s)
- Hojjat Rezaiezadeh
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box, Shiraz, 71345-1583, Iran
| | - Mohammad Amin Langarizadeh
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutics Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Marziye Ranjbar Tavakoli
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutics Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Sabokro
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Banazadeh
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran.
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
6
|
Alanazi ST, Salama SA, Althobaiti MM, Alotaibi RA, AlAbdullatif AA, Musa A, Harisa GI. Alleviation of Copper-Induced Hepatotoxicity by Bergenin: Diminution of Oxidative Stress, Inflammation, and Apoptosis via Targeting SIRT1/FOXO3a/NF-κB Axes and p38 MAPK Signaling. Biol Trace Elem Res 2024:10.1007/s12011-024-04401-3. [PMID: 39347884 DOI: 10.1007/s12011-024-04401-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Despite its biological importance, excess copper induces organ damage, especially to the liver. Disruption of critical signaling cascades that control redox status, inflammatory responses, and cellular apoptosis significantly contributes to the copper-induced hepatotoxicity. The present work explored the hepatoprotective ability of bergenin against the copper-induced hepatotoxicity using male Wistar rats as a mammalian model. The results revealed that bergenin suppressed the copper-evoked histopathological changes and hepatocellular necrosis as indicated by decreased activity of the liver enzymes ALT and AST in the sera of the copper-intoxicated rats. It decreased hepatic copper content and the copper-induced oxidative stress as revealed by reduced lipid peroxidation and improved activity of the antioxidant enzymes thioredoxin reductase, glutathione peroxidase, catalase, and superoxide dismutase. Bergenin downregulated the inflammatory cytokines TNF-α and IL-6, and the inflammatory cell infiltration to the liver tissues. Additionally, it inhibited the copper-induced apoptosis as indicated by significant reduction in caspase-3 activity. At the molecular level, bergenin activated the antioxidant transcription factor FOXO3a, inhibited the nuclear translocation of the inflammatory transcription factor NF-κB, and suppressed the inflammatory signaling molecules p38 MAPK and c-Fos. Interestingly, bergenin improved the expression of the anti-apoptotic protein Bcl2 and reduced the pro-apoptotic protein BAX. Bergenin markedly enhanced the expression of the histone deacetylase protein SIRT1 that regulates activity of NF-κB and FOXO3a. Collectively, these findings highlight the alleviating activity of bergenin against the copper-induced hepatotoxicity via controlling oxidative stress, inflammation, and apoptosis potentially through upregulation of SIRT1, activation of FOXO3a along with suppression of NF-κB and p38 MAPK signaling.
Collapse
Affiliation(s)
- Samyah T Alanazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, 11433, Riyadh, Saudi Arabia
| | - Samir A Salama
- Division of Biochemistry, Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia.
| | - Musaad M Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Rana A Alotaibi
- College of Pharmacy, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Ammar A AlAbdullatif
- Pharmaceutical Care Services, Ministry of the National Guard-Health Affairs, P.O. Box 4616, 31412, Dammam, Saudi Arabia
| | - Arafa Musa
- Department of Pharmacognosy, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Gamaleldin I Harisa
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Zhu Y, Yao L, Guo Y, Zhang J, Xia Y, Wei Z, Dai Y. Bergenin attenuates triptolide-caused premature ovarian failure in mice based on the antioxidant activity. Reprod Toxicol 2024; 126:108608. [PMID: 38735593 DOI: 10.1016/j.reprotox.2024.108608] [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: 09/06/2023] [Revised: 04/16/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
Tripterygium wilfordii (TW) preparations have been utilized in China for treating rheumatoid arthritis and autoimmune diseases. However, their clinical use is limited due to reproductive toxicity, notably premature ovarian failure (POF). Our study aimed to investigate the effect and mechanism of bergenin in attenuating POF induced by triptolide in mice. POF was induced in female ICR mice via oral triptolide administration (50 μg/kg) for 60 days. Mice received bergenin (25, 50, 100 mg/kg, i.g.) or estradiol valerate (EV) (0.1 mg/kg, i.g.) daily, 1 h before triptolide treatment. In vitro, ovarian granulosa cells (OGCs) were exposed to triptolide (100 nM) and bergenin (1, 3, 10 μM). Antioxidant enzyme activity, protein expression, apoptosis rate, and reactive oxygen species (ROS) levels were assessed. The results showed that triptolide-treated mice exhibited evident atrophy, along with an increase in atretic follicles. Bergenin (50, 100 mg/kg) and EV (0.1 mg/kg), orally administered, exerted significant anti-POF effect. Bergenin and EV also decreased apoptosis in mouse ovaries. In vitro, bergenin (1, 3, 10 μM) attenuated triptolide-induced OGCs apoptosis by reducing levels of apoptosis-related proteins. Additionally, bergenin reduced oxidative stress through downregulation of antioxidant enzymes activity and overall ROS levels. Moreover, the combined use with Sh-Nrf2 resulted in a reduced protection of bergenin against triptolide-induced apoptosis of OGCs. Together, bergenin counteracts triptolide-caused POF in mice by inhibiting Nrf2-mediated oxidative stress and preventing OGC apoptosis. Combining bergenin with TW preparations may effectively reduce the risk of POF.
Collapse
Affiliation(s)
- Yanrong Zhu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Lichen Yao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Yilei Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Jing Zhang
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Yufeng Xia
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China.
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China.
| |
Collapse
|
8
|
Shi F, Chen Z, Yao M, Huang Y, Xiao J, Ma L, Mo J, Lin L, Qin Z. Effects of glutaraldehyde and povidone-iodine on apoptosis of grass carp liver and hepatocytes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116078. [PMID: 38335575 DOI: 10.1016/j.ecoenv.2024.116078] [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: 10/21/2023] [Revised: 01/27/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
Since disinfectants are used all over the world to treat illnesses in people and other animals, they pose a major risk to human health. The comprehensive effects of disinfectant treatments on fish liver, especially the impacts on oxidative stress, toxicological effects, transcriptome profiles, and apoptosis, have not yet been fully analyzed. In the current investigation, healthy grass carp were exposed to 80 μg/L glutaraldehyde or 50 μg/L povidone-iodine for 30 days. First, the findings of enzyme activity tests demonstrated that the administration of glutaraldehyde could considerably increase oxidative stress by lowering T-SOD, CAT, and GPx and raising MDA. Furthermore, KEGG research revealed that exposure to glutaraldehyde and povidone-iodine stimulated the PPAR signal pathway. To further elucidate the transcriptome results, the relative expressions of related DEGs in the PPAR signal pathway were verified. Glutaraldehyde induced apoptosis in liver tissue of grass carp; however, it activated cytotoxicity and apoptosis in grass carp hepatocytes when exposed to glutaraldehyde or povidone-iodine. According to the current study, disinfectants can cause the impairment of the immune system, oxidative stress, and attenuation of the PPAR signal pathway in the liver of grass carp, making them detrimental as dietary supplements for grass carp, particularly in the aquaculture sector.
Collapse
Affiliation(s)
- Fei Shi
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Zhilong Chen
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Minshan Yao
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Yao Huang
- The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Jin Xiao
- Department of orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Lixin Ma
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Jilin Mo
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Li Lin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China.
| | - Zhendong Qin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China.
| |
Collapse
|
9
|
Pan S, Mou C, Chen Z. Red recombination enables a wide variety of markerless manipulation of porcine epidemic diarrhea virus genome to generate recombinant virus. Front Cell Infect Microbiol 2024; 13:1338740. [PMID: 38317792 PMCID: PMC10839022 DOI: 10.3389/fcimb.2023.1338740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/27/2023] [Indexed: 02/07/2024] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) is a member of the genera Alphacoronavirus that has been associated with acute watery diarrhea and vomiting in swine. Unfortunately, no effective vaccines and antiviral drugs for PEDV are currently available. Reverse genetics systems are crucial tools for these researches. Here, a PEDV full-length cDNA clone was constructed. Furtherly, three PEDV reporter virus plasmids containing red fluorescent protein (RFP), Nano luciferase (Nluc), or green fluorescence protein (GFP) were generated using Red recombination with the GS1783 E. coli strain. These reporter-expressing recombinant (r) PEDVs showed similar growth properties to the rPEDV, and the foreign genes were stable to culture up to P9 in Vero cells. Using the Nluc-expressing rPEDV, the replication of PEDV was easily quantified, and a platform for rapid anti-PEDV drug screening was constructed. Among the three drugs, Bergenin, Umifenovir hydrochloride (Arbidol), and Ganoderma lucidum triterpenoids (GLTs), we found that GLTs inhibited PEDV replication mainly after the stage of virus "Entry". Overall, this study will broaden insight into the method for manipulating the PEDV genome and provide a powerful tool for screening anti-PEDV agents.
Collapse
Affiliation(s)
- Shuonan Pan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Chunxiao Mou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Zhenhai Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
10
|
Alanazi ST, Harisa GI, Salama SA. Modulating SIRT1, Nrf2, and NF-κB signaling pathways by bergenin ameliorates the cadmium-induced nephrotoxicity in rats. Chem Biol Interact 2024; 387:110797. [PMID: 37949422 DOI: 10.1016/j.cbi.2023.110797] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
In light of the current industrial evolution, exposure to cadmium has become a significant public health concern. Cadmium accumulates in the renal tubular cells and causes nephrotoxicity largely through disruption of the redox homeostasis, induction of inflammation, and suppression of the histone deacetylase SIRT1 expression. The current work aimed at exploring the protective capability of bergenin, a naturally-occurring methyl gallic acid derivative, against the cadmium-evoked nephrotoxicity. Male Wistar rats were treated either with cadmium alone or with cadmium and bergenin for a 7-day experimental period followed by collection of kidney and blood specimens that were subjected to biochemical, molecular, and histological investigations. The results revealed the ability of bergenin to improve the renal functions in the cadmium-intoxicated rats as evidenced by increased glomerular filtration rate, and decreased serum creatinine and blood urea nitrogen. Equally important, bergenin reduced the renal tissue injury and enhanced its redox homeostasis as indicated by decreased protein expression of the kidney injury marker KIM-1, reduced lipid peroxidation, and improved antioxidant potential and histopathological picture of the renal tissues. Mechanistically, bergenin reduced the renal tissue cadmium content, markedly up-regulated protein expression of SIRT1 that regulates inflammation and the redox status of the renal tissues. Additionally, it improved the expression of the major antioxidant transcription factor Nrf2 and its responsive gene products heoxygenase-1 and NAD(P)H quinone dehydrogenase 1 in the cadmium-intoxicated rats. In the same context, bergenin down-regulated the acetylation and the nuclear translocation of the inflammatory transcription factor NF-κB and reduced levels of its responsive gene products TNF-α and IL-1β, as well as the activity of the inflammatory cell infiltration biomarker myeloperoxidase. Collectively, the current study underscores the ameliorating activity of bergenin against the cadmium-evoked nephrotoxicity and highlights modulation of SIRT1, Nrf2, and NF-κB signaling as potential underlining molecular mechanisms.
Collapse
Affiliation(s)
- Samyah T Alanazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Gamaleldin I Harisa
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Cairo, 11751, Egypt.
| | - Samir A Salama
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Cairo, 11751, Egypt; Division of Biochemistry, Department of Pharmacology, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| |
Collapse
|
11
|
Liu XY, Wang YN, Du JS, Chen BH, Liu KY, Feng L, Xiang GS, Zhang SY, Lu YC, Yang SC, Zhang GH, Hao B. Biosynthetic pathway of prescription bergenin from Bergenia purpurascens and Ardisia japonica. FRONTIERS IN PLANT SCIENCE 2024; 14:1259347. [PMID: 38239219 PMCID: PMC10794647 DOI: 10.3389/fpls.2023.1259347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 12/14/2023] [Indexed: 01/22/2024]
Abstract
Bergenin is a typical carbon glycoside and the primary active ingredient in antitussive drugs widely prescribed for central cough inhibition in China. The bergenin extraction industry relies on the medicinal plant species Bergenia purpurascens and Ardisia japonica as their resources. However, the bergenin biosynthetic pathway in plants remains elusive. In this study, we functionally characterized a shikimate dehydrogenase (SDH), two O-methyltransferases (OMTs), and a C-glycosyltransferase (CGT) involved in bergenin synthesis through bioinformatics analysis, heterologous expression, and enzymatic characterization. We found that BpSDH2 catalyzes the two-step dehydrogenation process of shikimic acid to form gallic acid (GA). BpOMT1 and AjOMT1 facilitate the methylation reaction at the 4-OH position of GA, resulting in the formation of 4-O-methyl gallic acid (4-O-Me-GA). AjCGT1 transfers a glucose moiety to C-2 to generate 2-Glucosyl-4-O-methyl gallic acid (2-Glucosyl-4-O-Me-GA). Bergenin production ultimately occurs in acidic conditions or via dehydration catalyzed by plant dehydratases following a ring-closure reaction. This study for the first time uncovered the biosynthetic pathway of bergenin, paving the way to rational production of bergenin in cell factories via synthetic biology strategies.
Collapse
Affiliation(s)
- Xiang-Yu Liu
- College of Agronomy and Biotechnology, National and Local Joint Engineering Research Center on Germplasm Innovation & Utilization of Chinese Medicinal Materials in Southwest China, Key Laboratory of Medicinal Plant Biology of Yunnan Province, Yunnan Agricultural University, Kunming, Yunnan, China
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
| | - Yi-Na Wang
- College of Agronomy and Biotechnology, National and Local Joint Engineering Research Center on Germplasm Innovation & Utilization of Chinese Medicinal Materials in Southwest China, Key Laboratory of Medicinal Plant Biology of Yunnan Province, Yunnan Agricultural University, Kunming, Yunnan, China
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
| | - Jiang-Shun Du
- College of Agronomy and Biotechnology, National and Local Joint Engineering Research Center on Germplasm Innovation & Utilization of Chinese Medicinal Materials in Southwest China, Key Laboratory of Medicinal Plant Biology of Yunnan Province, Yunnan Agricultural University, Kunming, Yunnan, China
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
| | - Bi-Huan Chen
- College of Agronomy and Biotechnology, National and Local Joint Engineering Research Center on Germplasm Innovation & Utilization of Chinese Medicinal Materials in Southwest China, Key Laboratory of Medicinal Plant Biology of Yunnan Province, Yunnan Agricultural University, Kunming, Yunnan, China
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
| | - Kun-Yi Liu
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
| | - Lei Feng
- College of Agronomy and Biotechnology, National and Local Joint Engineering Research Center on Germplasm Innovation & Utilization of Chinese Medicinal Materials in Southwest China, Key Laboratory of Medicinal Plant Biology of Yunnan Province, Yunnan Agricultural University, Kunming, Yunnan, China
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
| | - Gui-Sheng Xiang
- College of Agronomy and Biotechnology, National and Local Joint Engineering Research Center on Germplasm Innovation & Utilization of Chinese Medicinal Materials in Southwest China, Key Laboratory of Medicinal Plant Biology of Yunnan Province, Yunnan Agricultural University, Kunming, Yunnan, China
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
| | - Shuang-Yan Zhang
- College of Agronomy and Biotechnology, National and Local Joint Engineering Research Center on Germplasm Innovation & Utilization of Chinese Medicinal Materials in Southwest China, Key Laboratory of Medicinal Plant Biology of Yunnan Province, Yunnan Agricultural University, Kunming, Yunnan, China
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
| | - Ying-Chun Lu
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
| | - Sheng-Chao Yang
- College of Agronomy and Biotechnology, National and Local Joint Engineering Research Center on Germplasm Innovation & Utilization of Chinese Medicinal Materials in Southwest China, Key Laboratory of Medicinal Plant Biology of Yunnan Province, Yunnan Agricultural University, Kunming, Yunnan, China
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
| | - Guang-Hui Zhang
- College of Agronomy and Biotechnology, National and Local Joint Engineering Research Center on Germplasm Innovation & Utilization of Chinese Medicinal Materials in Southwest China, Key Laboratory of Medicinal Plant Biology of Yunnan Province, Yunnan Agricultural University, Kunming, Yunnan, China
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
| | - Bing Hao
- Yunnan Characteristic Plant Extraction Laboratory, Kunming, Yunnan, China
- College of Tobacco Science, Yunnan Agricultural University, Kunming, Yunnan, China
| |
Collapse
|
12
|
Akhilesh, Chouhan D, Ummadisetty O, Verma N, Tiwari V. Bergenin ameliorates chemotherapy-induced neuropathic pain in rats by modulating TRPA1/TRPV1/NR2B signalling. Int Immunopharmacol 2023; 125:111100. [PMID: 38149571 DOI: 10.1016/j.intimp.2023.111100] [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: 08/30/2023] [Revised: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 12/28/2023]
Abstract
Chemotherapy-induced neuropathic pain (CINP) is one of the most prominent and incapacitating complication associated with chemotherapeutic regimens. The exact mechanisms underlying CINP are not fully understood yet, which hampers the development of effective therapeutics. The current study has been designed to investigate the effect of bergenin on CINP and dissect the underlying cellular and molecular mechanisms. Behavioural responsiveness assays were conducted in rats before and after CINP induction and at different time points post-bergenin treatment. We also measured alterations in tight junction proteins, pro-inflammatory cytokines, microglia activity, transient receptor potential (TRP) channels (TRPV1, TRPA1 and TRPM8) and N-methyl-D-aspartate receptor subtype 2 (NR2B) in dorsal root ganglion (DRG) and spinal tissues of neuropathic rats. Bergenin treatment leads to a significant and dose-dependent reduction in evoked and spontaneous ongoing pain without causing central side effects in neuropathic rats. Furthermore, treatment with bergenin and gabapentin did not affect the baseline pain threshold in healthy, non-chemotherapy-treated rats, as evaluated through tail-flick and tail-clip assays. Chemotherapy administration leads to a significant activation of TRP channels, concurrent with microglial activation, disruption of spinal cord tight junction proteins, and subsequent infiltration of pro-inflammatory cytokines, as well as NR2B activation. Notably, bergenin treatment effectively reversed all of these alterations, with the exception of TRPM8, in both the DRG and spinal cord of neuropathic rats. Findings from the present study suggests that bergenin mitigates neuropathic pain by modulating the TRPA1/TRPV1/NR2B signalling and presents a promising therapeutic avenue for the treatment of chemotherapy-induced neuropathic pain.
Collapse
Affiliation(s)
- Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Obulapathi Ummadisetty
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Nivedita Verma
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India.
| |
Collapse
|
13
|
Zhang L, Tong Y, Fang Y, Pei J, Wang Q, Li G. Exploring the hypolipidemic effects of bergenin from Saxifraga melanocentra Franch: mechanistic insights and potential for hyperlipidemia treatment. Lipids Health Dis 2023; 22:203. [PMID: 38001454 PMCID: PMC10668478 DOI: 10.1186/s12944-023-01973-2] [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: 08/30/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
OBJECTIVE The goal of this study was to explore the hypolipidemic effects of bergenin extracted from Saxifraga melanocentra Franch (S. melanocentra), which is a frequently utilized Tibetan medicinal plant known for its diverse bioactivities. Establishing a quality control system for black stem saxifrage is crucial to ensure the rational utilization of its medicinal resources. METHODS A one-step polyamide medium-pressure liquid chromatography technique was applied to isolate and prepare bergenin from a methanol extract of S. melanocentra. A zebrafish model of hyperlipidemia was used to investigate the potential hypolipidemic effects of bergenin. RESULTS The results revealed that bergenin exhibited substantial hypo efficacy in vivo. Specifically, bergenin significantly reduced the levels of triglycerides (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-c) while simultaneously increasing high-density lipoprotein cholesterol (HDL-c) levels. At the molecular level, bergenin exerted its effects by inhibiting the expression of FASN, SREBF1, HMGCRα, RORα, LDLRα, IL-1β, and TNF while promoting the expression of IL-4 at the transcriptional level. Molecular docking analysis further demonstrated the strong binding affinity of bergenin to proteins such as FASN, SREBF1, HMGCRα, RORα, LDLRα, IL-4, IL-1β, and TNF. CONCLUSIONS Findings indicate that bergenin modulates lipid metabolism by regulating lipid and cholesterol synthesis as well as inflammatory responses through signaling pathways associated with FASN, SREBF1, and RORα. These results position bergenin as a potential candidate for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Li Zhang
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810001, P. R. China
| | - Yingying Tong
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810001, P. R. China
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai, 264005, P. R. China
| | - Yan Fang
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810001, P. R. China
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai, 264005, P. R. China
| | - Jinjin Pei
- Qinba State Key Laboratory of biological resources and ecological environment, Province Key Laboratory of Bioresources, College of Bioscience and bioengineering, QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C, Shaanxi University of Technology, Hanzhong, 723001, Shaanxi, China
| | - Qilan Wang
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810001, P. R. China.
| | - Gang Li
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai, 264005, P. R. China.
| |
Collapse
|
14
|
Sun C, Bai S, Liang Y, Liu D, Liao J, Chen Y, Zhao X, Wu B, Huang D, Chen M, Wu D. The role of Sirtuin 1 and its activators in age-related lung disease. Biomed Pharmacother 2023; 162:114573. [PMID: 37018986 DOI: 10.1016/j.biopha.2023.114573] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023] Open
Abstract
Aging is a major driving factor in lung diseases. Age-related lung disease is associated with downregulated expression of SIRT1, an NAD+-dependent deacetylase that regulates inflammation and stress resistance. SIRT1 acts by inducing the deacetylation of various substrates and regulates several mechanisms that relate to lung aging, such as genomic instability, lung stem cell exhaustion, mitochondrial dysfunction, telomere shortening, and immune senescence. Chinese herbal medicines have many biological activities, exerting anti-inflammatory, anti-oxidation, anti-tumor, and immune regulatory effects. Recent studies have confirmed that many Chinese herbs have the effect of activating SIRT1. Therefore, we reviewed the mechanism of SIRT1 in age-related lung disease and explored the potential roles of Chinese herbs as SIRT1 activators in the treatment of age-related lung disease.
Collapse
|
15
|
Zou H, Gong Y, Ye H, Yuan C, Li T, Zhang J, Ren L. Dietary regulation of peroxisome proliferator-activated receptors in metabolic syndrome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154904. [PMID: 37267691 DOI: 10.1016/j.phymed.2023.154904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 05/15/2023] [Accepted: 05/28/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Peroxisome proliferator-activated receptors (PPARs) are a class of ligand-activated nuclear transcription factors, members of the type nuclear receptor superfamily, with three subtypes, namely PPARα, PPARβ/δ, and PPARγ, which play a key role in the metabolic syndrome. In the past decades, a large number of studies have shown that natural products can act by regulating metabolic pathways mediated by PPARs. PURPOSE This work summarizes the physiological importance and clinical significance of PPARs and reviews the experimental evidence that natural products mediate metabolic syndrome via PPARs. METHODS This study reviews relevant literature on clinical trials, epidemiology, animals, and cell cultures published in NCBI PubMed, Scopus, Web of Science, Google Scholar, and other databases from 2001 to October 2022. Search keywords were "natural product" OR "botanical" OR "phytochemical" AND "PPAR" as well as free text words. RESULTS The modulatory involvement of PPARs in the metabolic syndrome has been supported by prior research. It has been observed that many natural products can treat metabolic syndrome by altering PPARs. The majority of currently described natural compounds are mild PPAR-selective agonists with therapeutic effects that are equivalent to synthetic medicines but less harmful adverse effects. CONCLUSION PPAR agonists can be combined with natural products to treat and prevent metabolic syndrome. Further human investigations are required because it is unknown how natural products cause harm and how they might have negative impacts.
Collapse
Affiliation(s)
- Haoyang Zou
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Yiyao Gong
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Haiqing Ye
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Cuiping Yuan
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun 130033, China
| | - Tiezhu Li
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun 130033, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Li Ren
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
16
|
Mao B, Yuan W, Wu F, Yan Y, Wang B. Autophagy in hepatic ischemia-reperfusion injury. Cell Death Discov 2023; 9:115. [PMID: 37019879 PMCID: PMC10076300 DOI: 10.1038/s41420-023-01387-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 04/07/2023] Open
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is a major complication of liver resection or liver transplantation that can seriously affect patient's prognosis. There is currently no definitive and effective treatment strategy for HIRI. Autophagy is an intracellular self-digestion pathway initiated to remove damaged organelles and proteins, which maintains cell survival, differentiation, and homeostasis. Recent studies have shown that autophagy is involved in the regulation of HIRI. Numerous drugs and treatments can change the outcome of HIRI by controlling the pathways of autophagy. This review mainly discusses the occurrence and development of autophagy, the selection of experimental models for HIRI, and the specific regulatory pathways of autophagy in HIRI. Autophagy has considerable potential in the treatment of HIRI.
Collapse
Affiliation(s)
- Benliang Mao
- College of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Wei Yuan
- Department of General Surgery, Guangzhou Red Cross Hospital affiliated to Jinan University, Guangzhou, China
| | - Fan Wu
- Department of General Surgery, Guangzhou Red Cross Hospital affiliated to Jinan University, Guangzhou, China
| | - Yong Yan
- Department of General Surgery, Guangzhou Red Cross Hospital affiliated to Jinan University, Guangzhou, China
| | - Bailin Wang
- College of Clinical Medicine, Guizhou Medical University, Guiyang, China.
- Department of General Surgery, Guangzhou Red Cross Hospital affiliated to Jinan University, Guangzhou, China.
| |
Collapse
|
17
|
Gan Y, Li X, Han S, Zhou L, Li W. Targeting Mcl-1 Degradation by Bergenin Inhibits Tumorigenesis of Colorectal Cancer Cells. Pharmaceuticals (Basel) 2023; 16:241. [PMID: 37259388 PMCID: PMC9965350 DOI: 10.3390/ph16020241] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 09/29/2023] Open
Abstract
Myeloid leukemia 1 (Mcl-1) is frequently overexpressed in human malignancies and emerged as a promising drug target. In this study, we verified the inhibitory effect of bergenin on colorectal cancer cells both in vivo and in vitro. In an in vitro setting, bergenin significantly reduced the viability and colony formation and promoted apoptosis of CRC cells dose-dependently. Bergenin decreased the activity of Akt/GSK3β signaling and enhanced the interaction between FBW7 and Mcl-1, which eventually induced Mcl-1 ubiquitination and degradation. Using the HA-Ub K48R mutant, we demonstrated that bergenin promotes Mcl-1 K48-linked polyubiquitination and degradation. In vivo studies showed that bergenin significantly reduced tumor size and weight without toxicity to vital organs in mice. Overall, our results support the role of bergenin in inhibiting CRC cells via inducing Mcl-1 destruction, suggesting that targeting Mcl-1 ubiquitination could be an alternative strategy for antitumor therapy.
Collapse
Affiliation(s)
- Yu Gan
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Shuangze Han
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Li Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, The Third Xiangya Hospital, Central South University, Changsha 410008, China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| |
Collapse
|
18
|
Luan Y, Luo Y, Deng M. New advances in Nrf2-mediated analgesic drugs. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154598. [PMID: 36603339 DOI: 10.1016/j.phymed.2022.154598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/24/2022] [Accepted: 12/10/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Oxidative stress is an inevitable process that occurs during life activities, and it can participate in the development of inflammation. Although great progress has been made according to research examining analgesic drugs and therapies, there remains a need to develop new analgesic drugs to fill certain gaps in both the experimental and clinical space. PURPOSE This review reports the research and preclinical progress of this class of analgesics by summarizing known nuclear factor E-2-related factor-2 (Nrf2) pathway-modulating substances. STUDY DESIGN We searched and reported experiments that intervene in the Nrf2 pathway and its various upstream and downstream molecules for analgesic therapy. METHODS The medical literature database (PubMed) was searched for experimental studies examining the reduction of pain in animals through the Nrf2 pathway, the research methods were analyzed, and the pathways were classified and reported according to the pathway of these experimental interventions. RESULTS Humans have identified a variety of substances that can fight pain by regulating the expression of Nrf2 and its upstream and downstream pathways. CONCLUSION The Nrf2 pathway exerts anti-inflammatory activity by regulating oxidative stress, thereby playing a role in the fight against pain.
Collapse
Affiliation(s)
- Yifan Luan
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410013, China
| | - Yaping Luo
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410013, China
| | - Meichun Deng
- Department of Biochemistry and Molecular Biology & Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, Hunan 410013, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
19
|
Hu Y, Lin L, Liu K, Liu E, Han S, Gong Z, Xiao W. L-Theanine alleviates heat stress-induced impairment of immune function by regulating the p38 MAPK signalling pathway in mice. Food Funct 2023; 14:335-343. [PMID: 36511090 DOI: 10.1039/d2fo02775e] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
With the current trend of global warming, heat stress-induced impairment could seriously endanger human health. L-Theanine is a non-protein amino acid in tea with various biological activities, including immunoregulatory, anti-anxiety, and anti-oxidation. However, its effect on immune function under heat stress and the underlying mechanism are currently unclear. In this study, male BALB/c mice were used as experimental objects to explore the effect of L-theanine on heat stress-induced changes in immune function and its mechanism. Three doses of L-theanine were used: low (100 mg kg-1 d-1), medium (200 mg kg-1 d-1), and high (400 mg kg-1 d-1). Treatment with L-theanine could attenuate the heat stress-induced reductions in body weight and feed intake in mice, alleviate damage in the liver and jejunum, and inhibit the inflammatory factors IL-6, IL-1β, and TNF-α. Aspartate aminotransferase and alanine transaminase activity levels and the malondialdehyde content decreased, while the IgA, IgM, and IgG contents increased in response to L-theanine. It is possible that L-theanine affects the P38 signalling pathway and inhibits the increase in p-P65/P65 caused by the overexpression of HSP27 and regulation of PPAR-γ and Foxp3 proteins, thereby alleviating immune dysfunction caused by heat stress.
Collapse
Affiliation(s)
- Yuan Hu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.,Hunan Agricultural University, Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha, Hunan 410128, China.,Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, China
| | - Ling Lin
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.,Hunan Agricultural University, Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha, Hunan 410128, China.,Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, China
| | - Kehong Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.,Hunan Agricultural University, Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha, Hunan 410128, China.,Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, China
| | - Enshuo Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.,Hunan Agricultural University, Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha, Hunan 410128, China.,Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, China
| | - Shumin Han
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.,Hunan Agricultural University, Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha, Hunan 410128, China.,Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, China
| | - Zhihua Gong
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.,Hunan Agricultural University, Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha, Hunan 410128, China.,Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, China
| | - Wenjun Xiao
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.,Hunan Agricultural University, Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha, Hunan 410128, China.,Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, China
| |
Collapse
|
20
|
Salama SA, Elshafey MM. Cross-talk between PPARγ, NF-κB, and p38 MAPK signaling mediates the ameliorating effects of bergenin against the iron overload-induced hepatotoxicity. Chem Biol Interact 2022; 368:110207. [DOI: 10.1016/j.cbi.2022.110207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/27/2022] [Accepted: 10/02/2022] [Indexed: 11/03/2022]
|
21
|
Ishtiaq SM, Arshad MI, Khan JA. PPARγ signaling in hepatocarcinogenesis: Mechanistic insights for cellular reprogramming and therapeutic implications. Pharmacol Ther 2022; 240:108298. [PMID: 36243148 DOI: 10.1016/j.pharmthera.2022.108298] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 11/30/2022]
Abstract
Liver cancer or hepatocellular carcinoma (HCC) is leading cause of cancer-related mortalities globally. The therapeutic approaches for chronic liver diseases-associated liver cancers aimed at modulating immune check-points and peroxisome proliferator-activated receptor gamma (PPARγ) signaling pathway during multistep process of hepatocarcinogenesis that played a dispensable role in immunopathogenesis and outcomes of disease. Herein, the review highlights PPARγ-induced effects in balancing inflammatory (tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, IL-1) and anti-inflammatory cytokines (IL-10, transforming growth factor beta (TGF-β), and interplay of PPARγ, hepatic stellate cells and fibrogenic niche in cell-intrinsic and -extrinsic crosstalk of hepatocarcinogenesis. PPARγ-mediated effects in pre-malignant microenvironment promote growth arrest, cell senescence and cell clearance in liver cancer pathophysiology. Furthermore, PPARγ-immune cell axis of liver microenvironment exhibits an immunomodulation strategy of resident immune cells of the liver (macrophages, natural killer cells, and dendritic cells) in concomitance with current clinical guidelines of the European Association for Study of Liver Diseases (EASL) for several liver diseases. Thus, mechanistic insights of PPARγ-associated high value targets and canonical signaling suggest PPARγ as a possible therapeutic target in reprogramming of hepatocarcinogenesis to decrease burden of liver cancers, worldwide.
Collapse
Affiliation(s)
- Syeda Momna Ishtiaq
- Institute of Physiology and Pharmacology, University of Agriculture, Faisalabad 38040, Pakistan
| | | | - Junaid Ali Khan
- Department of Pharmacology and Physiology, MNS University of Agriculture, Multan 60000, Pakistan.
| |
Collapse
|
22
|
Chen T, Su S, Yang Z, Zhang D, Li Z, Lu D. Srolo Bzhtang reduces inflammation and vascular remodeling via suppression of the MAPK/NF-κB signaling pathway in rats with pulmonary arterial hypertension. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115572. [PMID: 35872290 DOI: 10.1016/j.jep.2022.115572] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/14/2022] [Accepted: 07/17/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Srolo Bzhtang (SBT), which consists of Solms-laubachia eurycarpa, Bergenia purpurascens, Glycyrrhiza uralensis, and lac secreted by Laccifer lacca Kerr (Lacciferidae Cockerell), is a well-known traditional Tibetan medicinal formula and was documented to cure "lung-heat" syndrome by eliminating "chiba" in the ancient Tibetan medical work Four Medical Tantras (Rgyud bzhi). Clinically, it is a therapy for pulmonary inflammatory disorders, such as pneumonia, chronic bronchitis, and chronic obstructive pulmonary disease. However, whether and how SBT participates in pulmonary arterial hypertension (PAH) is still unclear. AIM OF THE STUDY We aimed to determine the role of SBT in attenuating pulmonary arterial pressure and vascular remodeling caused by monocrotaline (MCT) and hypoxia. To elucidate the potential mechanism underlying SBT-mediated PAH, we investigated the changes in inflammatory cytokines and mitogen-activated protein kinase (MAPK)/nuclear factor-kappa B (NF-κB) signaling pathway. MATERIALS AND METHODS MCT- and hypoxia-induced PAH rat models were used. After administering SBT for four weeks, the rats were tested for hemodynamic indicators, hematological changes, pulmonary arterial morphological changes, and the levels of interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α in serum and lung tissues. Protein expression of the MAPK/NF-κB signaling pathway was determined using western blotting. RESULTS SBT reduced pulmonary arterial pressure, vascular remodeling, and the levels of inflammatory cytokines induced by MCT and hypoxia in rats. Furthermore, SBT significantly suppressed the MAPK/NF-κB signaling pathway. CONCLUSIONS To our knowledge, this is the first study to demonstrate that SBT alleviates MCT- and hypoxia-induced PAH in rats, which is related to its anti-inflammatory actions involving inhibition of the MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Tingting Chen
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, PR China; Medical College, Qinghai University, Xining, 810001, PR China
| | - Shanshan Su
- Technical Center of Xining Customs (Key Laboratory of Food Safety Research In Qinghai Province), Xining, 810003, PR China
| | - Zhanting Yang
- Medical College, Qinghai University, Xining, 810001, PR China
| | - Dejun Zhang
- School of Ecological and Environmental Engineering, Qinghai University, Xining, 810016, PR China
| | - Zhanqiang Li
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, PR China; Medical College, Qinghai University, Xining, 810001, PR China.
| | - Dianxiang Lu
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, 810001, PR China; Medical College, Qinghai University, Xining, 810001, PR China.
| |
Collapse
|
23
|
Singla RK, Dhonchak K, Sodhi RK, Arockia Babu M, Madan J, Madaan R, Kumar S, Sharma R, Shen B. Bergenin ameliorates cognitive deficits and neuropathological alterations in sodium azide-induced experimental dementia. Front Pharmacol 2022; 13:994018. [PMID: 36249784 PMCID: PMC9556967 DOI: 10.3389/fphar.2022.994018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Bergenin, 4-O-methyl gallic acid glucoside, is a bioactive compound found in the cortex of Mallotus japonicus (L.f.) Müll.Arg. along with many other natural resources including that from Bergenia species. The present study delineates the neuroprotective potential of bergenin through the modulation of PPAR-γ receptors. Method: Dementia was induced in the Wistar rats by intraperitoneal (i.p.) administration of sodium azide (12.5 mg/kg for the first 5 days followed by 10 mg/kg for the next 9 days). The rats were then exposed to the Morris water maze test to assess the effect on cognitive abilities followed by a series of biochemical and histopathological evaluations. Results: Sodium azide-treated rats exhibited a severe deterioration of memory as suggested by poor performance in the spatial learning task in addition to the enhancement of brain acetylcholinesterase potential, oxidative stress, inflammation, and amyloid-β (Aβ) accumulation. Administration of bergenin to sodium azide-treated rats significantly recovered cognition and related biochemical variations. Further, co-administration of Bisphenol A diglycidyl ether (BADGE), a PPAR-γ antagonist with bergenin challenged its neuroprotective effects. Conclusions: The findings of our study exhibit that the cognitive restoration potential of bergenin may be attributed to its modulatory effects against cholinesterase, oxidative stress, and inflammatory markers, as well as its neuroprotective actions, thus aligning it as a possible therapy for Alzheimer's disease-related dementia. The study also fortifies the significance of PPAR-γ receptors in dementia.
Collapse
Affiliation(s)
- Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Konika Dhonchak
- Department of Pharmacology, Chandigarh College of Pharmacy, Mohali, India
| | - Rupinder K. Sodhi
- Department of Pharmacology, Chandigarh College of Pharmacy, Mohali, India
| | - M. Arockia Babu
- Department of Pharmacology, Chandigarh College of Pharmacy, Mohali, India
| | - Jitender Madan
- National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Reecha Madaan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Suresh Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
24
|
Bergenin has neuroprotective effects in mice with ischemic stroke through antioxidative stress and anti-inflammation via regulating Sirt1/FOXO3a/NF-κB signaling. Neuroreport 2022; 33:549-560. [DOI: 10.1097/wnr.0000000000001789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Li X, Bai R, Bai Y, Shi X, Yang Y, Xu S. ROS-mediated PPAR/RXR inhibition contributes to acetochlor-induced apoptosis and autophagy in Ctenopharyngodon idella hepatic cells. FISH & SHELLFISH IMMUNOLOGY 2022; 128:684-694. [PMID: 36028057 DOI: 10.1016/j.fsi.2022.08.053] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 06/15/2023]
Abstract
Acetochlor is a high-volume herbicide whose widespread use threatens ecosystems and affects aquaculture. Apoptosis and autophagy are important causes of hepatotoxicity caused by toxicants, which can be mediated by oxidative stress and the inhibition of PPAR/RXR pathway. However, the mechanism of acetochlor on fish hepatocyte damage still needs to be further investigated. Therefore, we treated the Ctenopharyngodon idella hepatic cell line (L8824 cells) with different concentrations (10, 20, and 40 μM) of acetochlor and/or ROS scavenger NAC (1 mM) for 24 h. The results showed that acetochlor decreased the cell viability in a dose-dependent manner. AO/EB staining and flow cytometry verified the increased apoptotic rates. Quantitative analysis of gene expression levels or protein expression levels displayed that the expression levels of Beclin1, P62, LC3B, BAX, and cleaved Casp3 were increased, and the expression of BCL2 was reduced. Besides, we detected the increased ROS contents and decreased PPAR/RXR pathway expressions after acetochlor treatment. The clearance of ROS alleviated the inhibition of the PPAR/RXR pathway and lightened apoptosis and autophagy under acetochlor stress. Overall, these results revealed that acetochlor exposure triggered BCL2/BAX/Casp3-cascaded apoptosis and Beclin1-dependent autophagy through ROS-mediated PPAR/RXR inhibition. The results partially explain the toxicological mechanism of acetochlor and provide targets for the development of its antidote.
Collapse
Affiliation(s)
- Xiaojing Li
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Ruichen Bai
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yichen Bai
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xu Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yuhong Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, 150030, PR China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
26
|
Ma Y, Jiao Z, Liu X, Zhang Q, Piao C, Xu J, Wang H. Protective effect of adipose-derived stromal cell-secretome attenuate autophagy induced by liver ischemia–reperfusion and partial hepatectomy. STEM CELL RESEARCH & THERAPY 2022; 13:427. [PMID: 35987696 PMCID: PMC9392224 DOI: 10.1186/s13287-022-03109-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/03/2022] [Indexed: 11/25/2022]
Abstract
Background The therapeutic effects of adipose-derived mesenchymal stromal cells (ADSCs) may be mainly mediated by their paracrine effects. The ADSC-secretome can ameliorate hepatic ischemia–reperfusion injury (IRI). We explored the therapeutic effect of the ADSC-secretome from the perspective of excessive hepatocyte autophagy induced by hepatic IRI. Methods We established a miniature pig model of hepatic ischemia–reperfusion (I/R) and hepatectomy using a laparoscopic technique and transplanted ADSCs and the ADSC-secretome into the liver parenchyma immediately after surgery. Liver injury and hepatocyte autophagy were evaluated by histopathological examination and assessment of relevant cytokines and other factors. Results The results showed that the ADSC-secretome alleviated the pathological changes of liver tissue and the microstructural damage of hepatocytes after IRI. Moreover, the expression levels of autophagy-related markers including Beclin-1, ATG5, ATG12, and LC3II/LC3I decreased, whereas those of p62 increased during phagophore expansion. Furthermore, the expression levels of markers related to the autophagy inhibition pathway phosphatidylinositol-3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR), including PI3K, Akt, and mTOR, increased. Conclusion The ADSC-secretome attenuates hepatic I/R and hepatectomy-induced liver damage by inhibiting autophagy, which is possibly mediated by activation of the PI3K/Akt/mTOR signaling pathway. In addition, there was no significant difference between ADSCs and the ADSC-secretome in the regulation of hepatocyte autophagy. Therefore, ADSCs may improve the excessive autophagy-induced injury of hepatocytes in hepatic I/R and hepatectomy through paracrine effect. Our findings provide new insight into the therapeutic potential of cell-free products, which could replace cell therapy in liver diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03109-2.
Collapse
|
27
|
Luteolin Pretreatment Attenuates Hepatic Ischemia-Reperfusion Injury in Mice by Inhibiting Inflammation, Autophagy, and Apoptosis via the ERK/PPARα Pathway. PPAR Res 2022; 2022:8161946. [PMID: 35966821 PMCID: PMC9366205 DOI: 10.1155/2022/8161946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/25/2022] [Accepted: 07/16/2022] [Indexed: 11/18/2022] Open
Abstract
Hepatic ischemia-reperfusion (IR) injury is a clinically significant process that frequently occurs in liver transplantation, partial hepatectomy, and hemorrhagic shock. The aim of this study was to explore the effectiveness of luteolin in hepatic IR injury and the underlying mechanism. BALB/c mice were randomly divided into six groups, including normal controls (NC), luteolin (50 mg/kg), sham procedure, IR+25 mg/kg luteolin, and IR+50 mg/kg luteolin group. Serum and tissue samples were collected at 6 and 24 h after reperfusion to assay liver enzymes, inflammatory factors, expression of proteins associated with apoptosis and autophagy, and factors associated with the extracellular signal-regulated kinase/peroxisome proliferator-activated receptor alpha (ERK/PPARα) pathway. Luteolin preconditioning decreased hepatocyte injury caused by ischemia-reperfusion, downregulated inflammatory factors, and inhibited apoptosis and autophagy. Luteolin also inhibited ERK phosphorylation and activated PPARα.
Collapse
|
28
|
Bergenin from Bergenia Species Produces a Protective Response against Myocardial Infarction in Rats. Processes (Basel) 2022. [DOI: 10.3390/pr10071403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Bergenin is a phenolic glycoside that has been reported to occur naturally in several plant species, reported as a cardioprotective. However, bergenin, one of the important phytochemicals in these plants, is still not reported as a cardioprotective. The present study was designed to investigate the cardioprotective effects of bergenin on isoproterenol-induced myocardial infarction in rats. Bergenin and atenolol were administered through intraperitoneal (i.p.) injection to Sprague Dawley (SD) rats in separate experiments for five (5) days. At the end of this period, rats were administered isoproterenol (80 mg/kg s.c.) to induce myocardial injury. After induction, rats were anaesthetized to record lead II ECG, then sacrificed, blood was collected to analyze cardiac marker enzymes, and a histopathological study of the heart tissues was also performed. Pretreatment with bergenin showed a significant decrease in ST-segment elevation, deep Q-wave, infarct size, and also normalized cardiac marker enzymes (cTnI, CPK, CK-MB, LDH, ALT, and AST), particularly at 3 mg/kg, as compared to isoproterenol treated group. Our findings revealed, for the first time, the use of glycoside bergenin as a potential cardioprotective agent against the isoproterenol-induced MI in rats.
Collapse
|
29
|
p62-Nrf2 Regulatory Loop Mediates the Anti-Pulmonary Fibrosis Effect of Bergenin. Antioxidants (Basel) 2022; 11:antiox11020307. [PMID: 35204190 PMCID: PMC8868171 DOI: 10.3390/antiox11020307] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 11/20/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) can severely disrupt lung function, leading to fatal consequences, and there is currently a lack of specific therapeutic drugs. Bergenin is an isocoumarin compound with lots of biological functions including antioxidant activity. This study evaluated the potential beneficial effects of bergenin on pulmonary fibrosis and investigated the possible mechanisms. We found that bergenin alleviated bleomycin-induced pulmonary fibrosis by relieving oxidative stress, reducing the deposition of the extracellular matrix (ECM) and inhibiting the formation of myofibroblasts. Furthermore, we showed that bergenin could induce phosphorylation and expression of p62 and activation of Nrf2, Nrf2 was required for bergenin-induced p62 upregulation, and p62 knockdown reduced bergenin-induced Nrf2 activity. More importantly, knockdown of Nrf2 or p62 could abrogate the antioxidant activity of bergenin and the inhibition effect of bergenin on TGF-β-induced ECM deposition and myofibroblast differentiation. Thereby, a regulatory loop is formed between p62 and Nrf2, which is an important target for bergenin aimed at treating pulmonary fibrosis.
Collapse
|
30
|
Jiang Y, Gong Q, Gong Y, Zhuo C, Huang J, Tang Q. Vitexin attenuates non-alcoholic fatty liver disease (NAFLD) lipid accumulation in high fat-diet fed mice by activating autophagy and reducing endoplasmic reticulum (ER) stress in liver. Biol Pharm Bull 2022; 45:260-267. [PMID: 35034930 DOI: 10.1248/bpb.b21-00716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become prevalent worldwide, but sufficient pharmaceutical treatments for this condition are lacking. Previous literature suggests that vitexin offers beneficial effects in the treatment of NAFLD, but the underlying mechanisms are not well understood. In this study, the in vivo effects of vitexin were investigated in high-fat-diet (HFD)-induced NAFLD mice. Liver pathology, biochemical parameters, lipid levels, hepatocyte ultrastructure, and related regulatory proteins were measured at the end of treatment. Treatment consisted of four weeks of daily administration of vitexin at a dose of 6 mg/kg of body weight. This treatment markedly improved hepatic architecture, attenuated lipid accumulation, and regulated lipid abnormalities. In addition, the treatment reduced endoplasmic reticulum (ER) stress, restored mitochondrial biological proteins, and increased autophagy. Furthermore, the treatment increased PPAR-r protein, which was inhibited by HFD. Thus, it was speculated that vitexin degraded lipids in HFD-induced NAFLD mice liver by inducing autophagy and restoring both ER and mitochondrial biological proteins.
Collapse
Affiliation(s)
- Yan Jiang
- Medical College, Guangxi University.,Guixi Key Laboratory for High Incidence Diseases, Youjiang Medical University for Nationalities
| | - Qiming Gong
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities
| | - Yuanxun Gong
- Guixi Key Laboratory for High Incidence Diseases, Youjiang Medical University for Nationalities
| | - Chenyi Zhuo
- Department of Hepatobiliary surgery, Affiliated hospital of Youjiang Medical University for Nationalities
| | - Jinmei Huang
- Graduate School, Guangxi University of Chinese Medicine
| | - Qianli Tang
- Medical College, Guangxi University.,Guixi Key Laboratory for High Incidence Diseases, Youjiang Medical University for Nationalities
| |
Collapse
|
31
|
Zhang G, Wang H, Zhang Q, Zhao Z, Zhu W, Zuo X. Bergenin alleviates H 2 O 2 -induced oxidative stress and apoptosis in nucleus pulposus cells: Involvement of the PPAR-γ/NF-κB pathway. ENVIRONMENTAL TOXICOLOGY 2021; 36:2541-2550. [PMID: 34499403 DOI: 10.1002/tox.23368] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/26/2021] [Accepted: 08/29/2021] [Indexed: 06/13/2023]
Abstract
Bergenin is a C-glucoside of 4-O-methyl gallic acid with a variety of biological activities, such as antioxidant and anti-inflammatory. Herein, we investigated the involvement of bergenin in the protective effect against H2 O2 -induced oxidative stress and apoptosis in human nucleus pulposus cells (HNPCs) and the underlying mechanisms. HNPCs were cotreated with various concentrations of bergenin and 200 μM H2 O2 for 24 h. Cell viability was detected by Cell Counting Kit-8 and lactate dehydrogenase release assays. Reactive oxygen species (ROS) was evaluated utilizing 2',7'-dichlorofluorescein-diacetate. Superoxide dismutase (SOD) and catalase (CAT) activities and glutathione (GSH) levels were measured to assess oxidative stress. Apoptosis was evaluated using terminal deoxynucleotidyl transferase dUTP nick end labeling and caspase-3/7 activity assays. Expression of protein was determined by western blotting. Results indicated that treatment with bergenin significantly alleviated H2 O2 -induced viability reduction and ROS overproduction in HNPCs in a dose-dependent manner. Bergenin alleviated H2 O2 -induced oxidative stress in HNPCs by increased activity of superoxide dismutase and level of glutathione peroxidase. H2 O2 -induced apoptosis and activity of caspase-3/7 were also suppressed by bergenin treatment in HNPCs. Western blotting showed that H2 O2 -induced decrease in expression of peroxisome proliferator-activated receptor γ (PPAR-γ) and increase in nuclear factor κB (NF-κB) were inhibited by bergenin. However, the inhibitory effect of bergenin on H2 O2 -induced viability reduction, oxidative stress and apoptosis were noticeably abrogated in PPAR-γ knockdown HNPCs. In conclusion, our results indicated that bergenin alleviates H2 O2 -induced oxidative stress and apoptosis in HNPCs by activating PPAR-γ and suppressing NF-κB pathway.
Collapse
Affiliation(s)
- Gaofeng Zhang
- Spondyloarthropathy Department, Nanyang Nanshi Hospital of He'nan Province, Nanyang, People's Republic of China
| | - Hai Wang
- Department of Image, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, People's Republic of China
| | - Qianxi Zhang
- Department of Pain Management, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, People's Republic of China
| | - Zhengyu Zhao
- Department of Image, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, People's Republic of China
| | - Wenyang Zhu
- Department of Image, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, People's Republic of China
| | - Xiaohua Zuo
- Department of Pain Management, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, People's Republic of China
| |
Collapse
|
32
|
Madaan R, Singla RK, Kumar S, Dubey AK, Kumar D, Sharma P, Bala R, Singla S, Shen B. Bergenin - a biologically active scaffold: Nanotechnological perspectives. Curr Top Med Chem 2021; 22:132-149. [PMID: 34649489 DOI: 10.2174/1568026621666211015092654] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 02/08/2023]
Abstract
Bergenin, 4-O-methyl gallic acid glucoside, is a bioactive compound present in various plants belonging to different families. The present work compiles scattered information on pharmacology, structure activity relationship and nanotechnological aspects of bergenin, collected from various electronic databases such as Sci Finder, PubMed, Google scholar, etc. Bergenin has been reported to exhibit hepatoprotective, anti-inflammatory, anticancer, neuroprotective, antiviral and antimicrobial activities. Molecular docking studies have shown that isocoumarin pharmacophore of bergenin is essential for its bioactivities. Bergenin holds a great potential to be used as lead molecule and also as a therapeutic agent for development of more efficacious and safer semisynthetic derivatives. Nanotechnological concepts can be employed to overcome poor bioavailability of bergenin. Finally, it is concluded that bergenin can be emerged as clinically potential medicine in modern therapeutics.
Collapse
Affiliation(s)
- Reecha Madaan
- Chitkara College of Pharmacy, Chitkara University Punjab. India
| | - Rajeev K Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan. China
| | - Suresh Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala- Punjab. India
| | - Ankit Kumar Dubey
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai-600036, Tamil Nadu. India
| | - Dinesh Kumar
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai-600036, Tamil Nadu. India
| | - Pooja Sharma
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala- Punjab. India
| | - Rajni Bala
- Chitkara College of Pharmacy, Chitkara University Punjab. India
| | - Shailja Singla
- iGlobal Research and Publishing Foundation, New Delhi. India
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan. China
| |
Collapse
|
33
|
Hu C, Zhao L, Zhang F, Li L. Regulation of autophagy protects against liver injury in liver surgery-induced ischaemia/reperfusion. J Cell Mol Med 2021; 25:9905-9917. [PMID: 34626066 PMCID: PMC8572770 DOI: 10.1111/jcmm.16943] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 08/10/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022] Open
Abstract
Transient ischaemia and reperfusion in liver tissue induce hepatic ischaemia/reperfusion (I/R) tissue injury and a profound inflammatory response in vivo. Hepatic I/R can be classified into warm I/R and cold I/R and is characterized by three main types of cell death, apoptosis, necrosis and autophagy, in rodents or patients following I/R. Warm I/R is observed in patients or animal models undergoing liver resection, haemorrhagic shock, trauma, cardiac arrest or hepatic sinusoidal obstruction syndrome when vascular occlusion inhibits normal blood perfusion in liver tissue. Cold I/R is a condition that affects only patients who have undergone liver transplantation (LT) and is caused by donated liver graft preservation in a hypothermic environment prior to entering a warm reperfusion phase. Under stress conditions, autophagy plays a critical role in promoting cell survival and maintaining liver homeostasis by generating new adenosine triphosphate (ATP) and organelle components after the degradation of macromolecules and organelles in liver tissue. This role of autophagy may contribute to the protection of hepatic I/R‐induced liver injury; however, a considerable amount of evidence has shown that autophagy inhibition also protects against hepatic I/R injury by inhibiting autophagic cell death under specific circumstances. In this review, we comprehensively discuss current strategies and underlying mechanisms of autophagy regulation that alleviates I/R injury after liver resection and LT. Directed autophagy regulation can maintain liver homeostasis and improve liver function in individuals undergoing warm or cold I/R. In this way, autophagy regulation can contribute to improving the prognosis of patients undergoing liver resection or LT.
Collapse
Affiliation(s)
- Chenxia Hu
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingfei Zhao
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Institute of Nephrology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fen Zhang
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lanjuan Li
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
34
|
Development of mode of action networks related to the potential role of PPARγ in respiratory diseases. Pharmacol Res 2021; 172:105821. [PMID: 34403731 DOI: 10.1016/j.phrs.2021.105821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 11/30/2022]
Abstract
The peroxisome proliferator-activated receptor γ (PPARγ) is a key transcription factor, operating at the intercept of metabolic control and immunomodulation. It is ubiquitously expressed in multiple tissues and organs, including lungs. There is a growing body of information supporting the role of PPARγ signalling in respiratory diseases. The aim of the present study was to develop mode of action (MoA) networks reflecting the relationships between PPARγ signalling and the progression/alleviation of a spectrum of lung pathologies. Data mining was performed using the resources of the NIH PubMed and PubChem information systems. By linking available data on pathological/therapeutic effects of PPARγ modulation, knowledge-based MoA networking at different levels of biological organization (molecular, cellular, tissue, organ, and system) was performed. Multiple MoA networks were developed to relate PPARγ modulation to the progress or the alleviation of pulmonary disorders, triggered by diverse pathogenic, genetic, chemical, or mechanical factors. Pharmacological targeting of PPARγ signalling was discussed with regard to ligand- and cell type-specific effects in the context of distinct disease inductor- and disease stage-dependent patterns. The proposed MoA networking analysis allows for a better understanding of the potential role of PPARγ modulation in lung pathologies. It presents a mechanistically justified basis for further computational, experimental, and clinical monitoring studies on the dynamic control of PPARγ signalling in respiratory diseases.
Collapse
|
35
|
Li X, Wang Y, Liang J, Bi Z, Ruan H, Cui Y, Ma L, Wei Y, Zhou B, Zhang L, Zhou H, Yang C. Bergenin attenuates bleomycin-induced pulmonary fibrosis in mice via inhibiting TGF-β1 signaling pathway. Phytother Res 2021; 35:5808-5822. [PMID: 34375009 DOI: 10.1002/ptr.7239] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/13/2021] [Accepted: 07/23/2021] [Indexed: 01/01/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease characterized by epithelial cell damage, fibroblast activation, and collagen deposition. IPF has high mortality and limited therapies, which urgently needs to develop safe and effective therapeutic drugs. Bergenin, a compound derived from a variety of medicinal plants, has demonstrated multiple pharmacological activities including anti-inflammatory and anti-tumor, also acts as a traditional Chinese medicine to treat chronic bronchitis, but its effect on the pulmonary fibrosis is unknown. In this study, we demonstrated that bergenin could attenuate bleomycin (BLM)-induced pulmonary fibrosis in mice. In vitro studies indicated that bergenin inhibited the transforming growth factor-β1 (TGF-β1)-induced fibroblast activation and the extracellular matrix accumulation by inhibiting the TGF-β1/Smad signaling pathway. Further studies showed that bergenin could induce the autophagy formation of myofibroblasts by suppressing the mammalian target of rapamycin signaling and that bergenin could promote the myofibroblast apoptosis. In vivo experiments revealed that bergenin substantially inhibited the myofibroblast activation and the collagen deposition and promoted the autophagy formation. Overall, our results showed that bergenin attenuated the BLM-induced pulmonary fibrosis in mice by suppressing the myofibroblast activation and promoting the autophagy and the apoptosis of myofibroblasts.
Collapse
Affiliation(s)
- Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Yanhua Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Jingjing Liang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Zhun Bi
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Hao Ruan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Yunyao Cui
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Ling Ma
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Yuli Wei
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Bingchen Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Liang Zhang
- Department of Thoracic Surgery, Tian Jin First Central Hospital, Tianjin, China
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| |
Collapse
|
36
|
Mannan A, Garg N, Singh TG, Kang HK. Peroxisome Proliferator-Activated Receptor-Gamma (PPAR-ɣ): Molecular Effects and Its Importance as a Novel Therapeutic Target for Cerebral Ischemic Injury. Neurochem Res 2021; 46:2800-2831. [PMID: 34282491 DOI: 10.1007/s11064-021-03402-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
Cerebral ischemic injury is a leading cause of death and long-term disability throughout the world. Peroxisome proliferator-activated receptor gamma (PPAR-ɣ) is a ligand-activated nuclear transcription factor that is a member of the PPAR family. PPAR-ɣ has been shown in several in vitro and in vivo models to prevent post-ischemic inflammation and neuronal damage by negatively controlling the expression of genes modulated by cerebral ischemic injury, indicating a neuroprotective effect during cerebral ischemic injury. A extensive literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out to understand the nature of the extensive work done on the mechanistic role of Peroxisome proliferator activated receptor gamma and its modulation in Cerebral ischemic injury. PPAR-ɣ can interact with specific DNA response elements to control gene transcription and expression when triggered by its ligand. It regulates lipid metabolism, improves insulin sensitivity, modulates antitumor mechanisms, reduces oxidative stress, and inhibits inflammation. This review article provides insights on the current state of research into the neuroprotective effects of PPAR-ɣ in cerebral ischemic injury, as well as the cellular and molecular mechanisms by which these effects are modulated, such as inhibition of inflammation, reduction of oxidative stress, suppression of pro-apoptotic production, modulation of transcription factors, and restoration of injured tissue through neurogenesis and angiogenesis.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Harmeet Kaur Kang
- Chitkara School of Health Sciences, Chitkara University, Punjab, India
| |
Collapse
|
37
|
Li J, Guo C, Wu J. The Agonists of Peroxisome Proliferator-Activated Receptor-γ for Liver Fibrosis. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2619-2628. [PMID: 34168433 PMCID: PMC8219117 DOI: 10.2147/dddt.s310163] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/07/2021] [Indexed: 12/18/2022]
Abstract
Liver fibrosis is a common link in the transformation of acute and chronic liver diseases to cirrhosis. It is of great clinical significance to study the factors associated with the induction of liver fibrosis and elucidate the method of reversal. Peroxisome proliferator-activated receptors (PPARs) are a class of nuclear transcription factors that can be activated by peroxisome proliferators. PPARs play an important role in fibrosis of various organs, especially the liver, by regulating downstream targeted pathways, such as TGF-β, MAPKs, and NF-κB p65. In recent years, the development and screening of PPAR-γ ligands have become a focus of research. The PPAR-γ ligands include synthetic hypolipidemic and antidiabetic drugs. In addition, microRNAs, lncRNAs, circRNAs and nano new drugs have attracted research interest. In this paper, the research progress of PPAR-γ in the pathogenesis and treatment of liver fibrosis was discussed based on the relevant literature in recent years.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Gastroenterology, Putuo People's Hospital, Tongji University, Shanghai, 200060, People's Republic of China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, People's Republic of China
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People's Hospital, Tongji University, Shanghai, 200060, People's Republic of China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, People's Republic of China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University, Shanghai, 200060, People's Republic of China
| |
Collapse
|
38
|
Li H, Shen X, Tong Y, Ji T, Feng Y, Tang Y, Mai R, Ye J, Que T, Luo X. Aggravation of hepatic ischemia‑reperfusion injury with increased inflammatory cell infiltration is associated with the TGF‑β/Smad3 signaling pathway. Mol Med Rep 2021; 24:580. [PMID: 34132369 PMCID: PMC8223105 DOI: 10.3892/mmr.2021.12219] [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: 11/15/2020] [Accepted: 05/18/2021] [Indexed: 11/06/2022] Open
Abstract
Ischemia‑reperfusion (IR) injury is a major challenge influencing the outcomes of hepatic transplantation. Transforming growth factor‑β (TGF‑β) and its downstream gene, SMAD family member 3 (Smad3), have been implicated in the pathogenesis of chronic hepatic injuries, such as hepatic fibrosis. Thus, the present study aimed to investigate the role of the TGF‑β/Smad3 signaling pathway on hepatic injury induced by IR in vivo. In total, 20 129S2/SvPasCrl wild‑type (WT) mice were randomized into two groups; 10 mice underwent IR injury surgery and 10 mice were sham‑operated. Histopathological changes in liver tissues and serum levels of alanine aminotransferase (ALT) were examined to confirm hepatic injury caused by IR surgery. The expression levels of TGF‑β1, Smad3 and phosphorylated‑Smad3 (p‑Smad3) were detected via western blotting. Furthermore, a total of five Smad3‑/‑ 129S2/SvPasCrl mice (Smad3‑/‑ mice) and 10 Smad3+/+ littermates received IR surgery, while another five Smad3‑/‑ mice and 10 Smad3+/+ littermates received the sham operation. Histopathological changes in liver tissues and serum levels of ALT were then compared between the groups. Furthermore, hepatic apoptosis and inflammatory cell infiltration after IR were evaluated in the liver tissues of Smad3‑/‑ mice and Smad3+/+ mice. The results demonstrated that the expression levels of TGF‑β1, Smad3 and p‑Smad3 were elevated in hepatic tissue from WT mice after IR injury. Aggravated hepatic injury, increased apoptosis and enhanced inflammatory cell infiltration induced by hepatic IR injury were observed in the Smad3‑/‑ mice compared with in Smad3+/+ mice. Collectively, the current findings suggested that activation of the TGF‑β/Smad3 signaling pathway was present alongside the hepatic injury induced by IR. However, the TGF‑β/Smad3 signaling pathway may have an effect on protecting against liver tissue damage caused by IR injury in vivo.
Collapse
Affiliation(s)
- Haixia Li
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoyun Shen
- Key Laboratory of Endoscopic Technology Research, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Yifan Tong
- Key Laboratory of Endoscopic Technology Research, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Tong Ji
- Key Laboratory of Endoscopic Technology Research, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Yan Feng
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yanping Tang
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Rongyun Mai
- Research Department, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jiaxiang Ye
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ting Que
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoling Luo
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
39
|
Álvarez-Mercado AI, Rojano-Alfonso C, Micó-Carnero M, Caballeria-Casals A, Peralta C, Casillas-Ramírez A. New Insights Into the Role of Autophagy in Liver Surgery in the Setting of Metabolic Syndrome and Related Diseases. Front Cell Dev Biol 2021; 9:670273. [PMID: 34141709 PMCID: PMC8204012 DOI: 10.3389/fcell.2021.670273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/23/2021] [Indexed: 01/18/2023] Open
Abstract
Visceral obesity is an important component of metabolic syndrome, a cluster of diseases that also includes diabetes and insulin resistance. A combination of these metabolic disorders damages liver function, which manifests as non-alcoholic fatty liver disease (NAFLD). NAFLD is a common cause of abnormal liver function, and numerous studies have established the enormously deleterious role of hepatic steatosis in ischemia-reperfusion (I/R) injury that inevitably occurs in both liver resection and transplantation. Thus, steatotic livers exhibit a higher frequency of post-surgical complications after hepatectomy, and using liver grafts from donors with NAFLD is associated with an increased risk of post-surgical morbidity and mortality in the recipient. Diabetes, another MetS-related metabolic disorder, also worsens hepatic I/R injury, and similar to NAFLD, diabetes is associated with a poor prognosis after liver surgery. Due to the large increase in the prevalence of MetS, NAFLD, and diabetes, their association is frequent in the population and therefore, in patients requiring liver resection and in potential liver graft donors. This scenario requires advancement in therapies to improve postoperative results in patients suffering from metabolic diseases and undergoing liver surgery; and in this sense, the bases for designing therapeutic strategies are in-depth knowledge about the molecular signaling pathways underlying the effects of MetS-related diseases and I/R injury on liver tissue. A common denominator in all these diseases is autophagy. In fact, in the context of obesity, autophagy is profoundly diminished in hepatocytes and alters mitochondrial functions in the liver. In insulin resistance conditions, there is a suppression of autophagy in the liver, which is associated with the accumulation of lipids, being this is a risk factor for NAFLD. Also, oxidative stress occurring in hepatic I/R injury promotes autophagy. The present review aims to shed some light on the role of autophagy in livers undergoing surgery and also suffering from metabolic diseases, which may lead to the discovery of effective therapeutic targets that could be translated from laboratory to clinical practice, to improve postoperative results of liver surgeries when performed in the presence of one or more metabolic diseases.
Collapse
Affiliation(s)
- Ana Isabel Álvarez-Mercado
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center, Parque Tecnológico Ciencias de la Salud, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Complejo Hospitalario Universitario de Granada, Granada, Spain
| | - Carlos Rojano-Alfonso
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marc Micó-Carnero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Carmen Peralta
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Araní Casillas-Ramírez
- Hospital Regional de Alta Especialidad de Ciudad Victoria “Bicentenario 2010”, Ciudad Victoria, Mexico
- Facultad de Medicina e Ingeniería en Sistemas Computacionales de Matamoros, Universidad Autónoma de Tamaulipas, Matamoros, Mexico
| |
Collapse
|
40
|
PGC-1 α Protects against Hepatic Ischemia Reperfusion Injury by Activating PPAR α and PPAR γ and Regulating ROS Production. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6677955. [PMID: 34104311 PMCID: PMC8159639 DOI: 10.1155/2021/6677955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/05/2021] [Accepted: 04/26/2021] [Indexed: 01/19/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) α and γ have been shown to be protective in hepatic ischemia/reperfusion (I/R) injury. However, the precise role of PPARγ coactivator-1α (PGC-1α), which can coactivate both of these receptors, in hepatic I/R injury, remains largely unknown. This study was designed to test our hypothesis that PGC-1α is protective during hepatic I/R injury in vitro and in vivo. Our results show that endogenous PGC-1α is basally expressed in normal livers and is moderately increased by I/R. Ectopic PGC-1α protects against hepatic I/R and hepatocyte anoxia/reoxygenation (A/R) injuries, whereas knockdown of endogenous PGC-1α aggravates such injuries, as evidenced by assessment of the levels of serum aminotransferases and inflammatory cytokines, necrosis, apoptosis, cell viability, and histological examination. The EMSA assay shows that the activation of PPARα and PPARγ is increased or decreased by the overexpression or knockdown of PGC-1α, respectively, during hepatic I/R and hepatocyte A/R injuries. In addition, the administration of specific antagonists of either PPARα (MK886) or PPARγ (GW9662) can effectively decrease the protective effect of PGC-1α against hepatic I/R and hepatocyte A/R injuries. We also demonstrate an important regulatory role of PGC-1α in reactive oxygen species (ROS) metabolism during hepatic I/R, which is correlated with the induction of ROS-detoxifying enzymes and is also dependent on the activations of PPARα and PPARγ. These data demonstrate that PGC-1α protects against hepatic I/R injury, mainly by regulating the activation of PPARα and PPARγ. Thus, PGC-1α may be a promising therapeutic target for the protection of the liver against I/R injury.
Collapse
|
41
|
Wu L, Li J, Feng J, Ji J, Yu Q, Li Y, Zheng Y, Dai W, Wu J, Guo C. Crosstalk between PPARs and gut microbiota in NAFLD. Biomed Pharmacother 2021; 136:111255. [PMID: 33485064 DOI: 10.1016/j.biopha.2021.111255] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/03/2021] [Accepted: 01/03/2021] [Indexed: 02/08/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most common liver disorder in both China and worldwide. It ranges from simple steatosis and progresses over time to nonalcoholic steatohepatitis (NASH), advanced liver fibrosis, cirrhosis, or hepatocellular carcinoma(HCC). Furthermore, NAFLD and its complications impose a huge health burden to society. The microbiota is widely connected and plays an active role in human physiology and pathology, and it is a hidden 'organ' in determining the state of the host, in terms of homeostasis, or disease. Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptorsuperfamily and can regulate multiple pathways involved in metabolism, and serve as effective targets forthe treatment of many types of metabolic syndromes, including NAFLD. The purpose of this review is to integrate related articles on gut microbiota, PPARs and NAFLD, and present a balanced overview on how the microbiota can possibly influence the development of NAFLD through PPARs.
Collapse
Affiliation(s)
- Liwei Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, 200060, China; Department of Gastroenterology, Shanghai Tenth People'sHospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, 200060, China; Department of Gastroenterology, Shanghai Tenth People'sHospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People'sHospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jie Ji
- Department of Gastroenterology, Shanghai Tenth People'sHospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People'sHospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yan Li
- Department of Gastroenterology, Shanghai Tenth People'sHospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yuanyuan Zheng
- Department of Gastroenterology, Shanghai Tenth People'sHospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, 200060, China; Department of Gastroenterology, Shanghai Tenth People'sHospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, 200060, China.
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People'sHospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
42
|
Zheng M, Li K, Chen T, Liu S, He L. Geniposide protects depression through BTK/JAK2/STAT1 signaling pathway in lipopolysaccharide-induced depressive mice. Brain Res Bull 2021; 170:65-73. [PMID: 33561536 DOI: 10.1016/j.brainresbull.2021.02.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/15/2022]
Abstract
The purpose of this study was to investigate the antidepressant mechanism of GEN (geniposide) on depression mice induced by LPS. The mice were intragastrically treated with GEN (10 mg/kg/d or 40 mg/kg/d) or ibrutinib for continuous 7 days prior to LPS injection. The anxiety- and depression-like behaviors of mice were assessed via behavioral tests (sucrose preference test (SPT), tail suspension test (TST), forced swimming test (FST), and open-field test (OFT)). Microglial BV2 cells were treated with GEN or/and ibrutinib and stimulated with LPS. The productions of pro-inflammatory cytokines IL-6 and TNF-α in hippocampus, serum, and supernatant were detected by ELISA. The correlative proteins BTK, p-BTK, JAK2, p-JAK2, STAT1, p-STAT1, BDNF, TrkB, and p-TrkB were assessed through western blot. As a result, GEN ameliorated the anxiety- and depression-like behaviors of mice in behavioral tests. GEN treatment also regulated microglia polarization towards anti-inflammatory phenotype M2 and inhibited the production of pro-inflammatory cytokines IL-6 and TNF-α. In addition, with the application of ibrutinib, the selective inhibitor of BTK, it was proclaimed that the administration of GEN restrained the activation of JAK2/STAT1 pathway via attenuating the hyperphosphorylation of BTK both in mice and BV2 cells. Furthermore, it was also found that GEN activated BDNF/TrkB neuroprotective signaling pathway through the reduction of BTK phosphorylation. From the overall results, we suggested that GEN exerted a beneficial effect on LPS-induced depression in mice possibly through the modulation of BTK/JAK2/STAT1 signaling.
Collapse
Affiliation(s)
- Menglin Zheng
- Department of Pharmacology, China Pharmaceutical University, 639, Longmian Avenue, Nanjing, 211198, China
| | - Ke Li
- Department of Pharmacology, China Pharmaceutical University, 639, Longmian Avenue, Nanjing, 211198, China
| | - Tong Chen
- Department of Pharmacology, China Pharmaceutical University, 639, Longmian Avenue, Nanjing, 211198, China
| | - Shengnan Liu
- Department of Pharmacology, China Pharmaceutical University, 639, Longmian Avenue, Nanjing, 211198, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, 639, Longmian Avenue, Nanjing, 211198, China.
| |
Collapse
|
43
|
Zhang J, Cheng P, Dai W, Ji J, Wu L, Feng J, Wu J, Yu Q, Li J, Guo C. Fenofibrate Ameliorates Hepatic Ischemia/Reperfusion Injury in Mice: Involvements of Apoptosis, Autophagy, and PPAR- α Activation. PPAR Res 2021; 2021:6658944. [PMID: 33603777 PMCID: PMC7870311 DOI: 10.1155/2021/6658944] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/22/2020] [Accepted: 01/16/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatic ischemia and reperfusion injury is characterized by hepatocyte apoptosis, impaired autophagy, and oxidative stress. Fenofibrate, a commonly used antilipidemic drug, has been verified to exert hepatic protective effects in other cells and animal models. The purpose of this study was to identify the function of fenofibrate on mouse hepatic IR injury and discuss the possible mechanisms. A segmental (70%) hepatic warm ischemia model was established in Balb/c mice. Serum and liver tissue samples were collected for detecting pathological changes at 2, 8, and 24 h after reperfusion, while fenofibrate (50 mg/kg, 100 mg/kg) was injected intraperitoneally 1 hour prior to surgery. Compared to the IR group, pretreatment of FF could reduce the inflammatory response and inhibit apoptosis and autophagy. Furthermore, fenofibrate can activate PPAR-α, which is associated with the phosphorylation of AMPK.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Ping Cheng
- Department of Gerontology, Shanghai Minhang District Central Hospital, Shanghai 201100, China
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China
| | - Jie Ji
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai 200060, China
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
44
|
Yu Q, Cheng P, Wu J, Guo C. PPARγ/NF-κB and TGF-β1/Smad pathway are involved in the anti-fibrotic effects of levo-tetrahydropalmatine on liver fibrosis. J Cell Mol Med 2021; 25:1645-1660. [PMID: 33438347 PMCID: PMC7875896 DOI: 10.1111/jcmm.16267] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/10/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022] Open
Abstract
Liver fibrosis is a necessary stage in the development of chronic liver diseases to liver cirrhosis. This study aims to investigate the anti‐fibrotic effects of levo‐tetrahydropalmatine (L‐THP) on hepatic fibrosis in mice and cell models and its underlying mechanisms. Two mouse hepatic fibrosis models were generated in male C57 mice by intraperitoneal injection of carbon tetrachloride (CCl4) for 2 months and bile duct ligation (BDL) for 14 days. Levo‐tetrahydropalmatine was administered orally at doses of 20 and 40 mg/kg. An activated LX2 cell model induced by TGF‐β1 was also generated. The results showed that levo‐tetrahydropalmatine alleviated liver fibrosis by inhibiting the formation of extracellular matrix (ECM) and regulating the balance between TIMP1 and MMP2 in the two mice liver fibrosis models and cell model. Levo‐tetrahydropalmatine inhibited activation and autophagy of hepatic stellate cells (HSCs) by modulating PPARγ/NF‐κB and TGF‐β1/Smad pathway in vivo and in vitro. In conclusion, levo‐tetrahydropalmatine attenuated liver fibrosis by inhibiting ECM deposition and HSCs autophagy via modulation of PPARγ/NF‐κB and TGF‐β1/Smad pathway.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ping Cheng
- Department of Gerontology, Shanghai Minhang District Central Hospital, Shanghai, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
45
|
Bergenin Attenuates Hepatic Fibrosis by Regulating Autophagy Mediated by the PPAR- γ/TGF- β Pathway. PPAR Res 2020; 2020:6694214. [PMID: 33488687 PMCID: PMC7790590 DOI: 10.1155/2020/6694214] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/06/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis is a pathological process involving diffuse extracellular matrix (ECM) deposition in the liver. It is typical of many chronic liver diseases, including cirrhosis, and effective drugs are needed. In this study, we explored the protective effect of bergenin on liver fibrosis induced by carbon tetrachloride and bile duct ligation. A variety of molecular biological methods (qRT-PCR, western blotting, and immunohistochemistry) were employed to confirm the increased degree of hepatocyte injury and ECM formation in the disease model, consistent with autophagy and activation of the TGF-β pathway. Bergenin activated PPAR-γ and inhibited TGF-β and autophagy and decreased liver fibrosis by inhibiting hepatocyte necrosis and ECM formation in a dose-dependent manner. The results suggest that bergenin may be a promising drug candidate for the treatment of liver fibrosis.
Collapse
|
46
|
Hou Y, Ali I, Li Z, Sulaiman A, Aziz S, Chen L, Hussain H, Cui L, Wang D, Zheng X. Separation of constituents from Bergenia stracheyi (Hook. F. & Thoms.) Engl. by high-speed countercurrent chromatography with elution mode and its antidiabetic and antioxidant in vitro evaluation. J Sep Sci 2020; 44:767-776. [PMID: 33314692 DOI: 10.1002/jssc.202000999] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/20/2020] [Accepted: 11/24/2020] [Indexed: 01/18/2023]
Abstract
Diabetes, a metabolic disorder, is caused by a high blood sugar level. Diabetes is an increasing health issue and search for potent antidiabetic agents is desirable. Owing to its ethnomedicinal value, the Himalayan perennial herb Bergenia stracheyi (Hook. f. & Thoms.) Engl. (Saxifragaceae Juss) is used to treat diabetes. Herein, an efficient high-speed countercurrent chromatography with elution mode is reported for separation of active compounds from B. stracheyi. In current investigation, six main compounds including β-arbutin (1), bergenin (2), 6-O-galloylarbutin (3), gallic acid (4), 11-O-galloylbergenin (5), and (-)-epicatechin 3-O-gallate (6) with above 95% purity were efficiently separated in a single run using biphasic tert-butyl methyl ether/n-butanol/methanol/water (1:3:1:5, v/v/v/v) solvent system. The structures of these compounds were characterized using spectral techniques and compared with the literature. Antidiabetic and antioxidant activities evaluation of the study samples showed that β-arbutin (1) and 6-O-galloylarbutin (3) have a significant protective effect, especially at high dose against hydrogen peroxide induced oxidative injury. Our results might help further in-depth phytochemical and biological evaluation studies in search of potent antidiabetic compounds from B. stracheyi.
Collapse
Affiliation(s)
- Yue Hou
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Iftikhar Ali
- School of Pharmaceutical Sciences and Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, P. R. China.,Department of Chemistry, Karakoram International University, Gilgit, Pakistan
| | - Zhao Li
- School of Pharmaceutical Sciences and Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, P. R. China
| | - Atiqa Sulaiman
- Department of Chemistry, Karakoram International University, Gilgit, Pakistan
| | - Shahid Aziz
- School of Pharmaceutical Sciences and Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, P. R. China.,Department of Chemistry, Mirpur University of Science and Technology (MUST), Mirpur, Pakistan
| | - Long Chen
- School of Pharmaceutical Sciences and Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, P. R. China
| | - Hidayat Hussain
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle, Germany
| | - Li Cui
- School of Pharmaceutical Sciences and Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, P. R. China
| | - Daijie Wang
- School of Pharmaceutical Sciences and Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, P. R. China
| | - Xin Zheng
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| |
Collapse
|
47
|
Rajput SA, Mirza MR, Choudhary MI. Bergenin protects pancreatic beta cells against cytokine-induced apoptosis in INS-1E cells. PLoS One 2020; 15:e0241349. [PMID: 33347462 PMCID: PMC7751853 DOI: 10.1371/journal.pone.0241349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022] Open
Abstract
Beta cell apoptosis induced by proinflammatory cytokines is one of the hallmarks of diabetes. Small molecules which can inhibit the cytokine-induced apoptosis could lead to new drug candidates that can be used in combination with existing therapeutic interventions against diabetes. The current study evaluated several effects of bergenin, an isocoumarin derivative, in beta cells in the presence of cytokines. These included (i) increase in beta cell viability (by measuring cellular ATP levels) (ii) suppression of beta cell apoptosis (by measuring caspase activity), (iii) improvement in beta cell function (by measuring glucose-stimulated insulin secretion), and (iv) improvement of beta cells mitochondrial physiological functions. The experiments were carried out using rat beta INS-1E cell line in the presence or absence of bergenin and a cocktail of proinflammatory cytokines (interleukin-1beta, tumor necrosis factor-alpha, and interferon- gamma) for 48 hr. Bergenin significantly inhibited beta cell apoptosis, as inferred from the reduction in the caspase-3 activity (IC50 = 7.29 ± 2.45 μM), and concurrently increased cellular ATP Levels (EC50 = 1.97 ± 0.47 μM). Bergenin also significantly enhanced insulin secretion (EC50 = 6.73 ± 2.15 μM) in INS-1E cells, presumably because of the decreased nitric oxide production (IC50 = 6.82 ± 2.83 μM). Bergenin restored mitochondrial membrane potential (EC50 = 2.27 ± 0.83 μM), decreased ROS production (IC50 = 14.63 ± 3.18 μM), and improved mitochondrial dehydrogenase activity (EC50 = 1.39 ± 0.62 μM). This study shows for the first time that bergenin protected beta cells from cytokine-induced apoptosis and restored insulin secretory function by virtue of its anti-inflammatory, antioxidant and anti-apoptotic properties. To sum up, the above mentioned data highlight bergenin as a promising anti-apoptotic agent in the context of diabetes.
Collapse
Affiliation(s)
- Sajid Ali Rajput
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center of Chemical and Biological Sciences, University of Karachi, Karachi, Sindh, Pakistan
| | - Munazza Raza Mirza
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center of Chemical and Biological Sciences, University of Karachi, Karachi, Sindh, Pakistan
| | - M. Iqbal Choudhary
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center of Chemical and Biological Sciences, University of Karachi, Karachi, Sindh, Pakistan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Sindh, Pakistan
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- * E-mail:
| |
Collapse
|
48
|
Yu Q, Wu L, Ji J, Feng J, Dai W, Li J, Wu J, Guo C. Gut Microbiota, Peroxisome Proliferator-Activated Receptors, and Hepatocellular Carcinoma. J Hepatocell Carcinoma 2020; 7:271-288. [PMID: 33150145 PMCID: PMC7605923 DOI: 10.2147/jhc.s277870] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/10/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world. HCC incidence rate is sixth and mortality is fourth worldwide. However, HCC pathogenesis and molecular mechanisms remain unclear. The incidence of HCC is associated with genetic, environmental, and metabolic factors. The role of gut microbiota in the pathogenesis of HCC has attracted researchers' attention because of anatomical and functional interactions between liver and intestine. Studies have demonstrated the involvement of gut microbiota in the development of HCC and chronic liver diseases, such as alcoholic liver disease (ALD), nonalcoholic fatty liver disease (NAFLD), and liver cirrhosis. Peroxisome proliferator-activated receptors (PPARs) are a group of receptors with diverse biological functions. Natural and synthetic PPAR agonists show potential for treatment of NAFLD, liver fibrosis, and HCC. Recent studies have demonstrated that PPARs take part in gut microbiota inhabitation and adaptation. This manuscript reviews the role of gut microbiota in the development of HCC and precancerous diseases, the role of PPARs in modulation of gut microbiota and HCC, and potential of gut microbiota for HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University School of Medicine, Shanghai200060, People’s Republic of China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai200072, People’s Republic of China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai200072, People’s Republic of China
| | - Jie Ji
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai200072, People’s Republic of China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai200072, People’s Republic of China
| | - Weiqi Dai
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University School of Medicine, Shanghai200060, People’s Republic of China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai200072, People’s Republic of China
- Shanghai Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai200336, People’s Republic of China
| | - Jingjing Li
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University School of Medicine, Shanghai200060, People’s Republic of China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai200072, People’s Republic of China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University School of Medicine, Shanghai200060, People’s Republic of China
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People’s Hospital, Tongji University School of Medicine, Shanghai200060, People’s Republic of China
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai200072, People’s Republic of China
| |
Collapse
|
49
|
Garg S, Khan SI, Malhotra RK, Sharma MK, Kumar M, Kaur P, Nag TC, RumaRay, Bhatia J, Arya DS. The molecular mechanism involved in cardioprotection by the dietary flavonoid fisetin as an agonist of PPAR-γ in a murine model of myocardial infarction. Arch Biochem Biophys 2020; 694:108572. [PMID: 32926843 DOI: 10.1016/j.abb.2020.108572] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 02/07/2023]
Abstract
The methodology exploring the cardioprotective potential of the flavonoid Fisetin through its ability to modulate PPAR-γ was unraveled in the present study. Computational modelling through molecular docking based binding study of interactions between Fiestin and PPAR-γ revealed the potential role of Fisetin as an agonist of PPAR-γ. A murine model of cardiac ischemia-reperfusion injury was used to explore this further. Male Wistar Rats were randomly assigned to five groups. Fisetin (20 mg/kg; p. o) was administered for 28 days. Ischemia was induced for 45 min on the 29th day followed by 60 min of reperfusion. Fisetin pretreatment upregulated the expression of PPAR-γ in heart tissue significantly Cardioprotection was assessed by measurement of hemodynamic parameters, infarct size, ELISA for oxidative stress, immunohistochemistry and TUNEL assay for apoptosis, and western blot analysis for MAPK proteins and inflammation. PPAR-γ activation by fisetin led to significantly reduced infarct size, suppression of oxidative stress, reduction of cardiac injury markers, alleviation of inflammation, and inhibition of apoptosis The MAPK-based molecular mechanism showed a rise in a key prosurvival kinase, ERK1/ERK2 and suppression of JNK and p38 proteins. The aforementioned beneficial findings of fisetin were reversed on the administration of a specific antagonist of PPAR-γ. In conclusion, through our experiments, we have proved that fisetin protects the heart against ischemia-reperfusion injury and the evident cardioprotection is PPAR-γ dependant. In conclusion, our study has revealed a prime mechanism involved in the cardioprotective effects of fisetin. Hence, Fisetin may be evaluated in further clinical studies as a cardioprotective agent in patients undergoing reperfusion interventions.
Collapse
Affiliation(s)
- Shanky Garg
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Sana Irfan Khan
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Rajiv Kumar Malhotra
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Manish Kumar Sharma
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Manoj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - RumaRay
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Jagriti Bhatia
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India
| | - Dharamvir Singh Arya
- Department of Pharmacology, Cardiovascular Research Laboratory, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
50
|
Villarreal CF, Santos DS, Lauria PSS, Gama KB, Espírito-Santo RF, Juiz PJL, Alves CQ, David JM, Soares MBP. Bergenin Reduces Experimental Painful Diabetic Neuropathy by Restoring Redox and Immune Homeostasis in the Nervous System. Int J Mol Sci 2020; 21:ijms21144850. [PMID: 32659952 PMCID: PMC7420298 DOI: 10.3390/ijms21144850] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetic neuropathy is a frequent complication of diabetes. Symptoms include neuropathic pain and sensory alterations—no effective treatments are currently available. This work characterized the therapeutic effect of bergenin in a mouse (C57/BL6) model of streptozotocin-induced painful diabetic neuropathy. Nociceptive thresholds were assessed by the von Frey test. Cytokines, antioxidant genes, and oxidative stress markers were measured in nervous tissues by ELISA, RT-qPCR, and biochemical analyses. Single (3.125–25 mg/kg) or multiple (25 mg/kg; twice a day for 14 days) treatments with bergenin reduced the behavioral signs of diabetic neuropathy in mice. Bergenin reduced both nitric oxide (NO) production in vitro and malondialdehyde (MDA)/nitrite amounts in vivo. These antioxidant properties can be attributed to the modulation of gene expression by the downregulation of inducible nitric oxide synthase (iNOS) and upregulation of glutathione peroxidase and Nrf2 in the nervous system. Bergenin also modulated the pro- and anti-inflammatory cytokines production in neuropathic mice. The long-lasting antinociceptive effect induced by bergenin in neuropathic mice, was associated with a shift of the cytokine balance toward anti-inflammatory predominance and upregulation of antioxidant pathways, favoring the reestablishment of redox and immune homeostasis in the nervous system. These results point to the therapeutic potential of bergenin in the treatment of painful diabetic neuropathy.
Collapse
Affiliation(s)
- Cristiane F. Villarreal
- Faculdade de Farmácia, Universidade Federal da Bahia, CEP 40.170-115 Salvador, Brazil; (D.S.S.); (P.S.S.L.); (R.F.E.-S.)
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, CEP 40.296-710 Salvador, Brazil; (K.B.G.); (M.B.P.S.)
- Correspondence: ; Tel.: +55-(71)3283-6933
| | - Dourivaldo S. Santos
- Faculdade de Farmácia, Universidade Federal da Bahia, CEP 40.170-115 Salvador, Brazil; (D.S.S.); (P.S.S.L.); (R.F.E.-S.)
| | - Pedro S. S. Lauria
- Faculdade de Farmácia, Universidade Federal da Bahia, CEP 40.170-115 Salvador, Brazil; (D.S.S.); (P.S.S.L.); (R.F.E.-S.)
| | - Kelly B. Gama
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, CEP 40.296-710 Salvador, Brazil; (K.B.G.); (M.B.P.S.)
| | - Renan F. Espírito-Santo
- Faculdade de Farmácia, Universidade Federal da Bahia, CEP 40.170-115 Salvador, Brazil; (D.S.S.); (P.S.S.L.); (R.F.E.-S.)
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, CEP 40.296-710 Salvador, Brazil; (K.B.G.); (M.B.P.S.)
| | - Paulo J. L. Juiz
- Universidade Federal do Recôncavo da Bahia, CEP 44.042-280 Feira de Santana, Brazil;
| | - Clayton Q. Alves
- Departamento de Ciências Exatas, Universidade Estadual de Feira de Santana, CEP 44.036-336 Feira de Santana, Brazil;
| | - Jorge M. David
- Instituto de Química, Universidade Federal da Bahia, CEP 40.170-280 Salvador, Brazil;
| | - Milena B. P. Soares
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, CEP 40.296-710 Salvador, Brazil; (K.B.G.); (M.B.P.S.)
| |
Collapse
|