1
|
La X, Zhang Z, Liang J, Li H, Pang Y, He X, Kang Y, Wu C, Li Z. Isolation and purification of flavonoids from quinoa whole grain and its inhibitory effect on lipid accumulation in nonalcoholic fatty liver disease by inhibiting the expression of CD36 and FASN. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:1330-1342. [PMID: 39305086 DOI: 10.1002/jsfa.13923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD), a chronic metabolic disorder marked by excessive lipid deposition, represents a considerable health burden with no established efficacious treatment strategy. Quinoa (Chenopodium quinoa Willd.), valued for its health benefits, is replete with flavonoid bioactives. The aims of this work are to isolate and purify flavonoids from quinoa whole grain that can intervene in NAFLD and to elucidate some of the underlying mechanisms. RESULTS Chenopodium quinoa Willd. flavonoids (CQWF) were obtained successfully through an optimized ultrasonic extraction methodology, followed by isolation and purification utilizing macroporous resin D101. The study then explored the therapeutic potential of CQWF and its eluted fractions in models emulating NAFLD conditions: an in vitro fatty liver cell model induced by oleic acid (OA) and palmitic acid (PA) in the HepG2 and BEL-7402 cell lines, and an in vivo high-fat diet (HFD)-induced NAFLD model in C57BL/6N mice. The findings revealed a comprehensive mitigating effect of CQWF30 on NAFLD, manifesting in reduced intracellular lipid accumulation in steatotic hepatocytes and a concerted downregulation of key lipid metabolism genes, CD36 and FASN. Administration of CQWF30 reduced triglyceride (TG) levels in both the cellular model and the livers of HFD-fed mice. It also reduced serum concentrations of TG, total cholesterol (T-CHO), low-density lipoprotein cholesterol (LDL-C), aspartate aminotransferase (AST), and alanine aminotransferase (ALT), while increasing high-density lipoprotein cholesterol (HDL-C) in the mice. CONCLUSION These results highlighted the promising therapeutic capacity of CQWF, particularly CQWF30. This research advances the exploration and utilization of flavonoids derived from quinoa whole grain, providing innovative dietary intervention strategies for NAFLD. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaoqin La
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan, China
| | - Zhaoyan Zhang
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Jingyi Liang
- Institute of Biotechnology, Shanxi University, Taiyuan, China
| | - Hanqing Li
- School of Life Science, Shanxi University, Taiyuan, China
| | - Yan Pang
- School of Life Science, Shanxi University, Taiyuan, China
| | - Xiaoting He
- School of Life Science, Shanxi University, Taiyuan, China
| | - Yurui Kang
- School of Life Science, Shanxi University, Taiyuan, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Zhuoyu Li
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan, China
- Institute of Biotechnology, Shanxi University, Taiyuan, China
| |
Collapse
|
2
|
Katsarou A, Tsioulos G, Kassi E, Chatzigeorgiou A. Current and experimental pharmacotherapy for the management of non-alcoholic fatty liver disease. Hormones (Athens) 2024; 23:621-636. [PMID: 39112786 DOI: 10.1007/s42000-024-00588-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/17/2024] [Indexed: 10/29/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease, with its incidence increasing in parallel with the global prevalence of obesity and type 2 diabetes mellitus. Despite our steadily increasing knowledge of its pathogenesis, there is as yet no available pharmacotherapy specifically tailored for NAFLD. To define the appropriate management, it is important to clarify the context in which the disease appears. In the case of concurrent metabolic comorbidities, NAFLD patients are treated by targeting these comorbidities, such as diabetes and obesity. Thus, GLP-1 analogs, PPAR, and SGLT2 inhibitors have recently become central to the treatment of NAFLD. In parallel, randomized trials are being conducted to explore new agents targeting known pathways involved in NAFLD progression. However, there is an imperative need to intensify the effort to design new, safe drugs with biopsy-proven efficacy. Of note, the main target of the pharmacotherapy should be directed to the regression of fibrotic NASH, as this histologic stage has been correlated with increased overall as well as liver-related morbidity and mortality. Herein we discuss the drugs currently at the forefront of NAFLD treatment.
Collapse
Affiliation(s)
- Angeliki Katsarou
- 251 Hellenic Airforce General Hospital, 1 P.Kanellopoulou Str, Athens, 11525, Greece.
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str, Athens, 11527, Greece.
| | - Georgios Tsioulos
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 1 Rimini Str, Athens, 12462, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 115 27, Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str, Athens, 11527, Greece
| |
Collapse
|
3
|
Quan J, Xie D, Li Z, Yu X, Liang Z, Chen Y, Wu L, Huang D, Lin L, Fan L. Luteolin alleviates airway remodeling in asthma by inhibiting the epithelial-mesenchymal transition via β-catenin regulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156090. [PMID: 39393303 DOI: 10.1016/j.phymed.2024.156090] [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: 04/19/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND Asthma is a prevalent long-term inflammatory condition that causes airway inflammation and remodeling. Increasing evidence indicates that epithelial-mesenchymal transition (EMT) holds a prominent implication in airway reconstruction in patients with asthma. Flavonoids obtained from Chinese Materia Medica (CMM), such as Luteolin (Lut), exhibit various beneficial effects in various asthma models. Lut has been shown to mitigate various asthma symptoms, including airway inflammation, hyperresponsiveness, bronchoconstriction, excessive mucus production, pulmonary autophagy, and neutrophilic asthma. However, whether flavonoids can suppress EMT-associated airway remodeling in asthma and the fundamental mechanisms involved remain unclear, with no studies specifically addressing Lut in this context. PURPOSE To evaluate the inhibition of airway remodeling in asthma by Lut and its potential mechanisms, while examining the significance of β-catenin in this process through cellular and animal studies. METHODS A BEAS-2B cell model stimulated by lipopolysaccharide (LPS) was established in vitro. Wound closure and Transwell assays were utilized to assess the cellular migratory ability. EMT- and fibrosis-related markers in LPS-stimulated cells were evaluated using RT-qPCR and western blotting. The status of the β-catenin/E-cadherin and β-catenin destruction complexes was evaluated using western blotting, immunofluorescence (IF) staining, and co-immunoprecipitation (Co-IP) analysis. The regulatory function of Lut in β-catenin-dependent EMT was further validated by β-catenin overexpression with adenovirus transduction and siRNA-mediated knockdown of β-catenin. Moreover, the counts of different types of bronchoalveolar lavage fluid (BALF) inflammatory cells from mice with asthma induced by ovalbumin (OVA) were evaluated in vivo using Congo red staining. Hematoxylin and eosin (H&E), Masson's trichrome, and periodic acid-Schiff (PAS) staining were used to evaluate collagen deposition, mucus production, and inflammation in murine lung tissues. Western blotting and immunohistochemistry (IHC) assays were used to assess EMT- and fibrosis-related markers in the lung tissues in vivo. RESULT Six naturally derived flavonoids, including Lut, attenuated cell migration and prevented EMT in LPS-treated BEAS-2B cells. Moreover, Lut suppressed TGF-β1, MMP-9, fibronectin (FN), and α-smooth muscle actin (α-SMA) levels in LPS-stimulated BEAS-2B cells. Additionally, Lut downregulated the levels of β-catenin by modulating the β-catenin/E-cadherin and β-catenin destruction complexes, highlighting the pivotal role of β-catenin in EMT inhibition by Lut in LPS-stimulated BEAS-2B cells. Furthermore, Lut suppressed airway inflammation and attenuated EMT-associated airway remodeling through β-catenin blockade in OVA-induced asthmatic mice. The bronchial wall thickness notably reduced from 37.24 ± 4.00 μm in the asthmatic model group to 30.06 ± 4.40 μm in the Lut low-dose group and 24.69 ± 2.87 μm in the Lut high-dose group. CONCLUSION According to our current understanding, this research is the first to reveal that Lut diminishes airway remodeling in asthma by inhibiting EMT via β-catenin regulation, thereby filling a research gap concerning Lut and flavonoids. These results provide a theoretical basis for treating asthma with anti-asthmatic CMM, as well as a candidate and complementary therapeutic approach to treat asthma.
Collapse
Affiliation(s)
- Jingyu Quan
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Dan Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Zihong Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Xuhua Yu
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Ziyao Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Yuanbin Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Lei Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Donghui Huang
- Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, China.
| | - Lin Lin
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China.
| | - Long Fan
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Department of Respiratory Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China.
| |
Collapse
|
4
|
Bonfiglio C, Tatoli R, Donghia R, Guido D, Giannelli G. Exploratory Role of Flavonoids on Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) in a South Italian Cohort. Antioxidants (Basel) 2024; 13:1286. [PMID: 39594428 PMCID: PMC11591465 DOI: 10.3390/antiox13111286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most recent definition for steatotic liver disease associated with metabolic syndrome. The results of recent metabolic and observational studies suggest a potential beneficial effect of food-derived flavonoids in some chronic diseases, including MASLD. The study aims to evaluate the protective role of diet flavonoids in subjects with and without MASLD belonging to a cohort living in the South of Italy. METHODS The study cohort comprised 1297 participants assessed in the NUTRIHEP cohort (2015-2018), divided into two groups, based on presence or absence of MASLD. RESULTS The results indicated statistically significant flavonoid consumption, showing a protective role against MASLD, at an optimal concentration of 165 mg/day, with an OR value of 0.63, (p = 0.001, 95% C.I.: 0.47; 0.83 t). The OR remained almost unchanged when the intake increased from 165 mg per day to 185 mg per day. CONCLUSIONS In conclusion, our study results show a protective role of flavonoids against MASLD. Consuming only 165 mg of flavonoids daily can activate this protective function, reducing the risk of MASLD.
Collapse
Affiliation(s)
- Caterina Bonfiglio
- Unit of Data Science, National Institute of Gastroenterology—IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.D.); (D.G.)
| | - Rossella Tatoli
- Unit of Data Science, National Institute of Gastroenterology—IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.D.); (D.G.)
| | - Rossella Donghia
- Unit of Data Science, National Institute of Gastroenterology—IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.D.); (D.G.)
| | - Davide Guido
- Unit of Data Science, National Institute of Gastroenterology—IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.D.); (D.G.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology—IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| |
Collapse
|
5
|
Sokal-Dembowska A, Jarmakiewicz-Czaja S, Filip R. Flavonoids and Their Role in Preventing the Development and Progression of MAFLD by Modifying the Microbiota. Int J Mol Sci 2024; 25:11187. [PMID: 39456969 PMCID: PMC11508831 DOI: 10.3390/ijms252011187] [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: 09/25/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
With the increasing prevalence and serious health consequences of metabolic-associated fatty liver disease (MAFLD), early diagnosis and intervention are key to effective treatment. Recent studies highlight the important role of dietary factors, including the use of flavonoids, in improving liver health. These compounds possess anti-inflammatory, antioxidant, and liver-protective properties. Flavonoids have been shown to affect the gut microbiota, which plays a key role in liver function and disease progression. Therefore, their role in preventing the development and progression of MAFLD through modulation of the microbiome seems to be of interest. This narrative review aims to consolidate the current evidence on the effects of selected flavonoids on MAFLD progression, their potential mechanisms of action, and the implications for the development of personalized dietary interventions for the management of liver disease.
Collapse
Affiliation(s)
- Aneta Sokal-Dembowska
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland.; (S.J.-C.)
| | - Sara Jarmakiewicz-Czaja
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland.; (S.J.-C.)
| | - Rafał Filip
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
- Department of Gastroenterology with IBD Unit, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| |
Collapse
|
6
|
Abdelmoneim D, Eldomany EB, El-Adl M, Farghali A, El-Sayed G, El-Sherbini ES. Possible protective effect of natural flavanone naringenin-reduced graphene oxide nanosheets on nonalcoholic fatty liver disease. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03495-9. [PMID: 39414698 DOI: 10.1007/s00210-024-03495-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
Utilizing naringenin as a safe, natural compound for reducing graphene oxide and to determine whether Nar-RGO more effectively mitigates the harmful effects of HFFD-induced NAFLD compared to crude naringenin. Using a straightforward experimental setup, we utilize the bioactive flavonoid naringenin (NAR) as the reducing agent to synthesize naringenin-reduced graphene oxide nanosheets (Nar-RGO). Naringenin loading on graphene oxide was validated using electroscopic methods (SEM and TEM) and zeta potential measurements. Utilization of reduced graphene oxide for naringenin encapsulation resulted in a significant improvement in hepatic steatosis, insulin resistance, oxidative stress, and signs of inflammation in HFFD-induced NAFLD compared to crude naringenin. This study demonstrates that Nar-RGO exhibits significantly greater efficacy compared to free naringenin. Therefore, it can be used as a promising medicine in counteracting high-fat-fructose diet (HFFD)-induced NAFLD.
Collapse
Affiliation(s)
- Doaa Abdelmoneim
- Biochemistry and Chemistry of Nutrition Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Ehab B Eldomany
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Mohamed El-Adl
- Biochemistry and Chemistry of Nutrition Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Ahmed Farghali
- Material Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Gehad El-Sayed
- Biochemistry and Chemistry of Nutrition Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - El Said El-Sherbini
- Biochemistry and Chemistry of Nutrition Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
7
|
Liang M, Dong S, Guo Y, Zhang Y, Xiao X, Ma J, Jiang X, Yu W. Exploration of the potential mechanism of aqueous extract of Artemisia capillaris for the treatment of non-alcoholic fatty liver disease based on network pharmacology and experimental verification. J Pharm Pharmacol 2024; 76:1328-1339. [PMID: 39186724 DOI: 10.1093/jpp/rgae061] [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: 01/10/2024] [Accepted: 08/06/2024] [Indexed: 08/28/2024]
Abstract
OBJECTIVES Non-alcoholic fatty liver disease (NAFLD) is a nutritional and metabolic disease with a high prevalence today. Artemisia capillaris has anti-inflammatory, antioxidant, and other effects. However, the mechanism of A. capillaris in treating NAFLD is still poorly understood. METHODS This study explored the mechanism of A. capillaris in the treatment of NAFLD through network pharmacology and molecular docking, and verified the results through in vivo experiments using a high-fat diet-induced mouse model and in vitro experiments using an oleic acid-induced HepG2 cell model. KEY FINDINGS Aqueous extract of A. capillaris (AEAC) can reduce blood lipids, reduce liver lipid accumulation and liver inflammation in NAFLD mice, and improve NAFLD. Network pharmacology analysis revealed that 51 drug ingredients in A. capillaris correspond to 370 targets that act on NAFLD. GEO data mining obtained 93 liver differentially expressed genes related to NAFLD. In the UHPLC-MS detection results, 36 components were characterized and molecular docked with JNK. Verified in vitro and in vivo, the results show that JNK and the phosphorylation levels of IL-6, IL-1β, c-Jun, c-Fos, and CCL2 are key targets and pathways. CONCLUSIONS This study confirmed that AEAC reduces lipid accumulation and inflammation in the liver of NAFLD mice by inhibiting the JNK/AP-1 pathway.
Collapse
Affiliation(s)
- Meng Liang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Siyu Dong
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yi Guo
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yuyi Zhang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xiao Xiao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jun Ma
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Heilongjiang Provincial Key Laboratory for Prevention and Control of Common Animal Diseases, Northeast Agricultural University, Harbin, 150030, China
| | - Xiaowen Jiang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Wenhui Yu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Heilongjiang Provincial Key Laboratory for Prevention and Control of Common Animal Diseases, Northeast Agricultural University, Harbin, 150030, China
| |
Collapse
|
8
|
La X, Zhang Z, Dong C, Li H, He X, Kang Y, Wu C, Li Z. Isorhamnetin in Quinoa Whole-Grain Flavonoids Intervenes in Non-Alcoholic Fatty Liver Disease by Modulating Bile Acid Metabolism through Regulation of FXR Expression. Foods 2024; 13:3076. [PMID: 39410111 PMCID: PMC11475887 DOI: 10.3390/foods13193076] [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: 08/29/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a severe hepatic health threat with no effective treatment. Based on the results that Chenopodium quinoa Willd. flavonoids eluted with 30% ethanol (CQWF30) can effectively alleviate NAFLD, this study employed ultrahigh-performance liquid chromatography-electrospray ionization-tandem mass spectrometry (UPLC-ESI-MS/MS) to analyze the components of CQWF30., and screened for flavonoids with potential NAFLD-mitigating effects through network pharmacology. In vitro models using HepG2 and BEL-7402 cell lines induced with free fatty acid (FFA) showed that isorhamnetin administration reduced intracellular lipid deposition and reversed elevated triglyceride (TG) and total cholesterol (T-CHO) levels. In vivo experiments in high-fat diet (HFD) mice demonstrated that isorhamnetin significantly lowered serum and liver fat content, mitigated liver damage, and modulated bile acid metabolism by upregulating FXR and BSEP and downregulating SLCO1B3. Consequently, isorhamnetin shows promise as a treatment for NAFLD due to its lipid-lowering and hepatoprotective activities.
Collapse
Affiliation(s)
- Xiaoqin La
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (Z.Z.); (C.W.)
| | - Zhaoyan Zhang
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (Z.Z.); (C.W.)
| | - Cunli Dong
- School of Life Science, Shanxi University, Taiyuan 030006, China; (C.D.); (H.L.); (X.H.); (Y.K.)
| | - Hanqing Li
- School of Life Science, Shanxi University, Taiyuan 030006, China; (C.D.); (H.L.); (X.H.); (Y.K.)
| | - Xiaoting He
- School of Life Science, Shanxi University, Taiyuan 030006, China; (C.D.); (H.L.); (X.H.); (Y.K.)
| | - Yurui Kang
- School of Life Science, Shanxi University, Taiyuan 030006, China; (C.D.); (H.L.); (X.H.); (Y.K.)
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; (Z.Z.); (C.W.)
- Shanxi Provincial Key Laboratory of Medical Molecular Cell Biology, Shanxi University, Taiyuan 030006, China
| | - Zhuoyu Li
- Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
9
|
Wen J, Tian YE, Skampardoni I, Yang Z, Cui Y, Anagnostakis F, Mamourian E, Zhao B, Toga AW, Zalesky A, Davatzikos C. The genetic architecture of biological age in nine human organ systems. NATURE AGING 2024; 4:1290-1307. [PMID: 38942983 PMCID: PMC11446180 DOI: 10.1038/s43587-024-00662-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 05/30/2024] [Indexed: 06/30/2024]
Abstract
Investigating the genetic underpinnings of human aging is essential for unraveling the etiology of and developing actionable therapies for chronic diseases. Here, we characterize the genetic architecture of the biological age gap (BAG; the difference between machine learning-predicted age and chronological age) across nine human organ systems in 377,028 participants of European ancestry from the UK Biobank. The BAGs were computed using cross-validated support vector machines, incorporating imaging, physical traits and physiological measures. We identify 393 genomic loci-BAG pairs (P < 5 × 10-8) linked to the brain, eye, cardiovascular, hepatic, immune, metabolic, musculoskeletal, pulmonary and renal systems. Genetic variants associated with the nine BAGs are predominantly specific to the respective organ system (organ specificity) while exerting pleiotropic links with other organ systems (interorgan cross-talk). We find that genetic correlation between the nine BAGs mirrors their phenotypic correlation. Further, a multiorgan causal network established from two-sample Mendelian randomization and latent causal variance models revealed potential causality between chronic diseases (for example, Alzheimer's disease and diabetes), modifiable lifestyle factors (for example, sleep duration and body weight) and multiple BAGs. Our results illustrate the potential for improving human organ health via a multiorgan network, including lifestyle interventions and drug repurposing strategies.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA.
| | - Ye Ella Tian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Andrew Zalesky
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Wang R, Yan R, Jiao J, Li F, Zhang H, Chang Z, Wei H, Yan S, Li J. Fruit and vegetable intake and the risk of non-alcoholic fatty liver disease: a meta-analysis of observational studies. Front Nutr 2024; 11:1398184. [PMID: 38974809 PMCID: PMC11224539 DOI: 10.3389/fnut.2024.1398184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024] Open
Abstract
Purpose This systematic review and meta-analysis of clinical observational studies aims to clarify the correlation between the intake levels of fruits and vegetables and non-alcoholic fatty liver disease (NAFLD). Materials and methods PubMed, Embase, Web of Science, and the Cochrane Library were searched for studies on the association between vegetable or fruit intake with the risk of NAFLD from the foundation of each database up until September 2023. The relative risk (OR) and the 95% confidence interval (CI) were pooled for both the highest and lowest consumption levels of vegetables and fruits to explore their association with the incidence of NAFLD. Results The meta-analysis encompassed 11 studies with a total of 493,682 patients. A higher consumption of vegetables (OR = 0.78, 95% CI = 0.67-0.91) and fruits (OR = 0.88, 95% CI = 0.83-0.93) was found to have a negative correlation with the risk of NAFLD, denoting an inverse association. This correlation, however, varied among different ethnic groups and gender. Conclusions Our results indicate that increased consumption of vegetables and fruits is associated with a reduced likelihood of developing NAFLD. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/#searchadvanced, identifier: CRD42023460430.
Collapse
Affiliation(s)
- Rui Wang
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Ruijuan Yan
- Department of Hepatology, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Junzhe Jiao
- Department of Hepatology, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Feilong Li
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Haibo Zhang
- Advanced Instituted of Medicine Sciences, Dalian Medical University, Dalian, China
| | - Zhanjie Chang
- Department of Hepatology, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Hailiang Wei
- Department of General Surgery, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Shuguang Yan
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jingtao Li
- Department of Hepatology, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
11
|
Wen J, Tian YE, Skampardoni I, Yang Z, Cui Y, Anagnostakis F, Mamourian E, Zhao B, Toga AW, Zaleskey A, Davatzikos C. The Genetic Architecture of Biological Age in Nine Human Organ Systems. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.06.08.23291168. [PMID: 37398441 PMCID: PMC10312870 DOI: 10.1101/2023.06.08.23291168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Understanding the genetic basis of biological aging in multi-organ systems is vital for elucidating age-related disease mechanisms and identifying therapeutic interventions. This study characterized the genetic architecture of the biological age gap (BAG) across nine human organ systems in 377,028 individuals of European ancestry from the UK Biobank. We discovered 393 genomic loci-BAG pairs (P-value<5×10-8) linked to the brain, eye, cardiovascular, hepatic, immune, metabolic, musculoskeletal, pulmonary, and renal systems. We observed BAG-organ specificity and inter-organ connections. Genetic variants associated with the nine BAGs are predominantly specific to the respective organ system while exerting pleiotropic effects on traits linked to multiple organ systems. A gene-drug-disease network confirmed the involvement of the metabolic BAG-associated genes in drugs targeting various metabolic disorders. Genetic correlation analyses supported Cheverud's Conjecture1 - the genetic correlation between BAGs mirrors their phenotypic correlation. A causal network revealed potential causal effects linking chronic diseases (e.g., Alzheimer's disease), body weight, and sleep duration to the BAG of multiple organ systems. Our findings shed light on promising therapeutic interventions to enhance human organ health within a complex multi-organ network, including lifestyle modifications and potential drug repositioning strategies for treating chronic diseases. All results are publicly available at https://labs-laboratory.com/medicine.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Ye Ella Tian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Filippos Anagnostakis
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Andrew Zaleskey
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
12
|
Zhao B, Liu K, Liu X, Li Q, Li Z, Xi J, Xie F, Li X. Plant-derived flavonoids are a potential source of drugs for the treatment of liver fibrosis. Phytother Res 2024; 38:3122-3145. [PMID: 38613172 DOI: 10.1002/ptr.8193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 02/28/2024] [Accepted: 03/10/2024] [Indexed: 04/14/2024]
Abstract
Liver fibrosis is a dynamic pathological process that can be triggered by any chronic liver injury. If left unaddressed, it will inevitably progress to the severe outcomes of liver cirrhosis or even hepatocellular carcinoma. In the past few years, the prevalence and fatality of hepatic fibrosis have been steadily rising on a global scale. As a result of its intricate pathogenesis, the quest for pharmacological interventions targeting liver fibrosis has remained a formidable challenge. Currently, no pharmaceuticals are exhibiting substantial clinical efficacy in the management of hepatic fibrosis. Hence, it is of utmost importance to expedite the development of novel therapeutics for the treatment of this condition. Various research studies have revealed the ability of different natural flavonoid compounds to alleviate or reverse hepatic fibrosis through a range of mechanisms, which are related to the regulation of liver inflammation, oxidative stress, synthesis and secretion of fibrosis-related factors, hepatic stellate cells activation, and proliferation, and extracellular matrix synthesis and degradation by these compounds. This review summarizes the progress of research on different sources of natural flavonoids with inhibitory effects on liver fibrosis over the last decades. The anti-fibrotic effects of natural flavonoids have been increasingly studied, making them a potential source of drugs for the treatment of liver fibrosis due to their good efficacy and biosafety.
Collapse
Affiliation(s)
- Bolin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kai Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xing Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiuxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhibei Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingjing Xi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fan Xie
- Hospital of Chengdu University of Traditional Chinese Medicine 610032, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
13
|
Thompson AS, Jennings A, Bondonno NP, Tresserra-Rimbau A, Parmenter BH, Hill C, Perez-Cornago A, Kühn T, Cassidy A. Higher habitual intakes of flavonoids and flavonoid-rich foods are associated with a lower incidence of type 2 diabetes in the UK Biobank cohort. Nutr Diabetes 2024; 14:32. [PMID: 38778045 PMCID: PMC11111454 DOI: 10.1038/s41387-024-00288-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
AIM To examine the associations of a diet high in flavonoid-rich foods, as reflected by a "Flavodiet Score" (FDS), the major individual food contributors to flavonoid intake, and flavonoid subclasses with type 2 diabetes (T2D) risk in the UK Biobank cohort. MATERIALS AND METHODS Flavonoid intakes were estimated from ≥2 dietary assessments among 113,097 study participants [age at enrolment: 56 ± 8 years; 57% female] using the U.S Department of Agriculture (USDA) databases. Multivariable Cox proportional hazards models were used to investigate associations between dietary exposures and T2D. RESULTS During 12 years of follow-up, 2628 incident cases of T2D were identified. A higher FDS (compared to lower [Q4 vs. Q1]), characterised by an average of 6 servings of flavonoid-rich foods per day, was associated with a 26% lower T2D risk [HR: 0.74 (95% CI: 0.66-0.84), ptrend = <0.001]. Mediation analyses showed that lower body fatness and basal inflammation, as well as better kidney and liver function partially explain this association. In food-based analyses, higher intakes of black or green tea, berries, and apples were significantly associated with 21%, 15%, and 12% lower T2D risk. Among individual flavonoid subclasses, 19-28% lower risks of T2D were observed among those with the highest, compared to lowest intakes. CONCLUSIONS A higher consumption of flavonoid-rich foods was associated with lower T2D risk, potentially mediated by benefits to obesity/sugar metabolism, inflammation, kidney and liver function. Achievable increases in intakes of specific flavonoid-rich foods have the potential to reduce T2D risk.
Collapse
Affiliation(s)
- Alysha S Thompson
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Amy Jennings
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Nicola P Bondonno
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
- Danish Cancer Society Research Centre (DCRC), Copenhagen, Denmark
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Anna Tresserra-Rimbau
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
- Department of Nutrition, Food Science and Gastronomy, XIA, School of Pharmacy and Food Sciences, INSA, University of Barcelona, 08921, Barcelona, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029, Madrid, Spain
| | - Benjamin H Parmenter
- Nutrition & Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Claire Hill
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Aurora Perez-Cornago
- Nuffield Department of Population Health, Cancer Epidemiology Unit, University of Oxford, Oxford, UK
| | - Tilman Kühn
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK.
- Department of Nutritional Sciences, University of Vienna, Vienna, Austria.
- Medical University of Vienna, Center for Public Health, Vienna, Austria.
- Heidelberg Institute of Global Health (HIGH), Faculty of Medicine and University Hospital, Heidelberg, Germany.
| | - Aedín Cassidy
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK.
| |
Collapse
|
14
|
Huang F, Lyu B, Xie F, Li F, Xing Y, Han Z, Lai J, Ma J, Zou Y, Zeng H, Xu Z, Gao P, Luo Y, Bolund L, Tong G, Fengping X. From gut to liver: unveiling the differences of intestinal microbiota in NAFL and NASH patients. Front Microbiol 2024; 15:1366744. [PMID: 38638907 PMCID: PMC11024258 DOI: 10.3389/fmicb.2024.1366744] [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: 01/07/2024] [Accepted: 03/04/2024] [Indexed: 04/20/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is increasingly recognized for its global prevalence and potential progression to more severe liver diseases such as non-alcoholic steatohepatitis (NASH). The gut microbiota plays a pivotal role in the pathogenesis of NAFLD, yet the detailed characteristics and ecological alterations of gut microbial communities during the progression from non-alcoholic fatty liver (NAFL) to NASH remain poorly understood. Methods: In this study, we conducted a comparative analysis of gut microbiota composition in individuals with NAFL and NASH to elucidate differences and characteristics. We utilized 16S rRNA sequencing to compare the intestinal gut microbiota among a healthy control group (65 cases), NAFL group (64 cases), and NASH group (53 cases). Random forest machine learning and database validation methods were employed to analyze the data. Results: Our findings indicate a significant decrease in the diversity of intestinal flora during the progression of NAFLD (p < 0.05). At the phylum level, high abundances of Bacteroidetes and Fusobacteria were observed in both NAFL and NASH patients, whereas Firmicutes were less abundant. At the genus level, a significant decrease in Prevotella expression was seen in the NAFL group (AUC 0.738), whereas an increase in the combination of Megamonas and Fusobacterium was noted in the NASH group (AUC 0.769). Furthermore, KEGG pathway analysis highlighted significant disturbances in various types of glucose metabolism pathways in the NASH group compared to the NAFL group, as well as notably compromised flavonoid and flavonol biosynthesis functions. The study uncovers distinct microbiota characteristics and microecological changes within the gut during the transition from NAFL to NASH, providing insights that could facilitate the discovery of novel biomarkers and therapeutic targets for NAFLD.
Collapse
Affiliation(s)
- Furong Huang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Sanming Project of Medicine in Shenzhen, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Bo Lyu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Cell, Shenzhen, China
| | - Fanci Xie
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
- People's Hospital of Longhua, Shenzhen, China
| | - Fang Li
- BGI, Shenzhen, China
- Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, BGI Research, Qingdao, China
| | - Yufeng Xing
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Sanming Project of Medicine in Shenzhen, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Zhiyi Han
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Sanming Project of Medicine in Shenzhen, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Jianping Lai
- Department of Infectious Diseases, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | | | - Yuanqiang Zou
- BGI, Shenzhen, China
- Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
| | - Hua Zeng
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Sanming Project of Medicine in Shenzhen, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Zhe Xu
- BGI, Shenzhen, China
- Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, BGI Research, Qingdao, China
| | - Pan Gao
- BGI, Shenzhen, China
- Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, BGI Research, Qingdao, China
| | - Yonglun Luo
- Department of Sanming Project of Medicine in Shenzhen, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Cell, Shenzhen, China
- BGI, Shenzhen, China
- Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, BGI Research, Qingdao, China
| | - Lars Bolund
- Department of Sanming Project of Medicine in Shenzhen, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- BGI Cell, Shenzhen, China
- BGI, Shenzhen, China
- Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- Lars Bolund Institute of Regenerative Medicine, BGI-Qingdao, BGI Research, Qingdao, China
| | - Guangdong Tong
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- Department of Sanming Project of Medicine in Shenzhen, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xu Fengping
- Department of Sanming Project of Medicine in Shenzhen, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Cell, Shenzhen, China
- BGI, Shenzhen, China
| |
Collapse
|
15
|
Gao S, Wei L, Qin Y, Zhang P, Quan T, Liang F, Huang G. Network pharmacological analysis on the mechanism of Linggui Zhugan decoction for nonalcoholic fatty liver disease. Medicine (Baltimore) 2024; 103:e37281. [PMID: 38457573 PMCID: PMC10919485 DOI: 10.1097/md.0000000000037281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 03/10/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), represents a chronic progressive disease that imposes a significant burden on patients and the healthcare system. Linggui Zhugan decoction (LGZGD) plays a substantial role in treating NAFLD, but its exact molecular mechanism is unknown. Using network pharmacology, this study aimed to investigate the mechanism of action of LGZGD in treating NAFLD. Active ingredients and targets were identified through the integration of data from the TCMSP, GEO, GeneCards, and OMIM databases. Cytoscape 3.9.1 software, in conjunction with the STRING platform, was employed to construct network diagrams and screen core targets. The enrichment analysis of gene ontology and the Kyoto Encyclopedia of Genes and Genomes pathways were conducted by using the R. Molecular docking of the active ingredients and core targets was performed with AutoDock Vina software. We obtained 93 and 112 active ingredients and potential targets using the bioinformatic analysis of LGZGD in treating NAFLD. The primary ingredients of LGZGD included quercetin, kaempferol, and naringenin. The core targets were identified AKT1, MYC, HSP90AA1, HIF1A, ESR1, TP53, and STAT3. Gene ontology function enrichment analysis revealed associations with responses to nutrient and oxygen levels, nuclear receptor activity, and ligand-activated transcription factor activity. Kyoto Encyclopedia of Genes and Genomes signaling pathway analysis implicated the involvement of the PI3K-Akt, IL-17, TNF, Th17 cell differentiation, HIF-1, and TLR signaling pathways. Molecular docking studies indicated strong binding affinities between active ingredients and targets. LGZGD intervenes in NAFLD through a multi-ingredient, multi-target, and multi-pathway approach. Treatment with LGZGD can improve insulin resistance, oxidative stress, inflammation, and lipid metabolism associated with NAFLD.
Collapse
Affiliation(s)
- Songlin Gao
- Graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Liuting Wei
- Graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yan Qin
- Graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Peng Zhang
- Department of Nephrology, Liuzhou Traditional Chinese Medicine Hospital, Liuzhou, Guangxi, China
| | - Tingwei Quan
- Graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Fei Liang
- Graduate School of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Guihua Huang
- Department of Spleen and Stomach Liver Diseases, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
16
|
Xu H, Yuan M, Niu K, Yang W, Jiang M, Zhang L, Zhou J. Involvement of Bile Acid Metabolism and Gut Microbiota in the Amelioration of Experimental Metabolism-Associated Fatty Liver Disease by Nobiletin. Molecules 2024; 29:976. [PMID: 38474489 DOI: 10.3390/molecules29050976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Metabolism-associated fatty liver disease (MAFLD), a growing health problem worldwide, is one of the major risks for the development of cirrhosis and liver cancer. Oral administration of nobiletin (NOB), a natural citrus flavonoid, modulates the gut microbes and their metabolites in mice. In the present study, we established a mouse model of MAFLD by subjecting mice to a high-fat diet (HFD) for 12 weeks. Throughout this timeframe, NOB was administered to investigate its potential benefits on gut microbial balance and bile acid (BA) metabolism using various techniques, including 16S rRNA sequencing, targeted metabolomics of BA, and biological assays. NOB effectively slowed the progression of MAFLD by reducing serum lipid levels, blood glucose levels, LPS levels, and hepatic IL-1β and TNF-α levels. Furthermore, NOB reinstated diversity within the gut microbial community, increasing the population of bacteria that produce bile salt hydrolase (BSH) to enhance BA excretion. By exploring further, we found NOB downregulated hepatic expression of the farnesoid X receptor (FXR) and its associated small heterodimer partner (SHP), and it increased the expression of downstream enzymes, including cholesterol 7α-hydroxylase (CYP7A1) and cytochrome P450 27A1 (CYP27A1). This acceleration in cholesterol conversion within the liver contributes to mitigating MAFLD. The present findings underscore the significant role of NOB in regulating gut microbial balance and BA metabolism, revealing that long-term intake of NOB plays beneficial roles in the prevention or intervention of MAFLD.
Collapse
Affiliation(s)
- Hongling Xu
- School of Traditional Chinese Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Mingming Yuan
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| | - Kailin Niu
- School of Traditional Chinese Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei Yang
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| | - Maoyuan Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Lei Zhang
- School of Traditional Chinese Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| | - Jing Zhou
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| |
Collapse
|
17
|
Yang X, Zhuo S, Fang T. Interaction between dietary flavonoid intake and trouble sleeping on non-alcoholic fatty liver disease risk: a cross-sectional study. Eur J Gastroenterol Hepatol 2024; 36:210-219. [PMID: 38047726 DOI: 10.1097/meg.0000000000002687] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
OBJECTIVE The possible interaction of dietary flavonoid intake and sleep on non-alcoholic fatty liver disease (NAFLD) has not been well studied. This study investigated the interaction between dietary flavonoid intake and trouble sleeping on the risk of NAFLD. METHODS Three discrete National Health and Nutrition Examination Survey data cycles from 2007 to 2010 and 2017 to 2018 were used. NAFLD was diagnosed by a US Fatty Liver Index ≥30. A sleep questionnaire diagnosed trouble sleeping. Univariate and multivariate logistic regression, restricted cubic spline (RCS) and subgroup analyses were used to evaluate the association between dietary flavonoids, trouble sleeping and NAFLD. We employed the relative excess risk due to interaction, attributable proportion of interaction and synergy index to evaluate additive interactions. RESULTS Ultimately, 5056 participants were enrolled, and higher anthocyanidins and flavanones intake was negatively correlated with NAFLD. Conversely, trouble sleeping was positively associated with NAFLD. These correlations remained stable after adjusting for confounders, and there was a sex difference in this relationship. In the RCS model, anthocyanins were negatively non-linearly related to NAFLD, while flavanones showed a negative linear relationship. Moreover, there was a synergistic interplay between low dietary anthocyanin intake and trouble sleeping on the risk of NAFLD. A similar relationship existed for flavanone intake. CONCLUSION Anthocyanin and flavanone intake were negatively associated, whereas trouble sleeping was positively associated with NAFLD risk. There was a synergistic effect of low anthocyanin intake and trouble sleeping. The same relationship existed for low flavanone intake.
Collapse
Affiliation(s)
- Xinxia Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shitu Zhuo
- Department of Neurology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
18
|
Rai R, Ahmad Z, Jain SK, Jat D, Mishra SK. Naringenin suppresses aluminum-induced experimental hepato-nephrotoxicity in mice through modulation of oxidative stress and inflammation. Toxicol Res 2024; 40:97-110. [PMID: 38223678 PMCID: PMC10786781 DOI: 10.1007/s43188-023-00209-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 07/25/2023] [Accepted: 08/17/2023] [Indexed: 01/16/2024] Open
Abstract
Aluminum is a widely used metal substance in daily life activities that has been shown to cause severe hepato-nephrotoxicity with long-term exposure. Natural dietary flavonoids are being utilized as a newer pharmaceutical approach against various acute and chronic diseases. Naringenin (NAR) has shown efficient therapeutic properties, including effects against metal toxicities. However, the protective efficacy of NAR on aluminum chloride (AlCl3)-induced hepato-renal toxicity needs investigation as aluminum has shown serious environmental toxicity and bioaccumulation behavior. In this study, mice were treated with AlCl3 (10 mg/kg b.w./day) to assess toxicities, and a group of mice were co-treated with NAR (10 mg/kg b.w./day) to assess the protective effects of NAR against hepato-nephrotoxicity. The levels of blood serum enzymes, oxidative stress biomarkers, inflammatory cytokines, and the apoptosis marker caspase-3 were measured using histological examinations. NAR treatment in AlCl3-treated mice resulted in maintained levels of liver and kidney function enzymes and lipid profiles. NAR treatment attenuated oxidative stress by regulating the levels of nitric oxide, advance oxidation of protein products, protein carbonylation, and lipid peroxidation. NAR also replenished reduced antioxidant enzymes such as superoxide dismutase, catalase, glutathione peroxidase, glutathione reductase and reduced the levels of glutathione and oxidized glutathione. NAR regulated the levels of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) and elevated the levels of anti-inflammatory cytokines (IL-4, IL-10, and IFN-γ). The histological study further confirmed the protective effects of NAR against AlCl3-induced hepato-renal alterations. NAR decreased the expression of caspase-3 as a mechanism of protective effects against apoptotic damage in the liver and kidney of AlCl3-treated mice. In summary, this study demonstrated the antioxidant and anti-inflammatory properties of NAR, leading to the suppression of AlCl3-triggered hepato-renal apoptosis and histological alterations. The results suggest that aluminum toxicity needs to be monitored in daily life usage, and supplementation of the natural dietary flavonoid naringenin may help maintain liver and kidney health.
Collapse
Affiliation(s)
- Ravina Rai
- Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, 470003 M.P India
| | - Zaved Ahmad
- Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, 470003 M.P India
| | - Subodh Kumar Jain
- Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, 470003 M.P India
| | - Deepali Jat
- Department of Zoology, School of Biological Sciences, Dr. Harisingh Gour Central University, Sagar, 470003 M.P India
| | | |
Collapse
|
19
|
Wang R, Mao Y, Yu C, Rong Z, Wang R, Wang Y, Lv L, Gao Y, Wang Z, Zhang H. Research Progress of Natural Products with the Activity of Anti-nonalcoholic Steatohepatitis. Mini Rev Med Chem 2024; 24:1894-1929. [PMID: 38752645 DOI: 10.2174/0113895575306598240503054317] [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: 01/26/2024] [Revised: 03/19/2024] [Accepted: 03/29/2024] [Indexed: 10/16/2024]
Abstract
Nonalcoholic steatohepatitis (NASH), a multi-target disease, is becoming a global epidemic. Although several anti-NASH drug candidates are being evaluated in late-stage clinical trials, none have been approved by the FDA to date. Given the global prevalence of the disease, the lack of effective drugs, and the very limited therapeutic efficacy of most of the existing synthetic drugs focusing on a single target, there is an urgent need to continue to develop new therapeutic agents. In contrast, many natural products, including pure compounds and crude extracts, possess hepatoprotective activities. Usually, these natural components are characterized by multi-targeting and low side effects. Therefore, natural products are important resources for the development of new anti- NASH drugs. In this paper, we focus on reviewing the anti-NASH potential, structure, and some of the side effects of natural products based on structural classification. We hope this mini-review will help researchers design and develop new anti-NASH drugs, especially based on the structure of natural products.
Collapse
Affiliation(s)
- Rui Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuheng Mao
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Chunping Yu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhenji Rong
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ruyue Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yixin Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Linjin Lv
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yang Gao
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhigang Wang
- Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Hailong Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
20
|
Singh V, Singh R, Singh MP, Katrolia A. Therapeutic Role of Desmodium Species on its Isolated Flavonoids. Curr Mol Med 2024; 24:74-84. [PMID: 36515031 DOI: 10.2174/1566524023666221213111851] [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: 09/09/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 12/15/2022]
Abstract
Secondary metabolites are an important part to play a major role in society and it was isolated from plant flavonoids and useful in the treatment of various kinds of diseases in the human race. They are widely used as food and nutrition supplements as well as antioxidants. Traditionally, the Desmodium species are an important tool for the secondary metabolites to treat various diseases. Desmodium triquetrum (Fabaceae) one of the Indian medicinal plants is widely used in the treatment of asthma and inflammation. Three flavonoids isolated from Desmodium triquetrum Linn namely Baicalein, Naringin and Neohesperidin are useful as antioxidants, food and nutrition supplements, that help the body to function efficiently while protecting it against toxins as well stressors. The role of flavonoids may be due to the presence of the phenolic compound. Similarly, the flavonoids such as gangetin, gangetinin, desmocarpin and desmodin isolated from the species Desmodium gangeticum are responsible for antileishmanial, antioxidant, anti-arthritic, and immunomodulatory activities. Additionally, isolated flavanoids from the species Desmodium triflorum show antibacterial, antiepileptic, antifungal, and radioprotective activities. So, the aim of the present study, based on the literature miming from the desmodium species is to acknowledge the importance of flavonoids in human health as dietary food supplements and therapeutic uses.
Collapse
Affiliation(s)
- Vedpal Singh
- Department of Pharmacognosy, College of Pharmacy, JSS Academy of Technical Education, C-1/A sector 62 Noida, Uttar Pradesh, India
| | - Rohit Singh
- Department of Pharmaceutical Chemistry, College of Pharmacy, JSS Academy of Technical Education, C-1/A sector 62 Noida, Uttar Pradesh. India
| | - Manish Pal Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | - Archita Katrolia
- Department of Pharmaceutics, College of Pharmacy, JSS Academy of Technical Education, Noida, UP, India
| |
Collapse
|
21
|
Lopez-Pentecost M, Tamez M, Mattei J, Jacobs ET, Thomson CA, Garcia DO. Adherence to a Traditional Mexican Diet Is Associated with Lower Hepatic Steatosis in US-Born Hispanics of Mexican Descent with Overweight or Obesity. Nutrients 2023; 15:4997. [PMID: 38068856 PMCID: PMC10708445 DOI: 10.3390/nu15234997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Hispanics of Mexican descent have disproportionate rates of non-alcoholic fatty liver disease (NAFLD). The purpose of this work is to investigate the association between the traditional Mexican diet score (tMexS) and hepatic steatosis and fibrosis, two NAFLD-related clinical endpoints, in Hispanic adults of Mexican descent. Data from 280 Hispanic adults of Mexican descent (n = 102 men, 178 women) with overweight or obesity enrolled in a cross-sectional observational study were analyzed. The tMexS was calculated from 24 h dietary recalls. Hepatic steatosis and fibrosis measurements were assessed using transient elastography (Fibroscan®). Linear regression models testing the association between tMexS and hepatic steatosis and fibrosis were run individually and through the stratification of significant modifiers. Mean tMexS were 5.9 ± 2.1, hepatic steatosis scores were 288.9 ± 48.9 dB/m, and fibrosis scores were 5.6 ± 2.2 kPa. Among the US-born group, with every point increase in the tMexS, there was a statistically significant 5.7 lower hepatic steatosis point (95% CI: -10.9, -0.6, p-value = 0.07). Higher adherence to a traditional Mexican diet was associated with lower hepatic steatosis in US-born Hispanics of Mexican descent. Findings from the current work may serve to inform future culturally relevant interventions for NAFLD prevention and management in individuals of Mexican descent.
Collapse
Affiliation(s)
- Melissa Lopez-Pentecost
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Martha Tamez
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA; (M.T.); (J.M.)
| | - Josiemer Mattei
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA; (M.T.); (J.M.)
| | - Elizabeth T. Jacobs
- University of Arizona Cancer Center, The University of Arizona, Tucson, AZ 85724, USA (C.A.T.); (D.O.G.)
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ 85724, USA
| | - Cynthia A. Thomson
- University of Arizona Cancer Center, The University of Arizona, Tucson, AZ 85724, USA (C.A.T.); (D.O.G.)
- Department of Health Promotion Sciences, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ 85724, USA
| | - David O. Garcia
- University of Arizona Cancer Center, The University of Arizona, Tucson, AZ 85724, USA (C.A.T.); (D.O.G.)
- Department of Health Promotion Sciences, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
22
|
Datta S, Aggarwal D, Sehrawat N, Yadav M, Sharma V, Sharma A, Zghair AN, Dhama K, Sharma A, Kumar V, Sharma AK, Wang H. Hepatoprotective effects of natural drugs: Current trends, scope, relevance and future perspectives. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155100. [PMID: 37801892 DOI: 10.1016/j.phymed.2023.155100] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/02/2023] [Accepted: 09/17/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND The liver is a well-known player in the metabolism and removal of drugs. Drug metabolizing enzymes in the liver detoxify drugs and xenobiotics, ultimately leading to the acquisition of homeostasis. However, liver toxicity and cell damage are not only related to the nature and dosage of a particular drug but are also influenced by other factors such as aging, immune status, environmental contaminants, microbial metabolites, gender, obesity, and expression of individual genes Furthermore, factors such as drugs, alcohol, and environmental contaminants could induce oxidative stress, thereby impairing the regenerative potential of the liver and causing several diseases. Persons suffering from other ailments and those with comorbidities are found to be more prone to drug-induced toxicities. Moreover, drug composition and drug-drug interactions could further aggravate the risk of drug-induced hepatotoxicity. A plethora of mechanisms are responsible for initiating liver cell damage and further aggravating liver cell injury, followed by impairment of homeostasis, ultimately leading to the generation of reactive oxygen species, immune-suppression, and oxidative stress. OBJECTIVE To summarize the potential of phytochemicals and natural bioactive compounds to treat hepatotoxicity and other liver diseases. STUDY DESIGN A deductive qualitative content analysis approach was employed to assess the overall outcomes of the research and review articles pertaining to hepatoprotection induced by natural drugs, along with analysis of the interventions. METHODS An extensive literature search of bibliographic databases, including Web of Science, PUBMED, SCOPUS, GOOGLE SCHOLAR, etc., was carried out to understand the role of hepatoprotective effects of natural drugs. RESULTS Bioactive natural products, including curcumin, resveratrol, etc., have been seen as neutralizing agents against the side effects induced by the drugs. Moreover, these natural products are dietary and are readily available; thus, could be supplemented along with drugs to reduce toxicity to cells. Probiotics, prebiotics, and synbiotics have shown promise of improving overall liver functioning, and these should be evaluated more extensively for their hepatoprotective potential. Therefore, selecting an appropriate natural product or a bioactive compound that is free of toxicity and offers a reliable solution for drug-induced liver toxicity is quintessential. CONCLUSIONS The current review highlights the role of natural bioactive products in neutralizing drug-induced hepatotoxicity. Efforts have been made to delineate the possible underlying mechanism associated with the neutralization process.
Collapse
Affiliation(s)
- Sonal Datta
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India
| | - Diwakar Aggarwal
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India
| | - Nirmala Sehrawat
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India
| | - Mukesh Yadav
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India
| | - Varruchi Sharma
- Department of Biotechnology & Bioinformatics, Sri Guru Gobind Singh College, Chandigarh 160019, India
| | - Ajay Sharma
- Department of Chemistry, Career Point University, Tikker - Kharwarian, Hamirpur, Himachal Pradesh 176041, India
| | - Abdulrazzaq N Zghair
- College of Health and Medical Techniques, Middle Technical University, Baghdad, Iraq
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, UP, India
| | - Aanchal Sharma
- University Centre for Research and Development, University Institute of Biotechnology Chandigarh University, Gharuan, Mohali, India
| | - Vikas Kumar
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India
| | - Anil K Sharma
- Department of Biotechnology, Amity University, Sector-82-A, IT City Road, Mohali, Punjab 140306, India.
| | - Hailian Wang
- Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
23
|
Al Jadani JM, Albadr NA, Alshammari GM, Almasri SA, Alfayez FF, Yahya MA. Esculeogenin A, a Glycan from Tomato, Alleviates Nonalcoholic Fatty Liver Disease in Rats through Hypolipidemic, Antioxidant, and Anti-Inflammatory Effects. Nutrients 2023; 15:4755. [PMID: 38004149 PMCID: PMC10675668 DOI: 10.3390/nu15224755] [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: 10/09/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
This study examined the preventative effects of esculeogenin A (ESGA), a newly discovered glycan from tomato, on liver damage and hepatic steatosis in high-fat-diet (HFD)-fed male rats. The animals were divided into six groups (each of eight rats): a control group fed a normal diet, control + ESGA (200 mg/kg), HFD, and HFD + ESAG in 3 doses (50, 100, and 200 mg/kg). Feeding and treatments were conducted for 12 weeks. Treatment with ESGA did not affect gains in the body or fat weight nor increases in fasting glucose, insulin, and HOMA-IR or serum levels of free fatty acids (FFAs), tumor-necrosis factor-α, and interleukin-6 (IL-6). On the contrary, it significantly reduced the serum levels of gamma-glutamyl transpeptidase (GGT), aspartate aminotransferase (AST), alanine aminotransferase (ALT), total triglycerides (TGs), cholesterol (CHOL), and low-density lipoprotein cholesterol (LDL-c) in the HFD-fed rats. In addition, it improved the liver structure, attenuating the increase in fat vacuoles; reduced levels of TGs and CHOL, and the mRNA levels of SREBP1 and acetyl CoA carboxylase (ACC); and upregulated the mRNA levels of proliferator-activated receptor α (PPARα) and carnitine palmitoyltransferase I (CPT I) in HFD-fed rats. These effects were concomitant with increases in the mRNA, cytoplasmic, and nuclear levels of nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), and heme oxygenase-1 (HO); a reduction in the nuclear activity of nuclear factor-kappa beta (NF-κB); and inhibition of the activity of nuclear factor kappa B kinase subunit beta (IKKβ). All of these effects were dose-dependent effects in which a normal liver structure and normal levels of all measured parameters were seen in HFD + ESGA (200 mg/kg)-treated rats. In conclusion, ESGA prevents NAFLD in HFD-fed rats by attenuating hyperlipidemia, hepatic steatosis, oxidative stress, and inflammation by acting locally on Nrf2, NF-κB, SREBP1, and PPARα transcription factors.
Collapse
Affiliation(s)
- Jwharah M. Al Jadani
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (J.M.A.J.); (G.M.A.); (S.A.A.); (M.A.Y.)
| | - Nawal A. Albadr
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (J.M.A.J.); (G.M.A.); (S.A.A.); (M.A.Y.)
| | - Ghedeir M. Alshammari
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (J.M.A.J.); (G.M.A.); (S.A.A.); (M.A.Y.)
| | - Soheir A. Almasri
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (J.M.A.J.); (G.M.A.); (S.A.A.); (M.A.Y.)
| | - Farah Fayez Alfayez
- Department of Medicine and Surgery, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Mohammed Abdo Yahya
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia; (J.M.A.J.); (G.M.A.); (S.A.A.); (M.A.Y.)
| |
Collapse
|
24
|
Oh E, Lee J, Cho S, Kim SW, Won K, Shin WS, Gwak SH, Ha J, Jeon SY, Park JH, Song IS, Thoudam T, Lee IK, Kim S, Choi SY, Kim KT. Gossypetin Prevents the Progression of Nonalcoholic Steatohepatitis by Regulating Oxidative Stress and AMP-Activated Protein Kinase. Mol Pharmacol 2023; 104:214-229. [PMID: 37595967 DOI: 10.1124/molpharm.123.000675] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 08/20/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a severe liver metabolic disorder, however, there are still no effective and safe drugs for its treatment. Previous clinical trials used various therapeutic approaches to target individual pathologic mechanisms, but these approaches were unsuccessful because of the complex pathologic causes of NASH. Combinatory therapy in which two or more drugs are administered simultaneously to patients with NASH, however, carries the risk of side effects associated with each individual drug. To solve this problem, we identified gossypetin as an effective dual-targeting agent that activates AMP-activated protein kinase (AMPK) and decreases oxidative stress. Administration of gossypetin decreased hepatic steatosis, lobular inflammation and liver fibrosis in the liver tissue of mice with choline-deficient high-fat diet and methionine-choline deficient diet (MCD) diet-induced NASH. Gossypetin functioned directly as an antioxidant agent, decreasing hydrogen peroxide and palmitate-induced oxidative stress in the AML12 cells and liver tissue of MCD diet-fed mice without regulating the antioxidant response factors. In addition, gossypetin acted as a novel AMPK activator by binding to the allosteric drug and metabolite site, which stabilizes the activated structure of AMPK. Our findings demonstrate that gossypetin has the potential to serve as a novel therapeutic agent for nonalcoholic fatty liver disease /NASH. SIGNIFICANCE STATEMENT: This study demonstrates that gossypetin has preventive effect to progression of nonalcoholic steatohepatitis (NASH) as a novel AMP-activated protein kinase (AMPK) activator and antioxidants. Our findings indicate that simultaneous activation of AMPK and oxidative stress using gossypetin has the potential to serve as a novel therapeutic approach for nonalcoholic fatty liver disease /NASH patients.
Collapse
Affiliation(s)
- Eunji Oh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Jae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Sungji Cho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Sung Wook Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Kyung Won
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Won Sik Shin
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Seung Hee Gwak
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Joohun Ha
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - So Yeon Jeon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Jin-Hyang Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Im-Sook Song
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Themis Thoudam
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - In-Kyu Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Seonyong Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Se-Young Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| | - Kyong-Tai Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang Republic of Korea (E.O., J.L., S.C., S.W.K., K.W.J., W.S.S., S.H.G., K-T.K.); Department of Biochemistry and Molecular Biology, Graduate School, College of Medicine, Kyung Hee University, Seoul, Republic of Korea (J.H.); College of Pharmacy, Dankook University, Cheonan, Republic of Korea (S.Y.J.); College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea (J-H.P., I.-M.S.); Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea (T.T., I.-K.L.); Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea (I.-K.L.); Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea (S.K., S-Y.C.); and Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Republic of Korea (K.-T.K.)
| |
Collapse
|
25
|
Fiecke C, Simsek S, Sharma AK, Gallaher DD. Effect of red wheat, aleurone, and testa layers on colon cancer biomarkers, nitrosative stress, and gut microbiome composition in rats. Food Funct 2023; 14:9617-9634. [PMID: 37814914 DOI: 10.1039/d3fo03438k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
We previously found greater reduction of colon cancer (CC) biomarkers for red wheat compared to white wheat regardless of refinement state. In the present study we examined whether the phenolic-rich aleurone and testa layers are drivers of chemoprevention by red wheat and their influence on gut microbiota composition using a 1,2-dimethylhydrazine-induced CC rat model. Rats were fed a low-fat diet (16% of energy as fat), high-fat diet (50% of energy as fat), or high-fat diet containing whole red wheat, refined red wheat, refined white wheat, or aleurone- or testa-enriched fractions for 12 weeks. Morphological markers (aberrant crypt foci, ACF) were assessed after methylene blue staining and biochemical markers (3-nitrotyrosine [3-NT], Dclk1) by immunohistochemical determination of staining positivity within aberrant crypts. Gut microbiota composition was evaluated from 16S rRNA gene sequencing of DNA extracted from cecal contents. Relative to the high-fat diet, the whole and refined red wheat, refined white wheat, and testa-enriched fraction decreased ACF, while only the refined red wheat and aleurone-enriched fraction decreased 3-NT. No significant differences were observed for Dclk1. An increase in microbial diversity was observed for the aleurone-enriched fraction (ACE index) and whole red wheat (Inverse Simpson Index). The diet groups significantly modified overall microbiome composition, including altered abundances of Lactobacillus, Mucispirillum, Phascolarctobacterium, and Blautia coccoides. These results suggest that red wheat may reduce CC risk through modifications to the gut microbiota and nitrosative stress, which may be due, in part, to the influence of dietary fiber and the phenolic-rich aleurone layer.
Collapse
Affiliation(s)
- Chelsey Fiecke
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, 55108, USA.
| | - Senay Simsek
- North Dakota State University, Department of Plant Sciences, Cereal Science Graduate Program, Fargo, ND, 58105, USA
| | - Ashok Kumar Sharma
- Department of Animal Science, University of Minnesota, St. Paul, MN, 55108, USA
| | - Daniel D Gallaher
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN, 55108, USA.
| |
Collapse
|
26
|
Park JS, Rustamov N, Roh YS. The Roles of NFR2-Regulated Oxidative Stress and Mitochondrial Quality Control in Chronic Liver Diseases. Antioxidants (Basel) 2023; 12:1928. [PMID: 38001781 PMCID: PMC10669501 DOI: 10.3390/antiox12111928] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Chronic liver disease (CLD) affects a significant portion of the global population, leading to a substantial number of deaths each year. Distinct forms like non-alcoholic fatty liver disease (NAFLD) and alcoholic fatty liver disease (ALD), though they have different etiologies, highlight shared pathologies rooted in oxidative stress. Central to liver metabolism, mitochondria are essential for ATP production, gluconeogenesis, fatty acid oxidation, and heme synthesis. However, in diseases like NAFLD, ALD, and liver fibrosis, mitochondrial function is compromised by inflammatory cytokines, hepatotoxins, and metabolic irregularities. This dysfunction, especially electron leakage, exacerbates the production of reactive oxygen species (ROS), augmenting liver damage. Amidst this, nuclear factor erythroid 2-related factor 2 (NRF2) emerges as a cellular protector. It not only counters oxidative stress by regulating antioxidant genes but also maintains mitochondrial health by overseeing autophagy and biogenesis. The synergy between NRF2 modulation and mitochondrial function introduces new therapeutic potentials for CLD, focusing on preserving mitochondrial integrity against oxidative threats. This review delves into the intricate role of oxidative stress in CLD, shedding light on innovative strategies for its prevention and treatment, especially through the modulation of the NRF2 and mitochondrial pathways.
Collapse
Affiliation(s)
| | | | - Yoon-Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea; (J.-S.P.); (N.R.)
| |
Collapse
|
27
|
Alwadani AH, Almasri SA, Aloud AA, Albadr NA, Alshammari GM, Yahya MA. The Synergistic Protective Effect of γ-Oryzanol (OZ) and N-Acetylcysteine (NAC) against Experimentally Induced NAFLD in Rats Entails Hypoglycemic, Antioxidant, and PPARα Stimulatory Effects. Nutrients 2022; 15:nu15010106. [PMID: 36615764 PMCID: PMC9823776 DOI: 10.3390/nu15010106] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
This study estimated that the combined effect of γ-Oryzanol and N-acetylcysteine (NAC) against high-fat diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) in rats also estimated some of their mechanisms of action. Adult male rats were divided into seven groups (n = 8 each) as control, control + NAC, control + γ-Oryzanol, HFD, HFD + NAC, HFD + γ-Oryzanol, and HFD + NAC + γ-Oryzanol. NAC was administered orally at a final concentration of 200 mg/kg, whereas γ-Oryzanol was added to diets at a concentration of 0.16. All treatments were conducted for 17 weeks and daily. Both NAC and γ-Oryzanol were able to reduce final body weights, fat weights, fasting glucose, fasting insulin, serum, and serum levels of liver function enzymes as well as the inflammatory markers such as tumor necrosis factor-α (TNF-α), interleukine-6 (IL-6), and leptin in HFD-fed rats. They also improved hepatic structure and glucose tolerance, increased adiponectin levels, and reduced serum and hepatic levels of triglycerides (TGs) and cholesterol (CHOL) in these rats. These effects were concomitant with a reduction in the hepatic levels of lipid peroxides (MDA) and serum levels of LDL-C, but also with an increment in the hepatic levels of superoxide dismutase (SOD) and glutathione (GSH). Interestingly, only treatment with γ-Oryzanol stimulated the mRNA levels of proliferator-activated receptor alpha (PPARα) and carnitine palmitoyltransferase 1 (CPT1) in the liver and white adipose tissue (WAT) of rats. Of note, the combination therapy of both drugs resulted in maximum effects and restored almost normal liver structure and basal levels of all the above-mentioned metabolic parameters. In conclusion, a combination therapy of γ-Oryzanol and NAC is an effective therapy to treat NAFLD, which can act via several mechanisms on the liver and adipose tissue.
Collapse
Affiliation(s)
- Ashwag H. Alwadani
- Department of of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
- Department of Home Economics, University College in Farasan, Jazan University, Jazan 54943, Saudi Arabia
| | - Soheir A. Almasri
- Department of of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
- Correspondence:
| | - Amal A. Aloud
- Department of of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nawal A. Albadr
- Department of of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ghedeir M. Alshammari
- Department of of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Abdo Yahya
- Department of of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|