1
|
Zhang X, Zheng Y, Zhou C, Cao J, Pan D, Cai Z, Wu Z, Xia Q. Comparative physiological and transcriptomic analysis of sono-biochemical control over post-acidification of Lactobacillus delbrueckii subsp. bulgaricus. Food Microbiol 2024; 122:104563. [PMID: 38839237 DOI: 10.1016/j.fm.2024.104563] [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: 01/03/2024] [Revised: 04/27/2024] [Accepted: 05/16/2024] [Indexed: 06/07/2024]
Abstract
Thermosonication (UT) prestress treatments combining with varied fermentation patterns has been revealed as an effective method to regulate post-acidification as exerted by Lactobacillus delbrueckii subsp. bulgaricus (L. delbrueckii), but sono-biochemical controlling mechanisms remain elusive. This study employed physiological and transcriptomic analysis to explore the response mechanism of L. delbrueckii to UT-induced microstress (600 W, 33 kHz, 10 min). UT stress-induced inhibition of acidification of L. delbrueckii during (post)-fermentation was first confirmed, relying on the UT process parameters such as stress exposure duration and UT power. The significantly enhanced membrane permeability in cells treated by 600 W for 10 min than the microbes stressed by 420 W for 20 min suggested the higher dependence of UT-derived stresses on the treatment durations, relative to the ultrasonic powers. In addition, ultrasonication treatment-induced changes in cell membrane integrity enhanced and/or disrupted permeability of L. delbrueckii, resulting in an imbalance in intracellular conditions associated with corresponding alterations in metabolic behaviors and fermentation efficiencies. UT-prestressed inoculum exhibited a 21.46% decrease in the membrane potential during the lag phase compared to untreated samples, with an intracellular pH of 5.68 ± 0.12, attributed to the lower activities of H+-ATPase and lactate dehydrogenase due to UT stress pretreatments. Comparative transcriptomic analysis revealed that UT prestress influenced the genes related to glycolysis, pyruvate metabolism, fatty acid synthesis, and ABC transport. The genes encoding 3-oxoacyl-[acyl-carrier-protein] reductases I, II, and III, CoA carboxylase, lactate dehydrogenase, pyruvate oxidase, glucose-6-phosphate isomerase, and glycerol-3-phosphate dehydrogenase were downregulated, thus identifying the relevance of the UT microstresses-downregulated absorption and utilization of carbohydrates with the attenuated fatty acid production and energy metabolisms. These findings could contribute to provide a better understanding of the inactivated effects on the post-acidification of L. delbrueckii by ultrasonic pretreatments, thus providing theoretical basis for the targeted optimization of acidification inhibition efficiencies for yogurt products during chilled preservation processes.
Collapse
Affiliation(s)
- Xiaohui Zhang
- College of Food Science and Engineering, Zhejiang Key Laboratory of Intelligent Food Logistic and Processing, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, 315211, China
| | - Yuanrong Zheng
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, 200436, China
| | - Changyu Zhou
- College of Food Science and Engineering, Zhejiang Key Laboratory of Intelligent Food Logistic and Processing, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, 315211, China
| | - Jinxuan Cao
- School of Food and Health, China Food Flavor and Nutrition Health Innovation Center, Beijing Technology and Business University, 11 Fucheng Road, Beijing, 100048, China
| | - Daodong Pan
- College of Food Science and Engineering, Zhejiang Key Laboratory of Intelligent Food Logistic and Processing, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, 315211, China
| | - Zhendong Cai
- College of Food Science and Engineering, Zhejiang Key Laboratory of Intelligent Food Logistic and Processing, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, 315211, China
| | - Zhen Wu
- College of Food Science and Engineering, Zhejiang Key Laboratory of Intelligent Food Logistic and Processing, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, 315211, China.
| | - Qiang Xia
- College of Food Science and Engineering, Zhejiang Key Laboratory of Intelligent Food Logistic and Processing, Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
2
|
Skoczyńska A, Ołdakowska M, Dobosz A, Adamiec R, Gritskevich S, Jonkisz A, Lebioda A, Adamiec-Mroczek J, Małodobra-Mazur M, Dobosz T. PPARs in Clinical Experimental Medicine after 35 Years of Worldwide Scientific Investigations and Medical Experiments. Biomolecules 2024; 14:786. [PMID: 39062500 PMCID: PMC11275227 DOI: 10.3390/biom14070786] [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: 05/22/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
This year marks the 35th anniversary of Professor Walter Wahli's discovery of the PPARs (Peroxisome Proliferator-Activated Receptors) family of nuclear hormone receptors. To mark the occasion, the editors of the scientific periodical Biomolecules decided to publish a special issue in his honor. This paper summarizes what is known about PPARs and shows how trends have changed and how research on PPARs has evolved. The article also highlights the importance of PPARs and what role they play in various diseases and ailments. The paper is in a mixed form; essentially it is a review article, but it has been enriched with the results of our experiments. The selection of works was subjective, as there are more than 200,000 publications in the PubMed database alone. First, all papers done on an animal model were discarded at the outset. What remained was still far too large to describe directly. Therefore, only papers that were outstanding, groundbreaking, or simply interesting were described and briefly commented on.
Collapse
Affiliation(s)
- Anna Skoczyńska
- Department of Internal and Occupational Medicine and Hypertension, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Monika Ołdakowska
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| | - Agnieszka Dobosz
- Department of Basic Medical Sciences and Immunology, Division of Basic Medical Sciences, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland
| | - Rajmund Adamiec
- Department of Diabetology and Internal Medicine, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
- Department of Internal Medicine, Faculty of Medical and Technical Sciences, Karkonosze University of Applied Sciences, Lwówiecka 18, 58-506 Jelenia Góra, Poland
| | - Sofya Gritskevich
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| | - Anna Jonkisz
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| | - Arleta Lebioda
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| | - Joanna Adamiec-Mroczek
- Department of Ophthalmology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Małgorzata Małodobra-Mazur
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| | - Tadeusz Dobosz
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland; (M.O.); (A.J.); (A.L.); (M.M.-M.); (T.D.)
| |
Collapse
|
3
|
Crosby-Galvan MM, Mendoza GD, Hernández-García PA, Martínez-García JA, Vázquez-Valladolid A, Cifuentes-López RO, Lee-Rangel HA. Influence of supplemental choline on milk yield, fatty acid profile, and weight changes in postpartum ewes and their offspring. Vet World 2024; 17:1265-1270. [PMID: 39077444 PMCID: PMC11283600 DOI: 10.14202/vetworld.2024.1265-1270] [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: 02/05/2024] [Accepted: 05/13/2024] [Indexed: 07/31/2024] Open
Abstract
Background and Aim The most intensive nutritional requirements occur during milk production's peak. Ewe milk contains more protein and fat than cow milk. The nutritional factors significantly determine the composition. The liver undergoes high stress during lactation but is relieved by essential nutrients. Choline acts metabolically as a lipotrope. This compound functions in cell structure construction, maintenance, and acetylcholine synthesis. The animal nutrition industry provides choline from various sources, such as synthetic and natural kinds. This study evaluated the influence of two distinct choline sources on dairy ewes' peripartum and postpartum milk production, composition, and offspring growth. Materials and Methods Twenty-four Rambouillet ewes, each weighing around 63.7 ± 1.7 kg, aged three with two previous births, spent 30-day pre-partum and post-partum in individual pens (2 × 2 m). They were given different experimental treatments 30 days before and after birth according to a randomized design; no choline (a), 4 g/day rumen-protected choline (RPC) (b), or 4 g/day thiocholine (c). Milk samples for milk composition and long-chain fatty acid (FA) analysis were taken every 30 days during milk collection. Results Significant differences (p < 0.05) in ewe body weight, lamb birth weight, and 30-day-old lamb body weight were observed at lambing and on day 30 of lactation due to choline treatment. Milk yield was significantly higher (1.57 kg/day) compared to the control (1.02 kg/day) and RPC (1.39 kg/day), due to the herbal choline source. There was no significant difference in the milk's protein, lactose, fat, non-fat solids, and total milk solids content between the treatments. Herbal choline lowers (p < 0.05) the concentrations of caproic, caprylic, capric, lauric, and myristic acids while boosting (p < 0.05) those of oleic and cis-11-eicosenoic acid, the changes influencing long-chain FA levels (p < 0.05). Conclusion Providing choline from both sources to ewes enhanced milk production and body weight at lambing and on 30-day post-lambing. The herbal choline supplement altered short-chain milk FAs, while representative concentration pathways affected medium-chain ones.
Collapse
Affiliation(s)
| | - German D. Mendoza
- Agriculture and Animal Science Department, Xochimilco Campus, Metropolitan Autonomous University, Mexico City, México
| | | | - José Antonio Martínez-García
- Agriculture and Animal Science Department, Xochimilco Campus, Metropolitan Autonomous University, Mexico City, México
| | | | | | - Héctor A. Lee-Rangel
- Agronomy and Veterinary Faculty, Bioscience Centre, San Luis Potosí Autonomous University, México
| |
Collapse
|
4
|
Zhang D, Ding H, Liu C, Huang Y, Tai W, Feng S, Wang X, Zhao C, Li Y. Circulating exosome-mediated AMPKα-SIRT1 pathway regulates lipid metabolism disorders in calf hepatocytes. Res Vet Sci 2024; 169:105177. [PMID: 38350170 DOI: 10.1016/j.rvsc.2024.105177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 02/15/2024]
Abstract
Subclinical ketosis (SCK) in dairy cows is often misdiagnosed because it lacks clinical signs and detection indicators. However, it is highly prevalent and may transform into clinical ketosis if not treated promptly. Due to the negative energy balance, a large amount of fat is mobilized, producing NEFA that exceeds the upper limit of liver processing, which in turn leads to the disturbance of liver lipid metabolism. The silent information regulator 1 (SIRT1) is closely related to hepatic lipid metabolism disorders. Exosomes as signal transmitters, also play a role in the circulatory system. We hypothesize that the circulating exosome-mediated adenosine 5'-monophosphate (AMP)-activated protein kinase alpha (AMPKα)-SIRT1 pathway regulates lipid metabolism disorders in SCK cows. We extracted the exosomes required for the experiment from the peripheral circulating blood of non-ketotic (NK) and SCK cows. We investigated the effect of circulating exosomes on the expression levels of mRNA and protein of the AMPKα-SIRT1 pathway in non-esterified fatty acid (NEFA)-induced dairy cow primary hepatocytes using in vitro cell experiments. The results showed that circulating exosomes increased the expression levels of Lipolysis-related genes and proteins (AMPKα, SIRT1, and PGC-1α) in hepatocytes treated with 1.2 mM NEFA, and inhibited the expression of lipid synthesis-related genes and protein (SREBP-1C). The regulation of exosomes on lipid metabolism disorders caused by 1.2 mM NEFA treatment showed the same trend as for SIRT1-overexpressing adenovirus. The added exosomes could regulate NEFA-induced lipid metabolism in hepatocytes by mediating the AMPKα-SIRT1 pathway, consistent with the effect of transfected SIRT1 adenovirus.
Collapse
Affiliation(s)
- Daoliang Zhang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, Anhui Province 230036, China
| | - Hongyan Ding
- Research Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, Anhui Province 230031, China
| | - Chang Liu
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, Anhui Province 230036, China
| | - Yingying Huang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, Anhui Province 230036, China
| | - Wenjun Tai
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, Anhui Province 230036, China
| | - Shibin Feng
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, Anhui Province 230036, China
| | - Xichun Wang
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, Anhui Province 230036, China
| | - Chang Zhao
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, Anhui Province 230036, China.
| | - Yu Li
- College of Animal Science and Technology, Anhui Agricultural University, 130 West Changjiang Road, Hefei, Anhui Province 230036, China.
| |
Collapse
|
5
|
Lu W, Yang J, Hu M, Zhong K, Wang Y, Yang Y, Loor JJ, Yang G, Han L. Effects of choline deficiency and supplementation on lipid droplet accumulation in bovine primary liver cells in vitro. J Dairy Sci 2023; 106:9868-9878. [PMID: 37678795 DOI: 10.3168/jds.2023-23452] [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: 03/03/2023] [Accepted: 07/03/2023] [Indexed: 09/09/2023]
Abstract
Rumen-protected choline (RPC) supplementation in the periparturient period has in some instances prevented and alleviated fatty liver disease in dairy cows. Mechanistically, however, it is unclear how choline prevents the accumulation of lipid droplets (LD) in liver cells. In this study, primary liver cells isolated from liver tissue obtained via puncture biopsy from 3 nonpregnant mid-lactation multiparous Holstein cows (∼160 d postpartum) were used. Analyses of LD via oil red O staining, protein abundance via Western blotting, and phospholipid content and composition measured by thin-layer chromatography and HPLC/mass spectrometry were performed in liver cells cultured in choline-deficient medium containing 150 μmol/L linoleic acid for 24 h. In a subsequent experiment, lipophagy was assessed in liver cells cultured with 30, 60, or 90 µmol/L choline-chloride. All data were analyzed statistically using SPSS 20.0 via t-tests or one-way ANOVA. Compared with liver cells cultured in Dulbecco's Modified Eagle Medium alone, choline deficiency increased the average diameter of LD (1.59 vs. 2.10 µm), decreased the proportion of small LD (<2 µm) from 75.3% to 56.6%, and increased the proportion of large LD (>4 µm) from 5.6% to 15.0%. In addition, the speed of LD fusion was enhanced by the absence of choline. Among phospholipid species, the phosphatidylcholine (PC) content of liver cells decreased by 34.5%. Seventeen species of PC (PC [18:2_22:6], PC [15:0_16:1], PC [14:0_20:4], and so on) and 6 species of lysophosphatidylcholine (LPC; LPC [15:0/0:0]), PC (22:2/0:0), LPC (20:2/0:0), and so on] were decreased, while PC (14:1_16:1) and LPC (0:0/20:1) were increased. Choline deficiency increased the triglyceride (TAG) content (0.57 vs. 0.39 μmol/mg) in liver cells and increased the protein abundance of sterol regulatory element binding protein 1, sterol regulatory element binding protein cleavage activation protein, and fatty acid synthase by 23.5%, 17%, and 36.1%, respectively. Upon re-supplementation with choline, the phenotype of LD (TAG content, size, proportion, and phospholipid profile) was reversed, and the ratio of autophagy marker LC3II/LC3I protein was significantly upregulated in a dose-dependent manner. Overall, at least in vitro in mid-lactation cows, these data demonstrated that PC synthesis is necessary for normal LD formation, and both rely on choline availability. According to the limitation of the source of liver cells used, further work should be conducted to ascertain that these effects are applicable to liver cells from postpartum cows, the physiological stage where the use of RPC has been implemented for the prevention and treatment of fatty liver.
Collapse
Affiliation(s)
- Wenyan Lu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, P.R. China
| | - Jingna Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, P.R. China
| | - Mingyue Hu
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, P.R. China
| | - Kai Zhong
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, P.R. China
| | - Yueying Wang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, P.R. China
| | - Yanbin Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, P.R. China
| | - Juan J Loor
- Department of Animal Science and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801
| | - Guoyu Yang
- Key Laboratory of Animal Growth and Development of Henan Province, College of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, P.R. China
| | - Liqiang Han
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, P.R. China.
| |
Collapse
|
6
|
Maynard CW, Gilbert E, Yan F, Cline MA, Dridi S. Peripheral and Central Impact of Methionine Source and Level on Growth Performance, Circulating Methionine Levels and Metabolism in Broiler Chickens. Animals (Basel) 2023; 13:1961. [PMID: 37370471 DOI: 10.3390/ani13121961] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/22/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
The present study was designed to evaluate the effects of DL-methionine (DL-Met) 2-hydroxy-4-(methylthio) butanoic acid (HMTBa), or S-(5'-Adenosyl)-L-methionine chloride (SAM), using feeding trial and central administration, on live performance, plasma metabolites, and the expression of feeding-related hypothalamic neuropeptides in broilers raised to a market age (35 d). Final average body weight (BW) and feed conversion ratio (FCR) from the feeding trial exceeded the performance measurements published by the primary breeder. At d35, the MTBHa group had better BW and lower feed intake, which resulted in a better FCR than the DL-Met group at 87 TSAA to lysine. At the molecular levels, the expression of hypothalamic neuropeptide (NPY) and monocarboxylate transporter (MCT) 2 did not differ between all treated groups; however, the mRNA abundances of hypothalamic MCT1 and orexin (ORX) were significantly upregulated in DL-Met- treated groups compared to the control. The ICV administration of SAM significantly reduced feed intake at all tested periods (from 30 to 180 min post injection) compared to the aCSF-treated group (control). The central administration of HMTBa increased feed intake, which reached a significant level only 60 min post administration, compared to the control group. ICV administration of DL-Met slightly increased feed intake compared to the control group, but the difference was not statistically discernable. Quantitative real-time PCR analysis showed that the hypothalamic expression of NPY, cocaine- and amphetamine-regulated transcript, MCT1, and MCT2 was significantly upregulated in the ICV-HMTBa group compared to the aCSF birds. The hypothalamic expression of the mechanistic target of rapamycin (mTOR), AMP-activated protein kinase (AMPKα1), D-amino acid oxidase, and hydroxyacid oxidase was significantly upregulated in DL-Met compared to the control group. The mRNA abundances of ORX were significantly increased in the hypothalamus of both DL-Met and HMTBa groups compared to the aCSF birds; however, mTOR gene expression was significantly downregulated in the SAM compared to the control group. Taken together, these data show, for the first time, that DL-Met and HMTBa have a common downstream (ORX) pathway, but also a differential central pathway, typically NPY-MCT for HMTBa and mTOR-AMPK for methionine.
Collapse
Affiliation(s)
- Craig W Maynard
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Elizabeth Gilbert
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Frances Yan
- Novus International, Saint Charles, MO 63304, USA
| | - Mark A Cline
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Sami Dridi
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
7
|
Li X, Du Y, Xue C, Kang X, Sun C, Peng H, Fang L, Han Y, Xu X, Zhao C. SIRT2 Deficiency Aggravates Diet-Induced Nonalcoholic Fatty Liver Disease through Modulating Gut Microbiota and Metabolites. Int J Mol Sci 2023; 24:8970. [PMID: 37240315 PMCID: PMC10219207 DOI: 10.3390/ijms24108970] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/05/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), characterized by excessive lipid accumulation in hepatocytes, is an increasing global healthcare burden. Sirtuin 2 (SIRT2) functions as a preventive molecule for NAFLD with incompletely clarified regulatory mechanisms. Metabolic changes and gut microbiota imbalance are critical to the pathogenesis of NAFLD. However, their association with SIRT2 in NAFLD progression is still unknown. Here, we report that SIRT2 knockout (KO) mice are susceptible to HFCS (high-fat/high-cholesterol/high-sucrose)-induced obesity and hepatic steatosis accompanied with an aggravated metabolic profile, which indicates SIRT2 deficiency promotes NAFLD-NASH (nonalcoholic steatohepatitis) progression. Under palmitic acid (PA), cholesterol (CHO), and high glucose (Glu) conditions, SIRT2 deficiency promotes lipid deposition and inflammation in cultured cells. Mechanically, SIRT2 deficiency induces serum metabolites alteration including upregulation of L-proline and downregulation of phosphatidylcholines (PC), lysophosphatidylcholine (LPC), and epinephrine. Furthermore, SIRT2 deficiency promotes gut microbiota dysbiosis. The microbiota composition clustered distinctly in SIRT2 KO mice with decreased Bacteroides and Eubacterium, and increased Acetatifactor. In clinical patients, SIRT2 is downregulated in the NALFD patients compared with healthy controls, and is associated with exacerbated progression of normal liver status to NAFLD to NASH in clinical patients. In conclusion, SIRT2 deficiency accelerates HFCS-induced NAFLD-NASH progression by inducing alteration of gut microbiota and changes of metabolites.
Collapse
Affiliation(s)
- Xingyu Li
- Department of Infectious Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang 050011, China;
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; (Y.D.)
| | - Yimeng Du
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; (Y.D.)
| | - Chunyuan Xue
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; (Y.D.)
| | - Xiaofeng Kang
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; (Y.D.)
| | - Chao Sun
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; (Y.D.)
| | - Huanyan Peng
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; (Y.D.)
| | - Liaoxin Fang
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; (Y.D.)
| | - Yuchen Han
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; (Y.D.)
| | - Xiaojie Xu
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing 100850, China; (Y.D.)
| | - Caiyan Zhao
- Department of Infectious Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang 050011, China;
| |
Collapse
|
8
|
Zhang J, Gaowa N, Wang Y, Li H, Cao Z, Yang H, Zhang X, Li S. Complementary hepatic metabolomics and proteomics reveal the adaptive mechanisms of dairy cows to the transition period. J Dairy Sci 2023; 106:2071-2088. [PMID: 36567250 DOI: 10.3168/jds.2022-22224] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 09/06/2022] [Indexed: 12/24/2022]
Abstract
The transition period from late pregnancy to early lactation is a vital time of the lifecycle of dairy cows due to the marked metabolic challenges. Besides, the liver is the pivot point of metabolism in cattle. Nevertheless, the hepatic physiological molecular adaptation during the transition period has not been elucidated, especially from the metabolomics and proteomics view. Therefore, the present study aims to investigate the hepatic metabolic alterations in transition cows by using integrative metabolomics and proteomics methods. Gas chromatography quadrupole-time-of-flight mass spectrometry-based metabolomics and data-independent acquisition-based quantitative proteomics methods were used to analyze liver tissues collected from 8 healthy multiparous Holstein dairy cows 21 d before and after calving. In total, 44 metabolites and 250 proteins were identified as differentially expressed from 233 metabolites and 3,539 proteins detected from the liver biopsies during the transition period. Complementary functional analysis of different metabolites and proteins indicated the upregulated gluconeogenesis, tricarboxylic acid cycles, AA degradation, fatty acid oxidation, AMP-activated protein kinase signaling pathway, peroxisome proliferator-activated receptor signaling pathway, and ribosome proteins in postpartum dairy cows. In terms of the metabolites and proteins, glucose-6-phosphate, fructose-6-phosphate, carnitine palmitoyltransferase 1A, and phosphoenolpyruvate carboxykinase played a significant role in these pathways. The upregulated oxidative status may be accompanied by the pathways mentioned above. In addition, the upregulated glucagon and insulin signaling pathways also indicated the significant requirement for glucose in postpartum dairy cows. These outcomes, from the view of global metabolites and proteins, may present a better comprehension of the biology of the transition period, which can be helpful in further developing nutritional regulation strategies targeting the liver to help cows overcome this metabolically challenging time.
Collapse
Affiliation(s)
- Jun Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100 China; State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Naren Gaowa
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Yajing Wang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Huanxu Li
- Beijing Oriental Kingherd Biotechnology Company, Beijing 100193, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Hongjian Yang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Xiaoming Zhang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193 China.
| |
Collapse
|
9
|
Carneiro TJ, Vojtek M, Gonçalves-Monteiro S, Batista de Carvalho ALM, Marques MPM, Diniz C, Gil AM. Effect of Pd 2Spermine on Mice Brain-Liver Axis Metabolism Assessed by NMR Metabolomics. Int J Mol Sci 2022; 23:13773. [PMID: 36430252 PMCID: PMC9693583 DOI: 10.3390/ijms232213773] [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: 10/21/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022] Open
Abstract
Cisplatin (cDDP)-based chemotherapy is often limited by severe deleterious effects (nephrotoxicity, hepatotoxicity and neurotoxicity). The polynuclear palladium(II) compound Pd2Spermine (Pd2Spm) has emerged as a potential alternative drug, with favorable pharmacokinetic/pharmacodynamic properties. This paper reports on a Nuclear Magnetic Resonance metabolomics study to (i) characterize the response of mice brain and liver to Pd2Spm, compared to cDDP, and (ii) correlate brain-liver metabolic variations. Multivariate and correlation analysis of the spectra of polar and lipophilic brain and liver extracts from an MDA-MB-231 cell-derived mouse model revealed a stronger impact of Pd2Spm on brain metabolome, compared to cDDP. This was expressed by changes in amino acids, inosine, cholate, pantothenate, fatty acids, phospholipids, among other compounds. Liver was less affected than brain, with cDDP inducing more metabolite changes. Results suggest that neither drug induces neuronal damage or inflammation, and that Pd2Spm seems to lead to enhanced brain anti-inflammatory and antioxidant mechanisms, regulation of brain bioactive metabolite pools and adaptability of cell membrane characteristics. The cDDP appears to induce higher extension of liver damage and an enhanced need for liver regeneration processes. This work demonstrates the usefulness of untargeted metabolomics in evaluating drug impact on multiple organs, while confirming Pd2Spm as a promising replacement of cDDP.
Collapse
Affiliation(s)
- Tatiana J. Carneiro
- Department of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Martin Vojtek
- LAQV/REQUIMTE—Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755 Porto, Portugal
| | - Salomé Gonçalves-Monteiro
- LAQV/REQUIMTE—Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755 Porto, Portugal
| | | | - Maria Paula M. Marques
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Carmen Diniz
- LAQV/REQUIMTE—Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4150-755 Porto, Portugal
| | - Ana M. Gil
- Department of Chemistry, CICECO—Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
10
|
Hasson DC, Watanabe-Chailland M, Romick-Rosendale L, Koterba A, Miner DS, Lahni P, Ma Q, Goldstein SL, Devarajan P, Standage SW. Choline supplementation attenuates experimental sepsis-associated acute kidney injury. Am J Physiol Renal Physiol 2022; 323:F255-F271. [PMID: 35834274 PMCID: PMC9394731 DOI: 10.1152/ajprenal.00033.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 11/22/2022] Open
Abstract
Acute kidney injury (AKI) is common in critically ill patients, and sepsis is its leading cause. Sepsis-associated AKI (SA-AKI) causes greater morbidity and mortality than other AKI etiologies, yet the underlying mechanisms are incompletely understood. Metabolomic technologies can characterize cellular energy derangements, but few discovery analyses have evaluated the metabolomic profile of SA-AKI. To identify metabolic derangements amenable to therapeutic intervention, we assessed plasma and urine metabolites in septic mice and critically ill children and compared them by AKI status. Metabolites related to choline and central carbon metabolism were differentially abundant in SA-AKI in both mice and humans. Gene expression of enzymes related to choline metabolism was altered in the kidneys and liver of mice with SA-AKI. Treatment with intraperitoneal choline improved renal function in septic mice. Because pediatric patients with sepsis displayed similar metabolomic profiles to septic mice, choline supplementation may attenuate pediatric septic AKI.NEW & NOTEWORTHY Altered choline metabolism plays a role in both human and murine sepsis-associated acute kidney injury (SA-AKI), and choline administration in experimental SA-AKI improved renal function. These findings indicate that 1) mouse models can help interrogate clinically relevant mechanisms and 2) choline supplementation may ameliorate human SA-AKI. Future research will investigate clinically the impact of choline supplementation on human renal function in sepsis and, using model systems, how choline mediates its effects.
Collapse
Affiliation(s)
- Denise C Hasson
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Miki Watanabe-Chailland
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Lindsey Romick-Rosendale
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adeleine Koterba
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Dashiell S Miner
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Patrick Lahni
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Qing Ma
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stuart L Goldstein
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Prasad Devarajan
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Stephen W Standage
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
11
|
Gong J, Zhao S, Heng N, Wang Y, Hu Z, Wang H, Zhu H. The Dynamic Transcription Profiles of Proliferating Bovine Ovarian Granulosa When Exposed to Increased Levels of β-Hydroxybutyric Acid. Front Vet Sci 2022; 9:915956. [PMID: 35990259 PMCID: PMC9389329 DOI: 10.3389/fvets.2022.915956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
Ketosis is common in high-yield dairy cows. It is a condition that is characterized by the accumulation of serum β-hydroxybutyric acid (BHBA). Both subclinical ketosis and clinical ketosis can compromise the reproductive performance and cause long-lasting negative effects on reproductive efficiency by affecting the proliferation of follicular and granulosa cells. However, the regulatory mechanisms involved in the development of follicular cells and granulosa cells in cows experiencing subclinical ketosis and clinical ketosis remain largely unknown. To investigate the effect of a ketosis-triggered increase in BHBA on bovine follicular granulosa cell development, we detected a significant reduction in the proliferation of granulosa cells (P < 0.05) in the BHBA-1.2 mM and BHBA-2.4 mM groups and a significant increase in the number of granulosa cells in the G1 phase of the cell cycle (P < 0.05). RNA-seq and trend analysis were used to identify differentially expressed genes by comparing three clusters: low-concentration response to 1.2 mM BHBA, high-concentration response to 2.4 mM BHBA, and the similar trend (up or down) response following BHBA concentration increased. GO and KEGG enrichment analyses were performed separately for each cluster. Analysis showed that two novel down-regulated genes (G0S2 and S100A6), which are associated with cell proliferation and cycle progression, were enriched in the low-concentration response to 1.2 mM BHBA. Another differentially expressed gene (PARP), which plays a role in the apoptotic pathway, was enriched in the high-concentration response to 2.4 mM BHBA. We also found that CYP27B1 and CYP17A1, which are associated with Ca2+ homeostasis and estrogen synthesis, were enriched in a similar trend response. In conclusion, we describe the dynamic transcription profiles of granulosa cells under different levels of β-hydroxybutyric stress and report key regulators that may underlie the detrimental effects on the development of follicles and granulosa cells, thus representing potential therapeutic targets to improve fertility in dairy cows with subclinical ketosis or clinical ketosis.
Collapse
|
12
|
Riosa R, Ghaffari MH, Hammon HM, Süss D, Hoelker M, Drillich M, Parys C, Guyader J, Sauerwein H, Iwersen M. Identification and characterization of dairy cows with different backfat thickness antepartum in relation to postpartum loss of backfat thickness: A cluster analytic approach. J Dairy Sci 2022; 105:6327-6338. [PMID: 35525619 DOI: 10.3168/jds.2021-21434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/02/2022] [Indexed: 11/19/2022]
Abstract
The objectives of this study were (1) to characterize the interindividual variation in the relationship between antepartum (ap) backfat thickness (BFT) and subsequent BFT loss during early lactation in a large dairy herd using cluster analysis; (2) to compare the serum concentrations of metabolites (nonesterified fatty acids, β-hydroxybutyrate), metabolic hormones (leptin and adiponectin), and an inflammatory marker (haptoglobin) among the respective clusters; and (3) to compare lactation performance and uterine health status in the different clusters. An additional objective was (4) to investigate differences in these serum variables and in milk yield of overconditioned (OC) cows that differed in the extent of BFT loss. Using data from a large study of 1,709 multiparous Holstein cows, we first selected those animals from which serum samples and BFT results (mm) were available at d 25 (±10) ap and d 31 (±3 d) postpartum (pp). The remaining 713 cows (parity of 2 to 7) were then subjected to cluster analysis: different approaches based on the BFT of the cows were performed. K-means (unsupervised machine learning algorithm) clustering based on BFT-ap alone identified 5 clusters: lean (5-8 mm BFT, n = 50), normal (9-12 mm, n = 206), slightly fat (SF; 13-16 mm, n = 203), just fat (JF; 16-22 mm, n = 193), and very fat (VF; 23-43 mm, n = 61). Clustering by difference between BFT-ap and BFT-pp (ΔBFT) also revealed 5 clusters: extreme loss (17-23 mm ΔBFT, n = 16), moderate loss (9-15 mm, n = 119), little loss (4-8 mm, n = 326), no loss (0-3 mm, n = 203), and gain (-8 to -1 mm, n = 51). Based on the blood variables measured, our results confirm that cows with greater BFT losses had higher lipid mobilization and ketogenesis than cows with less BFT loss. The serum variables of cows that gained BFT did not differ from normal cows. Milk yield was affected by the BFT-ap cluster, but not by the ΔBFT cluster. Cows categorized as VF had lesser milk yield than other clusters. We further compared the OC cows that had little or no BFT loss (i.e., 2% of VF, 12% of JF, and 31% of SF, OC-no loss, n = 85) with the OC cows that lost BFT (OC-loss, n = 135). Both NEFA and BHB pp concentrations and milk yield were greater in OC-loss cows compared with the OC-no loss cows. The serum concentration of leptin ap was greater in OC-loss than in the OC-no loss cows. Overall, OC cows lost more BFT than normal or lean cows. However, those OC cows with a smaller loss of BFT produced less milk than OC cows with greater losses.
Collapse
Affiliation(s)
- R Riosa
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany; College of Medical, Veterinary and Life Sciences, School of Veterinary Medicine, University of Glasgow, Garscube Estate, Switchback Road, Bearsden G611QH, United Kingdom
| | - M H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - H M Hammon
- Research Institute for Farm Animal Biology, 18196 Dummerstorf, Germany
| | - D Süss
- Clinical Unit for Herd Health Management in Ruminants, University Clinic for Ruminants, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - M Hoelker
- Institute of Animal Science, Department of Animal Breeding and Husbandry, University of Bonn, 53175 Bonn, Germany
| | - M Drillich
- Clinical Unit for Herd Health Management in Ruminants, University Clinic for Ruminants, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - C Parys
- Evonik Operations GmbH, 63457 Hanau, Germany
| | - J Guyader
- Evonik Operations GmbH, 63457 Hanau, Germany
| | - H Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| | - M Iwersen
- Clinical Unit for Herd Health Management in Ruminants, University Clinic for Ruminants, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| |
Collapse
|
13
|
Vaspin attenuates steatosis-induced fibrosis via GRP78 receptor by targeting AMPK signaling pathway. J Physiol Biochem 2022; 78:185-197. [PMID: 35001345 DOI: 10.1007/s13105-021-00852-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease that is rapidly becoming a public health problem. An imbalance in lipid distribution to the hepatocytes and metabolism causes hepatocyte steatosis. Vaspin is a newly discovered adipokine that has been linked to a variety of metabolic disorders. The effects of vaspin on steatosis and fibrosis pathogenesis and related mechanisms are unclear. Thus, this study investigated the molecular mechanism of vaspin on hepatocyte steatosis and fibrosis. HepG2 cells were treated with 1.2 mM free fatty acid and the intracellular lipid values were measured by flow cytometry and Nile red assay. RT-qPCR was used to assess the effect of vaspin and blocking of the GRP78 receptor on the expression of lipogenesis, oxidation, uptake, and secretion of fatty acid (FA), as well as AMPK activity. In co-cultured HepG2 and LX-2 cell lines, the expression of main proteins of hepatocyte fibrosis was analyzed using Western blot analysis. In the HepG2 cell line, we discovered that vaspin increased oxidation, FA secretion and gene expression, and AMPK activity and decreased lipogenesis and FA uptake and gene expression. Western blot analysis in co-cultured HepG2 and LX-2 cell lines showed that α-SMA and TGF-β1 protein expression decreased. The data demonstrated that vaspin acts as a novel regulator of hepatocyte steatosis through the GRP78 receptor, effectively reducing hepatocyte fibrosis through AMPK activation and decreasing NF-κB gene expression.
Collapse
|
14
|
Bayram HM, Majoo FM, Ozturkcan A. Polyphenols in the prevention and treatment of non-alcoholic fatty liver disease: An update of preclinical and clinical studies. Clin Nutr ESPEN 2021; 44:1-14. [PMID: 34330452 DOI: 10.1016/j.clnesp.2021.06.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/14/2021] [Accepted: 06/17/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS The prevention and treatment of non-alcoholic fatty liver disease (NAFLD) has become one of the most urgent problems to be solved. To date, only a lifestyle modification related to diet and physical activity is considered for these patients. Polyphenols are a group of plant natural products that when regularly consumed has been related to a reduction in the risk of several metabolic disorders associated with NAFLD. In this study, we aimed to present an overview of the relationship between polyphenols and NAFLD with current approaches. METHODS We performed a comprehensive literature search for articles on polyphenols and NAFLD published in English between January 2018 to August 2020. Keywords included in this review: "Phenolic" OR "Polyphenol" AND "Non-Alcoholic Fatty Liver Disease". The editorials, communications and conference abstracts were excluded. RESULTS Different polyphenols decreased the pro-inflammatory cytokines in both serum and liver that contribute to a decrease in fatty liver dysfunction. Additionally, polyphenols may improve the regulation of adipokines and prevent hepatic steatosis. According to human clinical studies, polyphenols are promising for NAFLD patients and associated diseases that lead to NAFLD. CONCLUSION Preclinical and clinical studies suggest that various polyphenols could prevent steatosis and its progression to non-alcoholic steatohepatitis, as well as ameliorate NAFLD. However, more clinical studies are needed to confirm this hypothesis.
Collapse
Affiliation(s)
- Hatice Merve Bayram
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Istanbul Gelisim University, Avcilar, 34310, Istanbul, Turkey.
| | - Fuzail Mohammed Majoo
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Istanbul Gelisim University, Avcilar, 34310, Istanbul, Turkey.
| | - Arda Ozturkcan
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Istanbul Gelisim University, Avcilar, 34310, Istanbul, Turkey.
| |
Collapse
|
15
|
Gao Y, Zhang S, Li J, Zhao J, Xiao Q, Zhu Y, Zhang J, Huang W. Effect and mechanism of ginsenoside Rg1-regulating hepatic steatosis in HepG2 cells induced by free fatty acid. Biosci Biotechnol Biochem 2020; 84:2228-2240. [PMID: 32654591 DOI: 10.1080/09168451.2020.1793293] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ginsenoside Rg1 (G-Rg1) is a bioactive phytochemical that has been found to be beneficial for the treatment of several diseases including nonalcoholic fatty liver disease (NAFLD). But there is a lack of literature reporting the effect of G-Rg1 on lipid metabolism balance in NAFLD. We investigated the effect and mechanism of G-Rg1 on lipid metabolism in vitro. We found that G-Rg1 decreased the levels of TG, TC, and MDA, and increased activity of SOD. Results of RT-PCR and western blotting showed that supplementation with G-Rg1 downregulated the expression of PPAR γ, FABP1, FATP2/5, CD36, SREBP1 c, and FASN, while the expression of PPAR ɑ, CPT1, ACOX1, MTTP, and ApoB100 was upregulated, after induction by a free fatty acid. Taken together, we conclude that G-Rg1 inhibits lipid synthesis and lipid uptake, and enhances lipid oxidation and lipid export to reduce hepatic steatosis of HepG2 cells by regulating PPAR ɑ and PPAR γ expression.
Collapse
Affiliation(s)
- Yue Gao
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Shujun Zhang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Jiajun Li
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Jinqiu Zhao
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Qing Xiao
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Yali Zhu
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Jia Zhang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Wenxiang Huang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University , Chongqing, China
| |
Collapse
|