1
|
Yan M, Hu J, Wang L, Zhang T, Xiao Z, Yuan Y, Yue T. Metabolic profiling of abdominal subcutaneous adipose tissue reveals effects of apple polyphenols for reversing high-fat diet induced obesity in C57BL/6 J mice. Food Chem 2025; 473:143055. [PMID: 39879748 DOI: 10.1016/j.foodchem.2025.143055] [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: 10/02/2024] [Revised: 01/07/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
Apple polyphenols (APP) can reduce obesity. However, the effects of APP on abdominal subcutaneous adipose tissue (aSAT) at metabolic level were unclear. In this study, 5-week APP intervenes were conducted on 10-week high-fat diet (HFD) feeding mice with doses of 200 and 500 mg/kg b.w./day, followed by ultra-high-performance liquid chromatography-mass spectrometry based untargeted metabolomics analysis. As expected, APP obviously reversed aSAT weight and index, as well as activities of myeloperoxidase, glutathione peroxidase, superoxide dismutase and catalase. Euclidean distance between HFD and normal chow diet (NCD) group was shortened. 64 and 127 differential metabolites were found in 200 and 500 mg/kg b.w./day group, with 12 and 13 changed pathways, respectively. Specifically, APP restored glycolysis, tricarboxylic acid cycle, amino acid metabolism, and lipid metabolism as dose-dependent manner. Finally, glucose-6-phosphate, xanthine and tyrosine were selected as critical junctures. Collectively, these findings underscore the potential of APP in reversing molecular alterations in aSAT.
Collapse
Affiliation(s)
- Min Yan
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Jinpeng Hu
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Lan Wang
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Ting Zhang
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Zhengcao Xiao
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Yahong Yuan
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China.
| | - Tianli Yue
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China.
| |
Collapse
|
2
|
Luo Q, Qiu J, Chen M, Yang N, Li X, Huang S, Ma Q, Li Z, Lou D, Du Y, Chen L, Shen Q, Chen F, Li C, Qiu P. Vine tea (Ampelopsis grossedentata) ameliorates chronic alcohol-induced hepatic steatosis, oxidative stress, and inflammation via YTHDF2/PGC-1α/SIRT3 axis. Food Res Int 2025; 209:116321. [PMID: 40253212 DOI: 10.1016/j.foodres.2025.116321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 04/21/2025]
Abstract
For over a millennium, the leaves of Ampelopsis grossedentata (Hand.-Mazz.) W. T. Wang, commonly known as vine tea, have been revered as a popular tea and traditional herbal remedy, possessing antioxidant, anti-inflammatory, hepatoprotective, and antiviral properties. In recent years, the incidence of alcohol-related liver injury has been on the rise, imposing a significant public health burden worldwide. Previous studies have indicated that extracts of vine tea (AGE) can ameliorate alcoholic liver disease (ALD), yet the pharmacological mechanisms underlying this effect remain poorly understood. In this study, we first employed UPLC-Q-TOF-MS to analyze the chemical constituents of AGE. Subsequently, an ALD model was established in mice fed with Lieber-DeCarli diet, and the hepatoprotective benefits of AGE were assessed by measuring biochemical indicators and hepatic pathological changes. Moreover, a suite of bioinformatics tools, including transcriptomics, weighted gene co-expression network analysis, and single-cell data mining, were utilized to reveal that the YTHDF2/PGC-1α/SIRT3 signaling axis may be the potential mechanism by which AGE exerts its anti-ALD effects. Additionally, Western blotting and immunofluorescence staining techniques were employed to further substantiate the aforementioned mechanism. Our findings demonstrate that administration of vine tea significantly alleviated chronic ethanol-induced hepatic lipid accumulation, oxidative stress, and inflammation. Notably, knockdown of YTHDF2 partially protected the liver from ethanol-induced injury. Mechanistically, bioinformatics analysis and in vitro and in vivo experiments identified YTHDF2 as a key pharmacological target of AGE in treating ALD, acting through the downstream PGC-1α/SIRT3 pathway. In summary, in this study, we provide the first evidence that AGE mitigates ethanol-induced liver injury by inhibiting YTHDF2 and enhancing the expression of PGC-1α and SIRT3. Vine tea, as a tea food with unique medicinal value, shows significant potential and value in the treatment of ALD.
Collapse
Affiliation(s)
- Qihan Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiang Qiu
- Department of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Minxia Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Na Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyue Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qing Ma
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zongyuan Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dayong Lou
- Zhuji People's Hospital of Zhejiang Province, Shaoxing, China
| | - Yu Du
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qing Shen
- Collaborative Innovation Center of Seafood Deep Processing, Zhejiang Province Joint Key Laboratory of Aquatic Products Processing, Institute of Seafood, Zhejiang Gongshang University, Hangzhou, China.
| | - Fangming Chen
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Changyu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Ping Qiu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
3
|
Wang Y, Yang Y, Yuan M, Yang F, Zeng W, Liu J, Zhan C. MC-LR induced apoptosis in human embryonic kidney (HEK293) cells through activation of TNF-R1/RIPK1 pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:339-348. [PMID: 39705070 DOI: 10.1080/15287394.2024.2439532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
In recent years, the outbreak of cyanobacterial blooms has become increasingly frequent. Microcystin-LR (MC-LR), a metabolite of cyanobacteria, poses a significant threat to the ecosystem and human health. Several studies have demonstrated that MC-LR might induce renal cell apoptosis, as a consequence of tissue damage. However, the molecular mechanisms underlying MC-LR-initiated renal injury remain to be determined. This investigation aimed to determine the role of apoptosis in MC-LR-induced kidney damage and its potential underlying mechanisms using the human embryonic kidney (HEK293) cell line. The results of TUNEL and immunofluorescence assays indicated that MC-LR induced increased apoptosis in HEK293 cells. Compared to control, the mRNA expression levels of RIPK1, caspase-8, and TNF-α were elevated following incubation with MC-LR, while the mRNA expression level of Bcl-2/Bax was decreased. The protein levels of RIPK1, TNF-R1, and caspase-8 were elevated in the MC-LR-treated HEK293 cells. Data demonstrated that MC-LR induced renal cell apoptosis through activation of the TNF-R1/RIPK1 pathway, providing new insights into understanding the toxic mechanisms attributed to MC-LR.
Collapse
Affiliation(s)
- Yaqi Wang
- Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, School of Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yue Yang
- Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, School of Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Mei Yuan
- The Second Affiliated Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Fei Yang
- Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, School of Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wen Zeng
- The Department of Public Health, The Central Hospital of Shaoyang, Shaoyang, China
| | - Jun Liu
- Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, School of Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chunhua Zhan
- Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, School of Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Department of Public Health, The Central Hospital of Shaoyang, Shaoyang, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, Hunan, China
| |
Collapse
|
4
|
Liu L, Jia R, Chen W, Chen W, Wang X, Guo Z. The lotus seed starch-EGCG complex modulates obesity in C57BL/6J mice through the regulation of the gut microbiota. Int J Biol Macromol 2025; 310:143256. [PMID: 40250649 DOI: 10.1016/j.ijbiomac.2025.143256] [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: 01/30/2025] [Revised: 03/23/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
The starch-polyphenol complex, identified as RS5-resistant starch, has been shown to regulate the gut environment and inhibit metabolic diseases, including obesity. In a study with C57BL/6 obese mice fed LSE, potential anti-obesity effects were demonstrated through physiological and biochemical assessments, gut microbiota analysis, and mechanistic insights. The study showed that LSE reduced mice body weight, serum total cholesterol, and triglycerides (P < 0.05). Serum inflammatory markers (TNF-α, IL-6, IL-1β) and LPS levels were significantly decreased, while glucose tolerance (AUC reduced by 29.29 %) and insulin sensitivity (AUC reduced by 31.79 %) were improved. Histological analysis indicated reduction in adipocyte size and attenuation of hepatic steatosis. Gut microbiota profiling demonstrated LSE increased beneficial bacteria genera Faecalibacterium, Bifidobacterium, and Akkermansia. This correlated with enhanced SCFA production (acetate 41.53 %, propionate 45.52 %, butyrate 57.49 % increase). These findings demonstrate that LSE exerts anti-obesity effects through modulation of the gut microbiota-SCFA-metabolic axis, supporting starch-polyphenol complexes as functional food candidates.
Collapse
Affiliation(s)
- Lu Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ru Jia
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wenjing Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wenyu Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiaoying Wang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Zebin Guo
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
5
|
Liu B, Yao Z, Song L, Sun C, Shen C, Cheng F, Cheng Z, Zhang R, Liu R. Vitexin alleviates lipid metabolism disorders and hepatic injury in obese mice through the PI3K/AKT/mTOR/SREBP-1c pathway. Eur J Med Chem 2025; 287:117379. [PMID: 39947052 DOI: 10.1016/j.ejmech.2025.117379] [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: 11/21/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/24/2025]
Abstract
Obesity is recognized as a metabolic disorder, and its treatment and management pose ongoing challenges worldwide. Hawthorn, a traditional Chinese herb used to alleviate digestive issues and reduce blood lipid levels, has unclear mechanisms of action regarding its active components in the treatment of obesity. This study investigated the anti-obesity effects of vitexin, a major flavonoid compound found in hawthorn, in high-fat diet (HFD)-induced C57BL/6 mice. The results demonstrated that vitexin significantly reduced body weight, liver weight, blood lipid levels, and inflammatory markers in obese mice, while also inhibiting hepatic lipid accumulation. Mechanistic studies revealed that vitexin likely suppresses adipogenesis by modulating the PI3K-AKT signaling pathway, as evidenced by reduced expression of PI3K, phosphorylated AKT, phosphorylated mTOR, and SREBP-1c in the livers of vitexin-treated obese mice. Additionally, vitexin inhibited NFκB expression by regulating IκBα phosphorylation, thereby alleviating obesity-induced liver injury. These findings suggest that vitexin may be the primary active component in hawthorn responsible for reducing blood lipid levels, highlighting its potential in the treatment of obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
- Bo Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ziqing Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lin Song
- Department of Pharmacy, Children' S Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Chen Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Changhong Shen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zefang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ruoqi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Rong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
6
|
Zhang Y, Chang M, Xue Q, Wang H, Liu Y, Wei H, Li J. Intermittent cold stimulation acclimates broilers to acute cold stress by affecting cardiac lipid metabolism. Anim Biosci 2025; 38:775-787. [PMID: 39483027 PMCID: PMC11917442 DOI: 10.5713/ab.24.0389] [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: 06/12/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE This study aimed to investigate whether intermittent cold stimulation can induce adaptation in broilers to acute cold stress (ACS) by regulating the lipid metabolism of hearts. METHODS CS0 were kept at normal rearing temperature, while CS3 and CS5 were exposed to 3°C for 3 and 5 hours, respectively, on alternate days lower than CS0 from 15d to 35d. On 50d, broilers in three groups were exposed to ACS at 10°C for 12 hours (Y12). The levels of corticosterone (CORT) and liothyronine (T3), mRNA and protein levels of heart adenosine monophosphate (AMP)-activated protein kinase/mammalian target of rapamycin (AMPK/mTOR) pathway genes were assessed at 36 d, 50 d and Y12. RESULTS At 36d, mRNA levels of AMPKα, acyl-CoA oxidase (ACO), mTOR, sterolregulatory element binding protein (SREBP), stearoyl-coA desaturase (SCD), acetyl-coA carboxylase (ACC), fatty acid synthase (FAS) and protein level of peroxisome proliferatorsactivated receptor α (PPARα) in CS3 and CS5 were significantly lower than those in CS0 (p<0.05). At 50d, compared to CS0, mRNA levels of PPARα, carnitine palmitoyltransferase1 (CPT1), ACO, tuberous sclerosis complex (TSC), SREBP and SCD, as well as protein levels of p-AMPKα/AMPKα, PPARα and SREBP were significantly increased in CS5 (p<0.05). At Y12, the levels of T3 in CS3 and CS5 were significantly higher than those in CS0 (p<0.05), mRNA levels of CPT1, ACO, SREBP, SCD and protein levels of p-AMPKα/AMPKα, SREBP, and FAS were significantly higher in CS5 than in CS0 and CS3 (p<0.05). However, compared to 50d, at Y12, mRNA levels of AMPKα, CPT1 and ACO in CS3 and CS5 significantly decreased (p<0.05), while protein levels of p-AMPKα/AMPKα significantly increased (p<0.05). CONCLUSION This study suggested that intermittent cold stimulation at 3°C lower than normal rearing temperature for 5h could help broilers adapt to the ACS by promoting heart lipid metabolism.
Collapse
Affiliation(s)
- Yong Zhang
- College of Life Science, Northeast Agricultural University, Harbin 150030,
China
| | - Minghang Chang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030,
China
| | - Qiang Xue
- College of Life Science, Northeast Agricultural University, Harbin 150030,
China
| | - Hongyu Wang
- College of Life Science, Northeast Agricultural University, Harbin 150030,
China
| | - Yuanyuan Liu
- College of Life Science, Northeast Agricultural University, Harbin 150030,
China
| | - Haidong Wei
- College of Life Science, Northeast Agricultural University, Harbin 150030,
China
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, Harbin 150030,
China
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030,
China
| |
Collapse
|
7
|
Song W, Zhu L, Yang C, Su K, Miao Y, Hu J, Chen B, Li L, Cui X, Luo Y, Sheng Q, Yue T. Ergosterol originated from Auricularia auricula attenuates high fat diet-induced obesity and cognitive impairment in mice. Food Funct 2025; 16:2780-2792. [PMID: 40084554 DOI: 10.1039/d4fo04817b] [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: 03/16/2025]
Abstract
Excessive intake of a high fat diet (HFD) leads to accumulation of fat and obesity. Ergosterol (ERG) is a characteristic sterol of fungi with various bioactive functions; however, there are few studies on the ERG function of ameliorating obesity and following cognitive impairment. It was previously found that Zhashui Auricularia auricula (AA) is rich in ERG; therefore it was selected to enrich ERG through the intervention of exogenous inducers of rice bran oil (RBO), methyl jasmonate (Me JA) and salicylic acid (SA). The accumulated ERG was used to investigate alleviative effects on mouse obesity and cognitive impairment. According to LEfSe analysis of intestinal flora species, ERG reduced the abundance of obesity or inflammation-related intestinal microbial genera, while increasing the relative abundance of beneficial bacteria. The ERG sourced from AA significantly ameliorated HFD-induced mouse obesity by reducing lipid levels and liver oxidative stress, recovering memory and learning abilities of the mice by restoring the hippocampus function and downregulating inflammatory factors.
Collapse
Affiliation(s)
- Wei Song
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| | - Lina Zhu
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| | - Chunyan Yang
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| | - Kaixin Su
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| | - Yaxing Miao
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| | - Jinhong Hu
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| | - Bing Chen
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| | - Lingling Li
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| | - Xiaole Cui
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| | - Yane Luo
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| | - Qinglin Sheng
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| | - Tianli Yue
- College of Food Science and Technology, Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Research Center of Food Safety Risk Assessment and Control, Northwest University, No. 229, North Taibai Road, Beilin District, Xi'an 710069, China.
| |
Collapse
|
8
|
Zhang C, Shu X, Yin C, Hu S, Liu P. The role of the mTOR pathway in breast cancer stem cells (BCSCs): mechanisms and therapeutic potentials. Stem Cell Res Ther 2025; 16:156. [PMID: 40158191 PMCID: PMC11954216 DOI: 10.1186/s13287-025-04218-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/11/2025] [Indexed: 04/01/2025] Open
Abstract
Breast cancer remains the most frequently diagnosed cancer globally, exerting a profound impact on women's health and healthcare systems. Central to its pathogenesis and therapeutic resistance are breast cancer stem cells (BCSCs), which possess unique properties such as self-renewal, differentiation, and resistance to conventional therapies, contributing to tumor initiation, metastasis, and recurrence. This comprehensive review elucidates the pivotal role of the mechanistic target of rapamycin (mTOR) pathway in regulating BCSCs and its implications for breast cancer progression and treatment resistance. We explore the cellular mechanisms by which mTOR influences metastasis, metabolism, autophagy, and ferroptosis in BCSCs, highlighting its contribution to epithelial-to-mesenchymal transition (EMT), metabolic reprogramming, and survival under therapeutic stress. On a molecular level, mTOR interacts with key signaling pathways including PI3K/Akt, Notch, IGF-1R, AMPK, and TGF-β, as well as regulatory proteins and non-coding RNAs, orchestrating a complex network that sustains BCSC properties and mediates chemoresistance and radioresistance. The review further examines various therapeutic strategies targeting the mTOR pathway in BCSCs, encompassing selective PI3K/Akt/mTOR inhibitors, monoclonal antibodies, natural products, and innovative approaches such as nanoparticle-mediated drug delivery. Clinical trials investigating mTOR inhibitors like sirolimus and combination therapies with agents such as everolimus and trastuzumab are discussed, underscoring their potential in eradicating BCSCs and improving patient outcomes. Additionally, natural compounds and repurposed drugs offer promising adjunctive therapies by modulating mTOR activity and targeting BCSC-specific vulnerabilities. In conclusion, targeting the mTOR pathway presents a viable and promising avenue for enhancing breast cancer treatment efficacy by effectively eliminating BCSCs, reducing tumor recurrence, and improving overall patient survival. Continued research and clinical validation of mTOR-targeted therapies are essential to translate these insights into effective clinical interventions, ultimately advancing personalized cancer management and therapeutic outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xu Shu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Chuanzheng Yin
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Shaobo Hu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Pian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
| |
Collapse
|
9
|
Alotaibi G, Alkhammash A. Pharmacological landscape of endoplasmic reticulum stress: Uncovering therapeutic avenues for metabolic diseases. Eur J Pharmacol 2025; 998:177509. [PMID: 40089262 DOI: 10.1016/j.ejphar.2025.177509] [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: 11/02/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
The endoplasmic reticulum (ER) plays a fundamental role in maintaining cellular homeostasis by ensuring proper protein folding, lipid metabolism, and calcium regulation. However, disruptions to ER function, known as ER stress, activate the unfolded protein response (UPR) to restore balance. Chronic or unresolved ER stress contributes to metabolic dysfunctions, including insulin resistance, non-alcoholic fatty liver disease (NAFLD), and neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease. Recent studies have also highlighted the importance of mitochondria-ER contact sites (MERCs) and ER-associated inflammation in disease progression. This review explores the current pharmacological landscape targeting ER stress, focusing on therapeutic strategies for rare metabolic and neurodegenerative diseases. It examines small molecules such as tauroursodeoxycholic acid (TUDCA) and 4-phenylbutyric acid (4-PBA), repurposed drugs like 17-AAG (17-N-allylamino-17demethoxygeldanamycin (tanespimycin)) and berberine, and phytochemicals such as resveratrol and hesperidin. Additionally, it discusses emerging therapeutic areas, including soluble epoxide hydrolase (sEH) inhibitors for metabolic disorders and MERCs modulation for neurological diseases. The review emphasizes challenges in translating these therapies to clinical applications, such as toxicity, off-target effects, limited bioavailability, and the lack of large-scale randomized controlled trials (RCTs). It also highlights the potential of personalized medicine approaches and pharmacogenomics in optimizing ER stress-targeting therapies.
Collapse
Affiliation(s)
- Ghallab Alotaibi
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra, 11961, Saudi Arabia.
| | - Abdullah Alkhammash
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra, 11961, Saudi Arabia.
| |
Collapse
|
10
|
Bian Y, Wu H, Jiang W, Kong X, Xiong Y, Zeng L, Zhang F, Song J, Wang C, Yang Y, Zhang X, Zhang Y, Pang P, Duo T, Wang Z, Pan T, Yang B. Anti-b diminishes hyperlipidaemia and hepatic steatosis in hamsters and mice by suppressing the mTOR/PPARγ and mTOR/SREBP1 signalling pathways. Br J Pharmacol 2025; 182:1254-1272. [PMID: 39614407 DOI: 10.1111/bph.17397] [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: 06/19/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND AND PURPOSE As a chronic metabolic syndrome, hyperlipidaemia is manifested as aberrantly elevated cholesterol and triglyceride (TG) levels, primarily attributed to disorders in lipid metabolism. Despite the promising outlook for hyperlipidaemia treatment, the need persists for the development of lipid-lowering agents with heightened efficiency and minimal toxicity. This investigation aims to elucidate the lipid-lowering effects and potential pharmacodynamic mechanisms of Anti-b, a novel low MW compound. EXPERIMENTAL APPROACH We employed high-fat diet (HFD) in hamsters and mice or oleic acid (OA) in cultures of HepG2 cells and LO2 cells to induce hyperlipidaemia models. We administered Anti-b to assess its therapeutic effects on dyslipidaemia and hepatic steatosis. We used western blotting, RNA sequencing, GO and KEGG analysis, oil red O staining, along with molecular docking and molecular dynamics simulation to elucidate the mechanisms underlying the effects of Anti-b. KEY RESULTS Anti-b exhibited a substantial reduction in HFD-induced elevation of blood lipids, liver weight to body weight ratio, liver diameter and hepatic fat accumulation. Moreover, Anti-b demonstrated therapeutic effects in alleviating total cholesterol (TC), TG levels, and lipid accumulation derived from OA in HepG2 cells and LO2 cells. Mechanistically, Anti-b selectively bound to the mTOR kinase protein and increased mTOR thermal stability, resulting in downregulation of phosphorylation level. Notably, Anti-b exerted anti-hyperlipidaemia effects by modulating PPARγ and SREBP1 signalling pathways and reducing the expression level of mSREBP1 and PPARγ proteins. CONCLUSION AND IMPLICATIONS In conclusion, our study has provided initial data of a novel low MW compound, Anti-b, designed and synthesised to target mTOR protein directly. Our results indicate that Anti-b may represent a novel class of drugs for the treatment of hyperlipidemia and hepatic steatosis.
Collapse
Affiliation(s)
- Yu Bian
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Han Wu
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Weitao Jiang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xue Kong
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yuting Xiong
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Linghua Zeng
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Feng Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jinglun Song
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Chunlei Wang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yang Yang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xinyue Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yuning Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ping Pang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Tianqi Duo
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zhuo Wang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Tengfei Pan
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Baofeng Yang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
11
|
Gupta S, Afzal M, Agrawal N, Almalki WH, Rana M, Gangola S, Chinni SV, Kumar K B, Ali H, Singh SK, Jha SK, Gupta G. Harnessing the FOXO-SIRT1 axis: insights into cellular stress, metabolism, and aging. Biogerontology 2025; 26:65. [PMID: 40011269 DOI: 10.1007/s10522-025-10207-0] [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: 02/02/2025] [Accepted: 02/15/2025] [Indexed: 02/28/2025]
Abstract
Aging and metabolic disorders share intricate molecular pathways, with the Forkhead box O (FOXO)- Sirtuin 1 (SIRT1) axis emerging as a pivotal regulator of cellular stress adaptation, metabolic homeostasis, and longevity. This axis integrates nutrient signaling with oxidative stress defence, modulating glucose and lipid metabolism, mitochondrial function, and autophagy to maintain cellular stability. FOXO transcription factors, regulated by SIRT1 deacetylation, enhance antioxidant defence mechanisms, activating genes such as superoxide dismutase (SOD) and catalase, thereby counteracting oxidative stress and metabolic dysregulation. Recent evidence highlights the dynamic role of reactive oxygen species (ROS) as secondary messengers in redox signaling, influencing FOXO-SIRT1 activity in metabolic adaptation. Additionally, key redox-sensitive regulators such as nuclear factor erythroid 2-related factor 2 (Nrf2) and Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) interact with this pathway, orchestrating mitochondrial biogenesis and adaptive stress responses. Pharmacological interventions, including alpha-lipoic acid (ALA), resveratrol, curcumin and NAD+ precursors, exhibit therapeutic potential by enhancing insulin sensitivity, reducing oxidative burden, and restoring metabolic balance. This review synthesizes current advancements in FOXO-SIRT1 regulation, its emerging role in redox homeostasis, and its therapeutic relevance, offering insights into future strategies for combating metabolic dysfunction and aging-related diseases.
Collapse
Affiliation(s)
- Saurabh Gupta
- Department of Pharmacology, Chameli Devi Institute of Pharmacy, Khandwa Road, Village Umrikheda, Near Tollbooth, Indore, Madhya Pradesh, 452020, India
| | - Muhammad Afzal
- Pharmacy Program, Department of Pharmaceutical Sciences, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gangola
- Department of Microbiology, Graphic Era Deemed to be University, Dehradun, 248002, India
| | - Suresh V Chinni
- Department of Biochemistry, Faculty of Medicine, Bioscience, and Nursing, MAHSA University, 42610, Jenjarom, Selangor, Malaysia
| | - Benod Kumar K
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, New Delhi, India
- Centre for Himalayan Studies, University of Delhi, Delhi, 110007, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| |
Collapse
|
12
|
Cheng X, Hemmati S, Pirhayati M, Zangeneh MM, Veisi H. Decoration of copper nanoparticles (Cu 2O NPs) over chitosan-guar gum: Its application in the Sonogashira cross-coupling reactions and treatment of human lung adenocarcinoma. Int J Biol Macromol 2025; 305:141122. [PMID: 39965696 DOI: 10.1016/j.ijbiomac.2025.141122] [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: 12/01/2024] [Revised: 02/02/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
This study outlines the sustainable synthesis of hybrid biopolymer hydrogels supported with octahedral Cu2O nanoparticles (NPs), alongside their biological assessments and characterizations. A composite hydrogel made of chitosan and guar gum (CS-GG) was employed as a template for the environmentally friendly synthesis of nanoparticles. Leveraging their electron-rich functional groups, the biopolymers acted as stabilizing agents for the Cu2O NPs and as green reductants, facilitating the reduction of copper ions. The material's physicochemical properties were thoroughly examined using advanced techniques, such as X-ray diffraction (XRD), Field-Emission Scanning Electron Microscopes (FE-SEM), Eneregy Dispersive X-ray Electron Spectroscopy (EDX), Fourier Transformed Infrared Spectroscopy (FT-IR), Transmission Electron Microscopy (TEM) and ICP-OES. The resulting CS-GG/Cu2O NPs nanocomposite was investigated as a reusable heterogeneous nanocatalyst, demonstrating its efficiency in the phosphine-free, palladium-free, and ligand-free synthesis of various stilbene derivatives with high yields through the Sonogashira coupling reaction. The catalyst showed no significant reduction in activity after being reused seven times consecutively. The cytotoxic effects of the CS-GG/Cu2O NPs nanocomposite on NCI-H661 lung cancer cells and normal cells (HUVEC) were assessed over 48 h using MTT assay. The cancer cell's viability decreased after exposure to the CS-GG/Cu2O NPs, with an IC50 value of 82 μg/mL. The CS-GG/Cu2O NPs nanocomposite controls the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) system, which in turn impacts apoptosis and cell proliferation in NCI-H661 cells, according to a detailed examination of the mTOR pathway. The pathway could act a role in the cell cycle inhibition and apoptosis induced by the CS-GG/Cu2O NPs nanocomposite. The CS-GG/Cu2O NPs nanocomposite could be a useful natural anti-cancer agent for the treatment of lung cancer.
Collapse
Affiliation(s)
- Xiongtao Cheng
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Saba Hemmati
- Department of Chemistry, Payame Noor University, Tehran, Iran
| | - Mozhgan Pirhayati
- Department of Applied Chemistry, Faculty of Science, Malayer University, Malayer, Iran.
| | - Mohamad Mehdi Zangeneh
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Hojat Veisi
- Department of Chemistry, Payame Noor University, Tehran, Iran.
| |
Collapse
|
13
|
Salazar-Barrantes KA, Abdala-Saiz A, Vega-Baudrit JR, Navarro-Hoyos M, Araya-Sibaja AM. Formulation and evaluation of atorvastatin calcium trihydrate Form I tablets. PLoS One 2025; 20:e0317407. [PMID: 39946379 PMCID: PMC11825022 DOI: 10.1371/journal.pone.0317407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 12/28/2024] [Indexed: 02/16/2025] Open
Abstract
Solid forms transformations and new crystal structures of an active pharmaceutical ingredient (API) can occur due to various manufacturing process conditions, especially if the drug substance is formulated as a hydrate. The conversion between hydrate and anhydrate forms caused by changes in temperature and humidity must be evaluated because of the risk of dehydration and phase transitions during the manufacturing process. Differences in physicochemical, mechanical, and rheological properties have been observed between solid forms of the same API that can cause manufacturing and product-related issues. Atorvastatin calcium trihydrate (ACT) is a synthetic lipid-lowering agent that was discovered during Lipitor® (its anhydrous form) Phase 3 clinical trials after passing Phase I and II. This case highlights the importance of routinely performing solid form screenings because of the probability of finding new solid forms during the development and scale-up process. Therefore, in this contribution, ACT tablet formulation was performed and evaluated starting from the compatibility of 1:1 proportions of drug and the excipients microcrystalline cellulose 101 (MCC 101), calcium carbonate, lactose monohydrate, croscarmellose sodium, hydroxypropyl cellulose, magnesium stearate, and polysorbate 80. Then, 40 mg ACT tablets were prepared on a small pilot scale, and manufacturing process assessment was conducted by sampling process stages selected as critically prone to solid forms formation or phase transition. Final product quality was evaluated regarding weight variation, hardness, disintegration, dissolution, and assay tests. Powder X-ray diffraction (PXRD), Fourier transform infrared spectroscopy (FT-IR), differential scanning calorimetry (DSC), and thermogravimetric analysis (TGA) were applied to solid state evaluation. The starting raw material was confirmed to be ACT Form I. From the preformulation studies, PXRD, FT-IR and TGA analyses showed no interactions between ACT and excipients, while DSC results revealed a physical interaction with MCC 101, not considered an incompatibility. The effect of the tablet manufacturing process was achieved by amorphization, while some ACT long-range crystalline structure remained, as confirmed by PXRD, FT-IR and DSC. However, the tablets' quality parameters were found to be within the acceptable range of both the pharmacopeia guidelines and manufacturer parameters regarding weight variation, hardness, disintegration, dissolution, and assay tests.
Collapse
Affiliation(s)
- Karen Andrea Salazar-Barrantes
- Escuela de Química, Universidad de Costa Rica, San José, Costa Rica
- Laboratorio Nacional de Nanotecnología LANOTEC-CeNAT-CONARE, San José, Costa Rica
| | - Ariadna Abdala-Saiz
- Departamento de Investigación y Desarrollo, Calox de Costa Rica, San José, Costa Rica
| | | | - Mirtha Navarro-Hoyos
- Laboratorio BIODESS, Escuela de Química, Universidad de Costa Rica, San José, Costa Rica
- Chemistry Department, Georgetown University, Washington, D.C., United States of America
| | | |
Collapse
|
14
|
Saadh MJ, Abosaoda MK, Baldaniya L, Kalia R, Arya R, Mishra S, Chauhan AS, Kumar A, Alizadeh M. The Effects of Chia Seed (Salvia hispanica L.) Consumption on Blood Pressure and Body Composition in Adults: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Clin Ther 2025; 47:168-175. [PMID: 39672763 DOI: 10.1016/j.clinthera.2024.11.012] [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: 10/16/2024] [Revised: 11/01/2024] [Accepted: 11/09/2024] [Indexed: 12/15/2024]
Abstract
PURPOSE Growing evidence has suggested that the consumption of chia seed can decrease blood pressure and obesity in adults. However, even studies have reported uncertain findings. The current meta-analysis aimed to assess the findings of randomized controlled trials (RCTs) on the efficacy of chia seed supplementation on blood pressure (systolic blood pressure [SBP], diastolic blood pressure [DBP]) and body composition (waist circumference [WC], weight, body mass index [BMI]) in adults. METHODS A systematic search of the literature was carried out in the PubMed, Web of Knowledge, Scopus, Cochrane Central Library, and EMBASE from inception up to October 2024. Data were extracted and analyzed using a random-effects model, and reported as weighted mean differences (WMD) with 95% confidence intervals (CI). FINDINGS A total of eight RCTs involving 372 participants were included in the meta-analysis. The results showed that chia consumption significantly reduced DBP (WMD: -7.49 mmHg; 95% CI: -9.64, -5.34; P < 0.001) and SBP (WMD: -5.61 mmHg; 95% CI: -8.77, -2.44; P = 0.001). Moreover, consuming chia seeds was linked to a notable decrease in WC (WMD: -1.46 cm; 95% CI: -2.68, -0.25; P = 0.01), but it had no significant effect on, BMI (WMD: -0.31 kg/m2; 95% CI: - 0.96, 0.34; P = 0.34) and weight (WMD: 0.09 kg; 95% CI: -0.76, 0.93; P = 0.84). IMPLICATIONS Chia consumption can significantly reduce SBP, DBP, and WC in adults, but no significant impact was showed on BMI and weight. To verify these results, more studies involving a greater number of participants are required.
Collapse
Affiliation(s)
| | - Munthar Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq; College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; College of Pharmacy, The Islamic University of Babylon, Babylon, Iraq
| | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences Marwadi University, Rajkot, Gujarat, India
| | - Rishiv Kalia
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - Shivang Mishra
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, India
| | - Ashish Singh Chauhan
- Division of Research and Innovation, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after the First President of Russia Boris Yeltsin, Ekaterinburg, Russia; Department of Technical Sciences, Western Caspian University, Baku, Azerbaijan; Department of Mechanical Engineering, Karpagam Academy of Higher Education, Coimbatore, 641021, India
| | - Mohamad Alizadeh
- Department of Medical, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Miao M, Zhang XY, Yu HX, Shi SR, Ma CN, Guo SD. Mechanisms underlying the effects of the conditional knockdown of hepatic PCSK9 in attenuating lipopolysaccharide-induced acute liver inflammation. Int J Biol Macromol 2025; 291:139066. [PMID: 39716700 DOI: 10.1016/j.ijbiomac.2024.139066] [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: 09/26/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/25/2024]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is known to promote hyperlipidemia primarily by inducing the degradation of the low-density lipoprotein receptor. Notably, recent studies have demonstrated that PCSK9 promotes inflammation in the vascular system, however, the roles of PCSK9 in hepatic inflammation remain unclear. As PCSK9 is primarily expressed in the liver, this study aimed to elucidate the roles of PCSK9 and the underlying mechanisms in lipopolysaccharide (LPS)-challenged hepatocytes. Next-generation sequencing analysis revealed that the conditional knockdown of hepatic PCSK9 significantly reduced the plasma levels of total cholesterol and modulated the expression of hundreds of genes. Importantly, PCSK9 knockdown attenuated hepatic inflammation by suppressing several signaling pathways related to inflammation, including the Toll-like receptor, mitogen-activated protein kinase (MAPK), and phosphoinositide-3 kinase/protein kinase B pathways. This subsequently altered the expression of nuclear factor kappa-B and activator protein 1. The underlying mechanisms were further confirmed by in vitro studies using primary hepatocytes and HepG2 cells, with a p38-MAPK inhibitor, a PCSK9 antibody, and two siRNAs against PCSK9. This study is the first to report that hepatic PCSK9 knockdown ameliorates LPS-induced acute liver inflammation via modulating multiple signaling pathways, thereby suggesting therapeutic potential of PCSK9 inhibitors in treating diseases related to hepatic inflammation.
Collapse
Affiliation(s)
- Miao Miao
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Xue-Ying Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Hai-Xin Yu
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shan-Rui Shi
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Chao-Nan Ma
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
16
|
Yang M, Zheng S, Zeng W, Zhan C, Yang Y, Yang F. Chronic exposure to low-dose MC-LR induces ileal inflammation in mice through the PI3K/AKT/mTOR pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025:1-9. [PMID: 39790025 DOI: 10.1080/15287394.2024.2441294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The global phenomenon of cyanobacterial bloom pollution is spreading globally due to climate change and eutrophication. It is well established that harmful cyanobacteria produce a wide range of toxins including microcystin-LR (MC-LR), a cyclic heptapeptide toxin known to damage various organs. The intestinal tract is the main site of MC-LR absorption and one of the targets susceptible to toxicity. Currently, studies on the enterotoxic effects of MC-LR predominantly focused on the colorectum, with limited investigations addressing the impact of microcystins on the small intestine. Therefore, the aim of our study was to examine the impact of chronic 9-month exposure of mice to low-dose 120 μg/L MC-LR in drinking water on ileal inflammation and potential mechanisms underlying these effects. Our findings showed that in mice chronically administered with low-dose MC-LR disorganized intestinal epithelial cells, lymphocytic infiltration and disturbed crypt arrangement were detected. The results of qPCR and Western blot demonstrated that, in comparison to control, the mRNA expression levels of pro-inflammatory factors IL-6, IL-17, IL-18, and IFN-γ were markedly elevated in the ileal tissue of mice treated with MC-LR, associated with significant increases in protein expression levels of p-PI3K, p-AKT, and p-mTOR. Taken together, evidence indicates that MC-LR induces ileal inflammation and histopathological damage involved activation of the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Mingjie Yang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Shuilin Zheng
- Changsha Center for Disease Control and Prevention, Changsha, China
| | - Wen Zeng
- The Department of Public Health, The Central Hospital of Shaoyang, Shaoyang, China
| | - Chunhua Zhan
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Yue Yang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- The Department of Public Health, The Central Hospital of Shaoyang, Shaoyang, China
| | - Fei Yang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
- Nuclear Medicine Department, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
17
|
Gataa IS, Abdullah Z, González Cabrera MV, S R, Verma S, Arora I, Monsi M, Muzammil K, Zainul R. Impact of whey protein on lipid profiles: A systematic review and meta-analysis. Nutr Metab Cardiovasc Dis 2025:103858. [PMID: 39939251 DOI: 10.1016/j.numecd.2025.103858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/26/2024] [Accepted: 01/03/2025] [Indexed: 02/14/2025]
Abstract
AIMS This research delved into a comprehensive examination and detailed analysis of the effects of whey protein (WP) supplementation on lipid profile in adults. DATA SYNTHESIS Data used in this research was obtained from diverse clinical trials. Thorough searches were carried out on multiple electronic platforms including PubMed, Embase, Web of Science, Scopus, Google Scholar, and the Cochrane Library from their inception until May 2024. Random effects models were assessed and pooled data were determined as weighted mean differences (WMDs) with a 95 % confidence interval (CI). RESULTS Overall, 20 randomized clinical trials (n = 1638 participants) met our inclusion criteria. The current meta-analysis demonstrates a significant reduction in TG (WMD: -12.21 mg/dL; %95CI: -20.16, -4.26; P = 0.003). Pooled analysis of 19 studies on HDL-c indicated a significant increase (WMD: 2.59 mg/dL; %95CI: 1.11, 4.07; P = 0.001). CONCLUSIONS We found that WP intake can improve TG and HDL-c significantly without significant effects on TC, and LDL-c levels. However, future well-designed with long duration RCTs is required on diverse populations to understand better the effects of these natural compounds and their constituents on lipid profile in adults.
Collapse
Affiliation(s)
| | - Z Abdullah
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University, Najaf, Iraq; Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| | - María Verónica González Cabrera
- Facultad de Ciencias Pecuarias, Escuela Superior Politécnica de Chimborazo (ESPOCH), Panamericana Sur km. 1½, Riobamba, 060155, Ecuador
| | - RenukaJyothi S
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Seema Verma
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Isha Arora
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Punjab, Mohali, 140307, India
| | - Mekha Monsi
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University, Abha, 62561, Saudi Arabia
| | - Rahadian Zainul
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Negeri Padang, Padang, Indonesia; Center for Advanced Material Processing, Artificial Intelligence, and Biophysics Informatics (CAMPBIOTICS), Universitas Negeri Padang, Padang, Indonesia; Researcher Fellow at Asia Pacific University of Technology and Innovations (APU), Malaysia.
| |
Collapse
|
18
|
Namini MS, Beheshtizadeh N, Ebrahimi-Barough S, Ai J. Human endometrial stem cell-derived small extracellular vesicles enhance neurite outgrowth and peripheral nerve regeneration through activating the PI3K/AKT signaling pathway. J Transl Med 2025; 23:6. [PMID: 39754260 PMCID: PMC11699817 DOI: 10.1186/s12967-024-06048-z] [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: 08/02/2024] [Accepted: 12/25/2024] [Indexed: 01/06/2025] Open
Abstract
Nowadays, extracellular vesicles (EVs) such as exosomes participate in cell-cell communication and gain attention as a new approach for cell-free therapies. Recently, various studies have demonstrated the therapeutic ability of exosomes, while the biological effect of human endometrial stem cell (hEnSC)-derived small EVs such as exosomes is still unclear. Herein, we obtained small EVs from hEnSC and indicated that these small EVs activate the vital cell signaling pathway and progress neurite outgrowth in PC-12 cell lines. For this purpose, hEnSC-derived small EVs were extracted by ultracentrifuge and characterized by DLS, SEM, TEM, and western blot. Also, dil-staining of hEnSC-derived small EVs was done to determine the penetration of hEnSC-derived small EVs into PC12 cells. The MTT assay, scratch assay, and western blot assay were applied to PC12 cells that were exposed to different concentrations of small EVs (0, 50, 100, and 150 µg/ml). Our results demonstrated that small EVs significantly increased neurite outgrowth, proliferation, and migration in PC12 cells in a dose-dependent manner. Moreover, the analysis of western blots showed increased expression of the PI3k/AKT signaling pathway in PC12 cells exposed to hEnSC-derived small EVs in a dose-dependent manner. Also, the results of this study indicated that hEnSC-derived small EVs can enhance cell proliferation and migration and promote neural outgrowth by activating the PI3k/AKT signaling pathway. Accordingly, hEnSC-derived small EVs became an effective strategy for cell-free therapies. Altogether, these positive effects make hEnSC-derived small EVs a new efficient approach in regenerative medicine, especially for the cure of neural injury.
Collapse
Affiliation(s)
- Mojdeh Salehi Namini
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Beheshtizadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Gupta J, Abosaoda MK, Shukla M, Ballal S, Kumar A, Chahar M, Saini S, Kapila I, Hadpoori A. Effect of soluble fiber supplementation on lipid parameters in subjects with type 2 diabetes: A systematic review and meta-analysis of randomized controlled trials. Prostaglandins Other Lipid Mediat 2025; 176:106939. [PMID: 39689417 DOI: 10.1016/j.prostaglandins.2024.106939] [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: 10/09/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND There is no consensus in the existing literature regarding the effect of soluble fiber on the lipid profile of patients with type 2 diabetes mellitus (T2DM). This systematic review and meta-analysis of randomized controlled trials aimed to assess the effect of soluble fiber on triglycerides (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) in T2DM patients. METHODS PubMed/MEDLINE, Scopus, and ISI Web of sciences were searched for RCTs up to 4 May 2024. Data from RCTs were pooled using the generic inverse variance method and expressed as weighted mean differences (WMD) with 95 % confidence interval (CIs). RESULTS Pooled data from 38 RCTs reporting patient outcomes were evaluated for mean effects. Results indicated that soluble fiber significantly altered TG (WMD: -16.97 mg/dL, 95 % CI: -29.16 to -4.78, P = 0.021), HDL-C (WMD: 1.74 mg/dL, 95 % CI: 1.02-2.46, P < 0.001), LDL-C (WMD: -11.14 mg/dL, 95 % CI: -15.41 to -6.87, P < 0.001), and TC (WMD: -13.87 mg/dL, 95 % CI: -17.99 to -9.75, P = 0.027). CONCLUSIONS Soluble fiber supplementation has the potential to improve lipid profile in patients with T2DM, and may provide a feasible approach for improving metabolic health in T2DM patients.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, UP 281406, India.
| | - Munthar Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq; College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; College of Pharmacy, The Islamic University of Babylon, Babylon, Babylon, Iraq
| | - Madhu Shukla
- Marwadi University Research Center, Department of Computer Engineering, Faculty of Engineering & Technology, Marwadi University, Rajkot, Gujarat 360003, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand 831001, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Suman Saini
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Ish Kapila
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India
| | | |
Collapse
|
20
|
Chen T, Wang T, Shi Y, Deng J, Yan X, Zhang C, Yin X, Liu W. Integrated network pharmacology, metabolomics and molecular docking analysis to reveal the mechanisms of quercetin in the treatment of hyperlipidemia. J Pharm Biomed Anal 2025; 252:116507. [PMID: 39383544 DOI: 10.1016/j.jpba.2024.116507] [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: 03/26/2024] [Revised: 09/23/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
Hyperlipidemia (HLP) is a significant contributor to cardiovascular diseases. Quercetin (QUE), a naturally occurring flavonoid with diverse bioactivities, has garnered attention due to its potential therapeutic effects. However, the precise mechanisms underlying the effects of QUE on HLP remain unclear. In this study, an ultra-high-performance liquid chromatography-quadrupole/electrostatic field Orbitrap high-resolution mass spectrometry (UPLC-Q-Exactive-MS) metabolomics strategy was employed to obtain metabolite profiles, and potential biomarkers were identified following data analysis. Network pharmacology and Drug Affinity Responsive Target Stability (DARTS) assays were utilized to explore the potential targets of QUE for HLP treatment. The results of metabolomics and network pharmacology were then integrated to identify the key targets and metabolic pathways involved in the therapeutic action of the QUE against HLP. Molecular docking and experimental validation were performed to confirm these key targets. A comprehensive database search identified 138 QUE-HLP-related targets. A protein-protein interaction (PPI) network was constructed using STRING, and the shared targets were filtered with Cytoscape. Among these, AKT1, TNF, VEGFA, mTOR, SREBP1, and SCD emerged as potential therapeutic targets. These findings were validated using in vitro cell experiments. Additionally, the mechanism of action of QUE against HLP was evaluated by integrating network pharmacology with metabolomics, identifying two metabolomic pathways crucial to HLP treatment. DARTS experiments confirmed the stable binding of QUE to FASN, p-mTOR, SREBP1, and p-AKT. In HepG2 cells treated with palmitic acid (PA), QUE significantly reduced the mRNA expression of ACLY, ACACA, FASN, and SCD (p < 0.05). Western blot analysis revealed that PA significantly increased protein expression of p-mTOR, SREBP1, FASN, and p-AKT (p < 0.05). In summary, our study provides novel insights into the protective mechanisms of QUE against HLP and offers valuable information regarding its potential benefits in clinical treatment.
Collapse
Affiliation(s)
- Tao Chen
- Department of Pharmacy, Hunan Provincial People's Hospital (The First Affiliate Hospital of Hunan Normal University), Changsha 410000, China
| | - Tongtong Wang
- Department of Pharmacy, Hunan Provincial People's Hospital (The First Affiliate Hospital of Hunan Normal University), Changsha 410000, China
| | - Yuanxiang Shi
- Institute of Clinical Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, PR China
| | - Jun Deng
- Department of Pharmacy, Hunan Provincial People's Hospital (The First Affiliate Hospital of Hunan Normal University), Changsha 410000, China
| | - Xiao Yan
- Department of Pharmacy, Hunan Provincial People's Hospital (The First Affiliate Hospital of Hunan Normal University), Changsha 410000, China
| | - Chenbin Zhang
- Department of Pharmacy, Hunan Provincial People's Hospital (The First Affiliate Hospital of Hunan Normal University), Changsha 410000, China
| | - Xin Yin
- Department of Pharmacy, Hunan Provincial People's Hospital (The First Affiliate Hospital of Hunan Normal University), Changsha 410000, China
| | - Wen Liu
- Department of Pharmacy, Hunan Provincial People's Hospital (The First Affiliate Hospital of Hunan Normal University), Changsha 410000, China.
| |
Collapse
|
21
|
Zeng G, Liu X, Zheng Z, Zhao J, Zhuo W, Bai Z, Lin E, Cai S, Cai C, Li P, Zou B, Li J. Knockdown of RASD1 improves MASLD progression by inhibiting the PI3K/AKT/mTOR pathway. Lipids Health Dis 2024; 23:424. [PMID: 39731125 DOI: 10.1186/s12944-024-02419-z] [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: 10/29/2024] [Accepted: 12/22/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND There is still no reliable therapeutic targets and effective pharmacotherapy for metabolic dysfunction-associated steatotic liver disease (MASLD). RASD1 is short for Ras-related dexamethasone-induced 1, a pivotal factor in various metabolism processes of Human. However, the role of RASD1 remains poorly illustrated in MASLD. Therefore, we designed a study to elucidate how RASD1 could impact on MASLD as well as the mechanisms involved. METHODS The expression level of RASD1 was validated in MASLD. Lipid metabolism and its underlying mechanism were investigated in hepatocytes and mice with either overexpression or knockdown of RASD1. RESULTS Hepatic RASD1 expression was upregulated in MASLD. Lipid deposition was significantly reduced in RASD1-knockdown hepatocytes and mice, accompanied by a marked downregulation of key genes in the signaling pathway of de novo lipogenesis. Conversely, RASD1 overexpression in hepatocytes had the opposite effect. Mechanistically, RASD1 regulated lipid metabolism in MASLD through the PI3K/AKT/mTOR signaling pathway. CONCLUSIONS We discovered a novel role of RASD1 in MASLD by regulating lipogenesis via the PI3K/AKT/mTOR pathway, thereby identifying a potential treatment target for MASLD.
Collapse
Affiliation(s)
- Guifang Zeng
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| | - Xialei Liu
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Zhouying Zheng
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Jiali Zhao
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Wenfeng Zhuo
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Zirui Bai
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - En Lin
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Shanglin Cai
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Chaonong Cai
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China
| | - Peiping Li
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| | - Baojia Zou
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| | - Jian Li
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, 519000, People's Republic of China.
| |
Collapse
|
22
|
Feng S, Zeng Y, Song F, Shen M, Yang F. Microcystins Exposure and the Risk of Metabolic Syndrome: A Cross-Sectional Study in Central China. Toxins (Basel) 2024; 16:542. [PMID: 39728800 PMCID: PMC11679381 DOI: 10.3390/toxins16120542] [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: 10/15/2024] [Revised: 11/08/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
A growing body of evidence indicates that microcystins (MCs) exposure may cause metabolic diseases. However, studies exploring the effects of MCs exposure on the risk of metabolic syndrome (MetS) in humans are currently lacking, and the underlying mechanisms remain unclear. Here, we conducted a cross-sectional study in central China to explore the effect of serum MCs on MetS, and assessed the mediation effects of the inflammation biomarker, white blood cell (WBC) level, in this relationship. The relationships among MCs and WBC level and risk of MetS were assessed using binary logistic and linear regression. Mediation analysis was used to explore possible mechanisms underlying those associations by employing R software (version 4.3.1). Compared to the lowest quartile of MCs, the highest quartile had an increased risk of MetS (odds ratio [OR] = 2.10, 95% confidence interval [CI]: 1.19, 3.70), with a dose-response relationship (p for trend < 0.05). WBCs mediated 11.14% of the association between serum MCs and triglyceride (TG) levels, but did not mediate the association of MCs exposure with MetS. This study firstly reveals that MCs exposure is an independent risk factor for MetS in a dose-response manner, and suggests that WBC level could partially mediate the association of MCs exposure with TG levels.
Collapse
Affiliation(s)
- Shuidong Feng
- Department of Epidemiology and Health Statistics, The Key Laboratory of Typical Environmental Pollution and Health Hazards of Hunan Province, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.F.); (Y.Z.); (F.S.)
| | - Yuke Zeng
- Department of Epidemiology and Health Statistics, The Key Laboratory of Typical Environmental Pollution and Health Hazards of Hunan Province, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.F.); (Y.Z.); (F.S.)
| | - Fengmei Song
- Department of Epidemiology and Health Statistics, The Key Laboratory of Typical Environmental Pollution and Health Hazards of Hunan Province, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.F.); (Y.Z.); (F.S.)
| | - Minxue Shen
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Department of Social Medicine and Health Management, Xiangya School of Public Health, Central South University, Changsha 410000, China
| | - Fei Yang
- Department of Epidemiology and Health Statistics, The Key Laboratory of Typical Environmental Pollution and Health Hazards of Hunan Province, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China; (S.F.); (Y.Z.); (F.S.)
| |
Collapse
|
23
|
Mamache W, Amira S, Benchikh F, Benabdallah H, Bencheikh A, Amira H, Ounis R, Torki MA, Kaoudoune C. The therapeutic potential of Salvia aegyptiaca extracts in ethanol-induced gastric ulcer: insights into macroscopic, histopathological, and biochemical mechanisms. Turk J Biol 2024; 49:40-51. [PMID: 40104582 PMCID: PMC11913356 DOI: 10.55730/1300-0152.2722] [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: 10/13/2024] [Revised: 02/24/2025] [Accepted: 12/02/2024] [Indexed: 03/20/2025] Open
Abstract
Background/aim This study explores the antiulcer activity of different doses of Salvia aegyptiaca (SAE) methanol (ME) and decocted extracts (DE) on ethanol-induced gastric ulcers in rats. Materials and methods Female Wistar rats weighing 180-200 g were divided into five groups: control, omeprazole (positive control), and extract-treated (100, 200, and 400 mg/kg). Ulcers were induced with absolute ethanol 30 min after treatment with the extracts. The experiment was followed by macroscopic and histopathological examination. In vitro tests were also conducted to assess lipid peroxidation, catalase activity, mucus content, glutathione, and protein levels. Results The study found that 100% ethanol caused significant damage, including colour and mucus loss, petechiae, haemorrhages, and oedema. However, pretreatment with ME SAE or DE SAE at doses of all three levels reduced the ethanol-induced damage. Histopathological analysis revealed reduced signs of haemorrhagic lesions, infiltration, and oedema in rats treated with ME SAE or DE SAE at doses of 100 or 200 mg/kg, whereas the 400 mg/kg dose provided complete protection. Comparable to the use of omeprazole, ingestion of DE SAE at doses of 100, 200, or 400 mg/kg demonstrated substantial protection against stomach ulcers produced by ethanol, with a range of 76%-84%. Both SAE extracts induced a dose-dependent increase in glutathione levels, with DE SAE showing a significant rise at 200 and 400 mg/kg. Conclusion The SAE extracts demonstrated a significant decrease in gastric lipid peroxidation, outperforming the effect of omeprazole.
Collapse
Affiliation(s)
- Walid Mamache
- Laboratory of Applied Phytotherapy to Chronic Disease, Department of Animal Biology, Faculty of Natural Life Sciences, Ferhat Abbas University, Setif, Algeria
| | - Smain Amira
- Laboratory of Applied Phytotherapy to Chronic Disease, Department of Animal Biology, Faculty of Natural Life Sciences, Ferhat Abbas University, Setif, Algeria
| | - Fatima Benchikh
- Laboratory of Applied Phytotherapy to Chronic Disease, Department of Animal Biology, Faculty of Natural Life Sciences, Ferhat Abbas University, Setif, Algeria
| | - Hassiba Benabdallah
- Laboratory of Applied Phytotherapy to Chronic Disease, Department of Animal Biology, Faculty of Natural Life Sciences, Ferhat Abbas University, Setif, Algeria
| | - Amor Bencheikh
- Laboratory of Applied Microbiology, Department of Microbiology, Faculty of Natural Life Sciences, Ferhat Abbas University, Setif, Algeria
| | - Hind Amira
- Laboratory of Applied Phytotherapy to Chronic Disease, Department of Animal Biology, Faculty of Natural Life Sciences, Ferhat Abbas University, Setif, Algeria
| | - Roumaissa Ounis
- Laboratory of Applied Phytotherapy to Chronic Disease, Department of Animal Biology, Faculty of Natural Life Sciences, Ferhat Abbas University, Setif, Algeria
| | - Mohammed Abdallah Torki
- Laboratory of Applied Phytotherapy to Chronic Disease, Department of Animal Biology, Faculty of Natural Life Sciences, Ferhat Abbas University, Setif, Algeria
| | - Chahrazed Kaoudoune
- Laboratory of Applied Phytotherapy to Chronic Disease, Department of Animal Biology, Faculty of Natural Life Sciences, Ferhat Abbas University, Setif, Algeria
| |
Collapse
|
24
|
Hao Z, Guo X, Wang Y, Yang G. Physical Activity Reduces Metabolic Risk via Iron Metabolism: Cross-National Evidence Using the Triglyceride-Glucose Index. Metabolites 2024; 14:651. [PMID: 39728432 DOI: 10.3390/metabo14120651] [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: 11/04/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Purpose: Studies suggest that the triglyceride-glucose index (TyG) is a novel and comprehensive marker of metabolic health. While most research indicates that increased physical activity (PA) is linked to improved metabolic health, some studies argue that the previous markers may not fully capture this relationship. This study uses TyG as a marker of metabolic health to examine the association between PA and TyG. Methods: Data are from cross-sectional surveys in three large population studies in China and the United States: CHARLS, CHNS, and NHANES. Regression models were applied to analyze the relationship between PA and TyG, with covariates adjusted in a stepwise manner. Stratified analysis was used to explore this relationship among different population groups, and, since it has been suggested that iron metabolism plays an important role in metabolic health, it was used as a mediating variable to construct a mediation model for analysis and discussion. Results: Higher PA was significantly associated with lower TyG levels across all three databases (p < 0.001), and this relationship remained robust after full adjustment for covariates. This negative association was more pronounced in older males (over 45 years). Iron metabolism also mediated this relationship, with mediation proportions ranging from 10% to 12.5%. Conclusions: There is a significant inverse association between PA and TyG, suggesting a link between increased PA and metabolic health, with iron metabolism moderating this relationship, especially among older males.
Collapse
Affiliation(s)
- Zikang Hao
- School of Physical Education, Shandong University, Jinan 250061, China
- Laboratory of Exercise Science, Ocean University of China, Qingdao 261000, China
| | - Xinmeng Guo
- School of Physical Education, Shandong University, Jinan 250061, China
| | - Yitao Wang
- Kunshan Hospital of Traditional Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Kunshan 215300, China
- School of Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Guang Yang
- Kunshan Hospital of Traditional Chinese Medicine, Nanjing University of Traditional Chinese Medicine, Kunshan 215300, China
- School of Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
25
|
Yang Y, Wu J, Zhou W, Ji G, Dang Y. Protein posttranslational modifications in metabolic diseases: basic concepts and targeted therapies. MedComm (Beijing) 2024; 5:e752. [PMID: 39355507 PMCID: PMC11442990 DOI: 10.1002/mco2.752] [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: 05/25/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024] Open
Abstract
Metabolism-related diseases, including diabetes mellitus, obesity, hyperlipidemia, and nonalcoholic fatty liver disease, are becoming increasingly prevalent, thereby posing significant threats to human health and longevity. Proteins, as the primary mediators of biological activities, undergo various posttranslational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, methylation, and SUMOylation, among others, which substantially diversify their functions. These modifications are crucial in the physiological and pathological processes associated with metabolic disorders. Despite advancements in the field, there remains a deficiency in contemporary summaries addressing how these modifications influence processes of metabolic disease. This review aims to systematically elucidate the mechanisms through which PTM of proteins impact the progression of metabolic diseases, including diabetes, obesity, hyperlipidemia, and nonalcoholic fatty liver disease. Additionally, the limitations of the current body of research are critically assessed. Leveraging PTMs of proteins provides novel insights and therapeutic targets for the prevention and treatment of metabolic disorders. Numerous drugs designed to target these modifications are currently in preclinical or clinical trials. This review also provides a comprehensive summary. By elucidating the intricate interplay between PTMs and metabolic pathways, this study advances understanding of the molecular mechanisms underlying metabolic dysfunction, thereby facilitating the development of more precise and effective disease management strategies.
Collapse
Affiliation(s)
- Yunuo Yang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Jiaxuan Wu
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Wenjun Zhou
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Guang Ji
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Yanqi Dang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| |
Collapse
|
26
|
Tan Q, Chu H, Wei J, Yan S, Sun X, Wang J, Zhu L, Yang F. Astaxanthin Alleviates Hepatic Lipid Metabolic Dysregulation Induced by Microcystin-LR. Toxins (Basel) 2024; 16:401. [PMID: 39330859 PMCID: PMC11435617 DOI: 10.3390/toxins16090401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024] Open
Abstract
Microcystin-LR (MC-LR), frequently generated by cyanobacteria, has been demonstrated to raise the likelihood of liver disease. Few previous studies have explored the potential antagonist against MC-LR. Astaxanthin (ASX) has been shown to possess various beneficial effects in regulating lipid metabolism in the liver. However, whether ASX could alleviate MC-LR-induced hepatic lipid metabolic dysregulation is as yet unclear. In this work, the important roles and mechanisms of ASX in countering MC-LR-induced liver damage and lipid metabolic dysregulation were explored for the first time. The findings revealed that ASX not only prevented weight loss but also enhanced liver health after MC-LR exposure. Moreover, ASX effectively decreased triglyceride, total cholesterol, aspartate transaminase, and alanine aminotransferase contents in mice that were elevated by MC-LR. Histological observation showed that ASX significantly alleviated lipid accumulation and inflammation induced by MC-LR. Mechanically, ASX could significantly diminish the expression of genes responsible for lipid generation (Srebp-1c, Fasn, Cd36, Scd1, Dgat1, and Pparg), which probably reduced lipid accumulation induced by MC-LR. Analogously, MC-LR increased intracellular lipid deposition in THLE-3 cells, while ASX decreased these symptoms by down-regulating the expression of key genes in the lipid synthesis pathway. Our results implied that ASX played a crucial part in lipid synthesis and effectively alleviated MC-LR-induced lipid metabolism dysregulation. ASX might be developed as a novel protectant against hepatic impairment and lipid metabolic dysregulation associated with MC-LR. This study offers new insights for further management of MC-LR-related metabolic diseases.
Collapse
Affiliation(s)
- Qinmei Tan
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.T.); (S.Y.); (X.S.)
| | - Hanyu Chu
- Hengyang Maternal and Child Health Hospital, Hengyang 421001, China;
| | - Jia Wei
- Xiangya School of Public Health, Central South University, Changsha 410078, China;
| | - Sisi Yan
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.T.); (S.Y.); (X.S.)
| | - Xiaoya Sun
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.T.); (S.Y.); (X.S.)
| | - Jiangping Wang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China;
| | - Lemei Zhu
- School of Public Health, Changsha Medical University, Changsha 410219, China;
| | - Fei Yang
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China; (Q.T.); (S.Y.); (X.S.)
- Affiliated Nanhua Hospital University of South China, Hengyang 421000, China
| |
Collapse
|
27
|
Li Y, He Q, Chen S, Dli H, Zhao J, Sun X, Yang P, Mao Q, Xia H. BI-7273, a BRD9 inhibitor, reduces lipid accumulation by downregulating the AKT/mTOR/SREBP1 signaling pathway. Biochem Pharmacol 2024; 226:116412. [PMID: 38971334 DOI: 10.1016/j.bcp.2024.116412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/24/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Increases in de novo lipogenesis that disturbed lipid homeostasis and caused lipid accumulation are a major cause of NAFLD and obesity. SREBP1 is a crucial regulatory factor controlling the expression of rate-limiting enzymes of lipid synthesis. A reduction in SREBP1expression can reduce lipid accumulation. Thus, we utilized an SREBP1-luciferase-KI HEK293 cell line constructed by our lab to screen 200 kinds of epigenetic drugs for their ability to downregulate SREBP1expression. BI-7273, an inhibitor of bromodomain-containing protein 9 (BRD9), was screened and found to decrease SREBP1 expression. What is more, BI-7273 has been confirmed that it could reduce lipid accumulation in HepG2 cells by BODIPY staining, and significantly decrease the protein expression of SREBP1 and FASN. To explore the potential mechanism BI-7273 reducing lipid accumulation, RNA sequencing (RNA-seq) was performed and demonstrated that BI-7273 reduced lipid accumulation by downregulating the AKT/mTOR/SREBP1 pathway in vitro. Finally, these results were verified in NAFLD and obesity mouse model induced by high fat diet (HFD). The results indicated that BI-7273 could decrease mouse body weight and improve insulin sensitivity, but also exhibited a strong negative correlation with serum lipid levels, and also demonstrated that BI-7273 reduced lipid accumulation via AKT/mTOR/SREBP1 pathway in vivo. In conclusion, our results revealed that BI-7273 decreases lipid accumulation by downregulating the AKT/mTOR/SREBP1 pathway in vivo and in vitro. This is the first report demonstrating the protective effect of this BRD9 inhibitor against NAFLD and obesity. BRD9 may be a novel target for the discovery of effective drugs to treat lipid metabolism disorders.
Collapse
Affiliation(s)
- Yu Li
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Qiongyan He
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Shuyu Chen
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Huma Dli
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Junli Zhao
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Xiaohong Sun
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Peiyan Yang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Qinwen Mao
- Department of Pathology, University of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China.
| |
Collapse
|
28
|
Jia W, Zhong L, Ren Q, Teng D, Gong L, Dong H, Li J, Wang C, He YX, Yang J. Microcystin-RR promote lipid accumulation through CD36 mediated signal pathway and fatty acid uptake in HepG2 cells. ENVIRONMENTAL RESEARCH 2024; 249:118402. [PMID: 38309560 DOI: 10.1016/j.envres.2024.118402] [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: 11/20/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/05/2024]
Abstract
Microcystins (MC)-RR is a significant analogue of MC-LR, which has been identified as a hepatotoxin capable of influencing lipid metabolism and promoting the progression of liver-related metabolic diseases. However, the toxicity and biological function of MC-RR are still not well understood. In this study, the toxic effects and its role in lipid metabolism of MC-RR were investigated in hepatoblastoma cells (HepG2cells). The results demonstrated that MC-RR dose-dependently reduced cell viability and induced apoptosis. Additionally, even at low concentrations, MC-RR promoted lipid accumulation through up-regulating levels of triglyceride, total cholesterol, phosphatidylcholines and phosphatidylethaolamine in HepG2 cells, with no impact on cell viability. Proteomics and transcriptomics analysis further revealed significant alterations in the protein and gene expression profiles in HepG2 cells treated with MC-RR. Bioinformatic analysis, along with subsequent validation, indicated the upregulation of CD36 and activation of the AMPK and PI3K/AKT/mTOR in response to MC-RR exposure. Finally, knockdown of CD36 markedly ameliorated MC-RR-induced lipid accumulation in HepG2 cells. These findings collectively suggest that MC-RR promotes lipid accumulation in HepG2 cells through CD36-mediated signal pathway and fatty acid uptake. Our findings provide new insights into the hepatotoxic mechanism of MC-RR.
Collapse
Affiliation(s)
- Wenjuan Jia
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China.
| | - Lin Zhong
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Qingmiao Ren
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Da Teng
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Lei Gong
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Haibin Dong
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Jun Li
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Chunxiao Wang
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Yong-Xing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Jun Yang
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China.
| |
Collapse
|
29
|
Uddandrao VVS, Chandrasekaran P, Saravanan G, Brahmanaidu P, Sengottuvelu S, Ponmurugan P, Vadivukkarasi S, Kumar U. Phytoformulation with hydroxycitric acid and capsaicin protects against high-fat-diet-induced obesity cardiomyopathy by reducing cardiac lipid deposition and ameliorating inflammation and apoptosis in the heart. J Tradit Complement Med 2024; 14:162-172. [PMID: 38481548 PMCID: PMC10927456 DOI: 10.1016/j.jtcme.2023.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/15/2023] [Accepted: 08/23/2023] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND AND AIM Phytoformulation therapy is a pioneering strategy for the treatment of metabolic disorders and related diseases. The aim of the present study was to investigate the protective effect of a phytoformulation consisting of hydroxycitric acid and capsaicin against obesity-related cardiomyopathy. EXPERIMENTAL PROCEDURE Sprague-Dawley rats were fed HFD for 21 weeks, and phytoformulation (100 mg/kg body weight) was administered orally for 45 days starting at week 16. RESULTS AND CONCLUSION We found that HFD supplementation resulted in significant hyperglycemia and caused an increase in cardiac lipid deposition, inflammation and apoptosis in the heart. Phytoformulation therapy not only significantly decreased blood levels of glucose, cholesterol, triglycerides, free fatty acids, and inflammatory cytokines in obese rats, but also protected cardiac tissue, as shown by histological analysis. Conversely, phytoformulation therapy decreased mRNA levels for sterol regulatory element-binding factor 1, fatty acid synthase, acetyl-CoA carboxylase, and fatty acid binding protein 1 genes involved in fatty acid synthesis and absorption in obese rats. It increased the levels of lysosomal acid lipase, hormone-sensitive lipase, and lipoprotein lipase genes involved in fatty acid degradation in the heart. In addition, the phytoformulation improved cardiac inflammation and apoptosis by downregulating the genes nuclear factor kappa-light-chain enhancer of activated B cells (NF-kB), tumour necrosis factor α, interleukin-6, toll-like receptor-4 (TLR-4), BCL2-associated X and caspase-3. In conclusion, our results show that the phytoformulation improved insulin sensitivity and attenuated myocardial lipid accumulation, inflammation, and apoptosis in the heart of HFD-induced obese rats by regulating fatty acid metabolism genes and downregulating NF-kB/TLR-4/caspase-3.
Collapse
Affiliation(s)
- V. V. Sathibabu Uddandrao
- Department of Biochemistry, K.S. Rangasamy College of Arts and Science (Autonomous), Tiruchengode, Namakkal District, Tamilnadu, 637215, India
| | - P. Chandrasekaran
- Department of Biochemistry, K.S. Rangasamy College of Arts and Science (Autonomous), Tiruchengode, Namakkal District, Tamilnadu, 637215, India
| | - G. Saravanan
- Department of Biochemistry, K.S. Rangasamy College of Arts and Science (Autonomous), Tiruchengode, Namakkal District, Tamilnadu, 637215, India
| | - Parim Brahmanaidu
- Animal Physiology and Biochemistry Laboratory, ICMR-National Animal Resource Facility for Biomedical Research (ICMR-NARFBR), Hyderabad, 500078, India
| | - S. Sengottuvelu
- Department of Pharmacology, Nandha College of Pharmacy, Erode, Tamilnadu, 638052, India
| | - P. Ponmurugan
- Department of Botany, Bharathiar University, Coimbatore, Tamilnadu, 641046, India
| | - S. Vadivukkarasi
- Department of Biochemistry, K.S. Rangasamy College of Arts and Science (Autonomous), Tiruchengode, Namakkal District, Tamilnadu, 637215, India
| | - Umesh Kumar
- School of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), Ghaziabad, Uttar Pradesh, 201015, India
| |
Collapse
|
30
|
Wang T, Yang J, Huang Z, Wang F, Liu R, Liu Y, Li X. Integrated 16s RNA sequencing and network pharmacology to explore the effects of polyphenol-rich raspberry leaf extract on weight control. Front Nutr 2024; 10:1306037. [PMID: 38260083 PMCID: PMC10800909 DOI: 10.3389/fnut.2023.1306037] [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: 10/03/2023] [Accepted: 12/08/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Obesity is recognized as a chronic low-grade inflammation associated with intestinal flora imbalance, leading to dyslipidemia and inflammation. Modern research has found that polyphenols have anti-obesity effects. However, the mechanism of action of raspberry leaf extract (RLE) with high polyphenols in regulating obesity is still unknown. This study investigated the improvement effect of supplementing RLE on high-fat diet (HFD) induced obesity in mice. Methods RLE was used to intervene in HFD induced C57BL/6J male mice during prevention stage (1-16 weeks) and treatment stage (17-20 weeks). Their weight changes and obesity-related biochemical indicators were measured. The changes in intestinal flora were analyzed using 16S rRNA sequencing, and finally the targets and pathways of the 7 typical polyphenols (quercetin-3-O-glucuronide, ellagic acid, kaempferol-3-O-rutinoside, chlorogenic acid, brevifolin carboxylic acid, quercetin-3-O-rutinoside, and quercetin) of RLE in the regulation of obesity were predicted by network pharmacology approach. Results and discussion The results showed that RLE effectively prevented and treated weight gain in obese mice induced by HFD, alleviated adipocyte hypertrophy, reduced Interleukin-6 and Tumor Necrosis Factor Alpha levels, and improved intestinal flora, especially Muriaculaceae, Alistipes and Alloprevotella, and decreased the Firmicutes/Bacteroidota ratio. Network pharmacology analysis selected 60 common targets for 7 RLE polyphenols and obesity. Combined with protein-protein interaction network, enrichment analysis and experimental results, TNF, IL-6, AKT1, and PPAR were predicted as potential key targets for RLE polyphenols. Conclusion The potential mechanism by which polyphenol-rich RLE regulates obesity may be attributed to the specific polyphenols of RLE and their synergistic effects, therefore RLE has a great anti-obesity potential and may be used as a means to alleviate obesity and related diseases.
Collapse
Affiliation(s)
- Tao Wang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan, Shanxi, China
- Dezhou Industrial Technology Research Institute of North University of China, Dezhou, Shandong, China
| | - Jing Yang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan, Shanxi, China
- Dezhou Industrial Technology Research Institute of North University of China, Dezhou, Shandong, China
| | - Ziang Huang
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan, Shanxi, China
- Dezhou Industrial Technology Research Institute of North University of China, Dezhou, Shandong, China
| | - Fei Wang
- The Hospital of North University of China, Taiyuan, Shanxi, China
| | - Ruzi Liu
- Dezhou Yongshengzhai Braised Chicken Group Co., Ltd., Dezhou, Shangdong, China
| | - Yongping Liu
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan, Shanxi, China
| | - Xiaojun Li
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan, Shanxi, China
| |
Collapse
|
31
|
Han LL, Zhang X, Zhang H, Li T, Zhao YC, Tian MH, Sun FL, Feng B. Alisol B 23-acetate promotes white adipose tissue browning to mitigate high-fat diet-induced obesity by regulating mTOR-SREBP1 signaling. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:83-92. [PMID: 38311542 DOI: 10.1016/j.joim.2024.01.003] [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: 07/08/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024]
Abstract
OBJECTIVE Obesity is a global health concern with management strategies encompassing bariatric surgery and anti-obesity drugs; however, concerns regarding complexities and side effects persist, driving research for more effective, low-risk strategies. The promotion of white adipose tissue (WAT) browning has emerged as a promising approach. Moreover, alisol B 23-acetate (AB23A) has demonstrated efficacy in addressing metabolic disorders, suggesting its potential as a therapeutic agent in obesity management. Therefore, in this study, we aimed to investigate the therapeutic potential of AB23A for mitigating obesity by regulating metabolic phenotypes and lipid distribution in mice fed a high-fat diet (HFD). METHODS An obesity mouse model was established by administration of an HFD. Glucose and insulin metabolism were assessed via glucose and insulin tolerance tests. Adipocyte size was determined using hematoxylin and eosin staining. The expression of browning markers in WAT was evaluated using Western blotting and quantitative real-time polymerase chain reaction. Metabolic cage monitoring involved the assessment of various parameters, including food and water intake, energy metabolism, respiratory exchange rates, and physical activity. Moreover, oil red O staining was used to evaluate intracellular lipid accumulation. A bioinformatic analysis tool for identifying the molecular mechanisms of traditional Chinese medicine was used to examine AB23A targets and associated signaling pathways. RESULTS AB23A administration significantly reduced the weight of obese mice, decreased the mass of inguinal WAT, epididymal WAT, and perirenal adipose tissue, improved glucose and insulin metabolism, and reduced adipocyte size. Moreover, treatment with AB23A promoted the expression of browning markers in WAT, enhanced overall energy metabolism in mice, and had no discernible effect on food intake, water consumption, or physical activity. In 3T3-L1 cells, AB23A inhibited lipid accumulation, and both AB23A and rapamycin inhibited the mammalian target of rapamycin-sterol regulatory element-binding protein-1 (mTOR-SREBP1) signaling pathway. Furthermore, 3-isobutyl-1-methylxanthine, dexamethasone and insulin, at concentrations of 0.25 mmol/L, 0.25 μmol/L and 1 μg/mL, respectively, induced activation of the mTOR-SREBP1 signaling pathway, which was further strengthened by an mTOR activator MHY1485. Notably, MHY1485 reversed the beneficial effects of AB23A in 3T3-L1 cells. CONCLUSION AB23A promoted WAT browning by inhibiting the mTOR-SREBP1 signaling pathway, offering a potential strategy to prevent obesity. Please cite this article as: Han LL, Zhang X, Zhang H, Li T, Zhao YC, Tian MH, Sun FL, Feng B. Alisol B 23-acetate promotes white adipose tissue browning to mitigate high-fat diet-induced obesity by regulating mTOR-SREBP1 signaling. J Integr Med. 2024; 22(1): 83-92.
Collapse
Affiliation(s)
- Lu-Lu Han
- Department of Neurology Three, The Fifth People's Hospital of Jinan, Jinan 250013, Shandong Province, China
| | - Xin Zhang
- Department of Gastroenterology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Hui Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Ting Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Yi-Chen Zhao
- Department of Geriatrics, the First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital), Jinan 250014, Shandong Province, China
| | - Ming-Hui Tian
- Chinese Medicine Culture and Literature Research Institute, Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Feng-Lei Sun
- Department of General Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong Province, China
| | - Bo Feng
- Department of Geriatrics, the First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital), Jinan 250014, Shandong Province, China; Department of Traditional Chinese Medicine, the Second People's Hospital of Haibei Prefecture, Zangzu Autonomous Prefecture of Haibei, 810300, Qinghai Province, China.
| |
Collapse
|
32
|
陈 静, 黄 英, 赵 景. [Trimethylamine N-Oxide Induces Renal Fibrosis Through the PI3K/AKT/SREBP1 Pathway]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:1105-1111. [PMID: 38162065 PMCID: PMC10752791 DOI: 10.12182/20231160106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Indexed: 01/03/2024]
Abstract
Objective To investigate the role and mechanism of trimethylamine N-oxide (TMAO), a uremic toxin, in renal fibrosis. Methods A total of 20 male BALB/c mice were randomly and evenly assigned to a Control group and a TMAO group. Mice in the Control group received intraperitoneal injection of normal saline, while mice in the TMAO group received intraperitoneal injection of TMAO (20 mg/[kg·d]). The injection was given once a day for 8 weeks. Histopathology and fibrosis of kidney were observed by H&E staining and Masson staining. Immunohistochemistry was performed to determine the levels of alpha smooth muscle actin (α-SMA), recombinant human fibronectin fragment (Fibronectin), and sterol-regulatory element binding protein 1 (SREBP1). Western blot was performed to determine α-SMA, SREBP1, phosphatidylinositol 3 kinase (PI3K), phospho-phosphatidylinositol 3 kinase (p-PI3K), protein kinase B (PKB, also known as AKT), and phospho-AKT (p-AKT) protein levels. HK2 cells were treated with SREBP1 small interfering RNA (siRNA) and PI3K/AKT inhibitor, respectively, and the reversal of the effects of TMAO was examined. Results Animal experiments showed that, compared with the Control group, the mice treated with TMAO experienced pathological damage and fibrosis of the kidney tissue and the expression levels of fibrosis markers, α-SMA and Fibronectin, in the kidney were increased (all P<0.05). According to the findings from further investigation, the TMAO-treatment group showed increased expression of SREBP1 and an up-regulation of PI3K phosphorylation ratio and AKT phosphorylation ratio compared with those of the Control group (all P<0.05). Cell experiments produced results similar to those of the animal experiment. After siRNA interference with SREBP1 expression, the expression levels of fibrosis marker proteins decreased (P<0.05). Besides, the high expression of SREBP1 caused by TMAO was inhibited after HK2 cells were incubated with LY294002, a PI3K-AKT pathway inhibitor (P<0.05). Conclusion TMAO may induce renal fibrosis by promoting the PI3K/AKT/SREBP1 pathway.
Collapse
Affiliation(s)
- 静 陈
- 陆军军医大学第二附属医院 肾脏内科 (重庆 400037)Department of Nephrology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - 英辉 黄
- 陆军军医大学第二附属医院 肾脏内科 (重庆 400037)Department of Nephrology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - 景宏 赵
- 陆军军医大学第二附属医院 肾脏内科 (重庆 400037)Department of Nephrology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
33
|
Liu Y, Xing L, Zhang Y, Liu X, Li T, Zhang S, Wei H, Li J. Mild Intermittent Cold Stimulation Affects Cardiac Substance Metabolism via the Neuroendocrine Pathway in Broilers. Animals (Basel) 2023; 13:3577. [PMID: 38003194 PMCID: PMC10668735 DOI: 10.3390/ani13223577] [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: 09/04/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
This study aimed to investigate the impact of cold adaptation on the neuroendocrine and cardiac substance metabolism pathways in broilers. The broilers were divided into the control group (CC), cold adaptation group (C3), and cold-stressed group (C9), and experimental period was divided into the training period (d 1-35), recovery period (d 36-43), and cold stress period (d 43-44). During the training period, the CC group was reared at ambient temperature, while C3 and C9 groups were reared at 3 °C and 9 °C lower than the ambient temperature, respectively, for 5 h/d at 1 d intervals. During the recovery period, all the groups were maintained at 20 °C. Lastly, during the cold stress period, the groups were divided into two sub-groups, and each sub-group was placed at 10 °C for 12 h (Y12) or 24 h (Y24) for acute cold stimulation. The blood, hypothalamic, and cardiac tissues samples were obtained from all the groups during the training, recovery, and acute stress periods. The results revealed that the transcription of calcium voltage-gated channel subunit alpha 1 C (CACNAIC) was increased in the hypothalamic tissues of the C3 group (p < 0.05). Moreover, compared to the CC group, the serum norepinephrine (NE) was increased in the C9 group (p < 0.05), but insulin (INS) was decreased in the C9 group (p < 0.05). In addition, the transcription of the phosphoinositide-3 kinase (PI3K), protein kinase B (Akt), mammalian target of rapamycin (mTOR), SREBP1c, FASN, ACC1, and SCD genes was down-regulated in the C3 and C9 groups (p < 0.05); however, their expression increased in the C3 and C9 groups after acute cold stimulation (p < 0.05). Compared to the CC group, the transcription of forkhead box O1 (FoxO1), PEPCK, G6Pase, GLUT1, HK1, PFK, and LDHB genes was up-regulated in the C3 and C9 groups (p < 0.05. Furthermore, compared to the CC and C9 groups, the protein and mRNA expressions of heat shock protein (HSP) 70 and HSP90 were significantly increased in the C3 group (p < 0.05). These results indicate that intermittent cold training can enhance cold stress tolerance in broilers by regulating their neuroendocrine and cardiac substance metabolism pathways.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Lu Xing
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Yong Zhang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Xiaotao Liu
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Tingting Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Shijie Zhang
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Haidong Wei
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, Harbin 150030, China; (Y.L.); (L.X.); (Y.Z.); (X.L.); (T.L.); (S.Z.)
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China
| |
Collapse
|
34
|
Niture S, Gadi S, Qi Q, Rios-Colon L, Khatiwada S, Vandana, Fernando RA, Levine KE, Kumar D. Cyanotoxins Increase Cytotoxicity and Promote Nonalcoholic Fatty Liver Disease Progression by Enhancing Cell Steatosis. Toxins (Basel) 2023; 15:411. [PMID: 37505679 PMCID: PMC10467139 DOI: 10.3390/toxins15070411] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Freshwater prokaryotic cyanobacteria within harmful algal blooms produce cyanotoxins which are considered major pollutants in the aquatic system. Direct exposure to cyanotoxins through inhalation, skin contact, or ingestion of contaminated drinking water can target the liver and may cause hepatotoxicity. In the current study, we investigated the effect of low concentrations of cyanotoxins on cytotoxicity, inflammation, modulation of unfolded protein response (UPR), steatosis, and fibrosis signaling in human hepatocytes and liver cell models. Exposure to low concentrations of microcystin-LR (MC-LR), microcystin-RR (MC-RR), nodularin (NOD), and cylindrospermopsin (CYN) in human bipotent progenitor cell line HepaRG and hepatocellular carcinoma (HCC) cell lines HepG2 and SK-Hep1 resulted in increased cell toxicity. MC-LR, NOD, and CYN differentially regulated inflammatory signaling, activated UPR signaling and lipogenic gene expression, and induced cellular steatosis and fibrotic signaling in HCC cells. MC-LR, NOD, and CYN also regulated AKT/mTOR signaling and inhibited autophagy. Chronic exposure to MC-LR, NOD, and CYN upregulated the expression of lipogenic and fibrosis biomarkers. Moreover, RNA sequencing (RNA seq) data suggested that exposure of human hepatocytes, HepaRG, and HCC HepG2 cells to MC-LR and CYN modulated expression levels of several genes that regulate non-alcoholic fatty liver disease (NAFLD). Our data suggest that low concentrations of cyanotoxins can cause hepatotoxicity and cell steatosis and promote NAFLD progression.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Sashi Gadi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Qi Qi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Leslimar Rios-Colon
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Sabin Khatiwada
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Vandana
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| | - Reshan A. Fernando
- NCCU-RTI Center for Applied Research in Environmental Sciences (CARES), RTI International, Durham, NC 27707, USA
| | - Keith E. Levine
- NCCU-RTI Center for Applied Research in Environmental Sciences (CARES), RTI International, Durham, NC 27707, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707, USA
| |
Collapse
|
35
|
Feng S, Cao M, Tang P, Deng S, Chen L, Tang Y, Zhu L, Chen X, Huang Z, Shen M, Yang F. Microcystins Exposure Associated with Blood Lipid Profiles and Dyslipidemia: A Cross-Sectional Study in Hunan Province, China. Toxins (Basel) 2023; 15:toxins15040293. [PMID: 37104231 PMCID: PMC10143012 DOI: 10.3390/toxins15040293] [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: 02/28/2023] [Revised: 04/01/2023] [Accepted: 04/05/2023] [Indexed: 04/28/2023] Open
Abstract
Increasing evidence from experimental research suggests that exposure to microcystins (MCs) may induce lipid metabolism disorder. However, population-based epidemiological studies of the association between MCs exposure and the risk of dyslipidemia are lacking. Therefore, we conducted a population-based cross-sectional study involving 720 participants in Hunan Province, China, and evaluated the effects of MCs on blood lipids. After adjusting the lipid related metals, we used binary logistic regression and multiple linear regression models to examine the associations among serum MCs concentration, the risk of dyslipidemia and blood lipids (triglyceride (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C)). Moreover, the additive model was used to explore the interaction effects on dyslipidemia between MCs and metals. Compared to the lowest quartile of MCs exposure, the risk of dyslipidemia [odds ratios (OR) = 2.27, 95% confidence interval (CI): 1.46, 3.53] and hyperTG (OR = 3.01, 95% CI: 1.79, 5.05) in the highest quartile was significantly increased, and showed dose-response relationships. MCs were positively associated with TG level (percent change, 9.43%; 95% CI: 3.53%, 15.67%) and negatively associated with HDL-C level (percent change, -3.53%; 95% CI: -5.70%, -2.10%). In addition, an additive antagonistic effect of MCs and Zn on dyslipidemia was also reported [relative excess risk due to interaction (RERI) = -1.81 (95% CI: -3.56, -0.05)], and the attributable proportion of the reduced risk of dyslipidemia due to the antagonism of these two exposures was 83% (95% CI: -1.66, -0.005). Our study first indicated that MCs exposure is an independent risk factor for dyslipidemia in a dose-response manner.
Collapse
Affiliation(s)
- Shuidong Feng
- Department of Epidemiology and Health Statistics, The Key Laboratory of Typical Environmental Pollution and Health Hazards of Hunan Province, School of Basic Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Mengyue Cao
- Department of Epidemiology and Health Statistics, The Key Laboratory of Typical Environmental Pollution and Health Hazards of Hunan Province, School of Basic Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Peng Tang
- Department of Epidemiology and Health Statistics, The Key Laboratory of Typical Environmental Pollution and Health Hazards of Hunan Province, School of Basic Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Shuxiang Deng
- Department of Epidemiology and Health Statistics, The Key Laboratory of Typical Environmental Pollution and Health Hazards of Hunan Province, School of Basic Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Limou Chen
- Department of Epidemiology and Health Statistics, The Key Laboratory of Typical Environmental Pollution and Health Hazards of Hunan Province, School of Basic Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yan Tang
- Department of Epidemiology and Health Statistics, The Key Laboratory of Typical Environmental Pollution and Health Hazards of Hunan Province, School of Basic Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Lemei Zhu
- School of Public Health, Changsha Medical University, Changsha 410219, China
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha 410008, China
- Furong Laboratory, Changsha 410008, China
| | - Zhijun Huang
- Furong Laboratory, Changsha 410008, China
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Minxue Shen
- Furong Laboratory, Changsha 410008, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Department of Social Medicine and Health Management, Central South University, Changsha 410000, China
| | - Fei Yang
- Department of Epidemiology and Health Statistics, The Key Laboratory of Typical Environmental Pollution and Health Hazards of Hunan Province, School of Basic Medicine, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Department of Social Medicine and Health Management, Central South University, Changsha 410000, China
| |
Collapse
|
36
|
Yang Y, Zheng S, Chu H, Du C, Chen M, Emran MY, Chen J, Yang F, Tian L. Subchronic Microcystin-LR Aggravates Colorectal Inflammatory Response and Barrier Disruption via Raf/ERK Signaling Pathway in Obese Mice. Toxins (Basel) 2023; 15:toxins15040262. [PMID: 37104200 PMCID: PMC10145857 DOI: 10.3390/toxins15040262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 04/05/2023] Open
Abstract
Microcystin-LR (MC-LR) is an extremely poisonous cyanotoxin that poses a threat to ecosystems and human health. MC-LR has been reported as an enterotoxin. The objective of this study was to determine the effect and the mechanism of subchronic MC-LR toxicity on preexisting diet-induced colorectal damage. C57BL/6J mice were given either a regular diet or a high-fat diet (HFD) for 8 weeks. After 8 weeks of feeding, animals were supplied with vehicle or 120 μg/L MC-LR via drinking water for another 8 weeks, and their colorectal were stained with H&E to detect microstructural alterations. Compared with the CT group, the HFD and MC-LR + HFD-treatment group induced a significant weight gain in the mice. Histopathological findings showed that the HFD- and MC-LR + HFD-treatment groups caused epithelial barrier disruption and infiltration of inflammatory cells. The HFD- and MC-LR + HFD-treatment groups raised the levels of inflammation mediator factors and decreased the expression of tight junction-related factors compared to the CT group. The expression levels of p-Raf/Raf and p-ERK/ERK in the HFD- and MC-LR + HFD-treatment groups were significantly increased compared with the CT group. Additionally, treated with MC-LR + HFD, the colorectal injury was further aggravated compared with the HFD-treatment group. These findings suggest that by stimulating the Raf/ERK signaling pathway, MC-LR may cause colorectal inflammation and barrier disruption. This study suggests that MC-LR treatment may exacerbate the colorectal toxicity caused by an HFD. These findings offer unique insights into the consequences and harmful mechanisms of MC-LR and provide strategies for preventing and treating intestinal disorders.
Collapse
Affiliation(s)
- Yue Yang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Shuilin Zheng
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, Department of Education, Hengyang Medical School, University of South China, Hengyang 421001, China
- Changsha Center for Disease Control and Prevention, Changsha 410004, China
| | - Hanyu Chu
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, Department of Education, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Can Du
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Mengshi Chen
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Mohammed Y. Emran
- National Institute for Materials Science (NIMS), 1-2-1 Sengen, Tsukuba 305-0047, Japan
| | - Jihua Chen
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Fei Yang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
- Hunan Province Key Laboratory of Typical Environmental Pollution and Health Hazards, The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, Department of Education, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Li Tian
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| |
Collapse
|