1
|
Tao J, Li H, Wang H, Tan J, Yang X. Metabolic dysfunction-associated fatty liver disease and osteoporosis: the mechanisms and roles of adiposity. Osteoporos Int 2024; 35:2087-2098. [PMID: 39136721 DOI: 10.1007/s00198-024-07217-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/26/2024] [Indexed: 11/21/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has recently been renamed metabolic dysfunction-associated fatty liver disease (MAFLD) by international consensus. Both MAFLD and osteoporosis are highly prevalent metabolic diseases. Recent evidence indicates that NAFLD increases the risk of low bone mineral density and osteoporosis, likely mediated by obesity. NAFLD has a close association with obesity and other metabolic disorders. Although obesity was previously thought to protect against bone loss, it now heightens osteoporotic fracture risk. This overview summarizes current clinical correlations between obesity, NAFLD, and osteoporosis, with a focus on recent insights into potential mechanisms interconnecting these three conditions. This study reviewed the scientific literature on the relationship between obesity, nonalcoholic fatty liver disease, and osteoporosis as well as the scientific literature that reveals the underlying pathophysiologic mechanisms between the three. Emerging evidence suggests obesity plays a key role in mediating the relationship between NAFLD and osteoporosis. Accumulating laboratory evidence supports plausible pathophysiological links between obesity, NAFLD, and osteoporosis, including inflammatory pathways, insulin resistance, gut microbiota dysbiosis, bone marrow adiposity, and alterations in insulin-like growth factor-1 signaling. Adiposity has important associations with NAFLD and osteoporosis, the underlying pathophysiologic mechanisms between the three may provide new therapeutic targets for this complex patient population.
Collapse
Affiliation(s)
- Jie Tao
- Department of General Practice, the Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Hong Li
- Department of Health Management Center, the Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Honggang Wang
- Department of Gastroenterology, the Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Juan Tan
- Department of General Practice, the Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu Province, China.
| | - Xiaozhong Yang
- Department of Gastroenterology, the Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu Province, China.
| |
Collapse
|
2
|
Ge H, Qi F, Shen Z, Wang H, Zhu S, Zhou S, Xie Z, Li D. Large-leaf yellow tea protein derived-peptides alleviated dextran sodium sulfate-induced acute colitis and restored intestinal microbiota balance in C57BL/6 J mice. Food Chem 2024; 456:139936. [PMID: 38865822 DOI: 10.1016/j.foodchem.2024.139936] [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: 03/17/2024] [Revised: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 06/14/2024]
Abstract
Large-leaf yellow tea (LYT)-derived peptides (TPP) are rich in amino acids required for damage repair, such as Glu, Arg, and Pro, and can be used to alleviate acute colitis. However, its effect and mechanisms against colitis remain unclear. This study utilized TPP to intervene in dextran sodium sulfate-induced acute colitis in C57BL/6 J mice. Results confirmed that TPP ameliorated acute colitis symptoms by inhibiting pro-inflammatory cytokines, restoring gut microbiota dysbiosis, particularly by increasing the abundance of beneficial bacteria Akkermansia and Lactobacillus while declining harmful microbiota Escherichia-Shigella. Besides, TPP intervention reshaped the gut microbiota phenotype by increasing the aerobic phenotype and reducing the potentially pathogenic phenotype. Levels of short-chain fatty acids, including acetic acid, propanoic acid, isobutyric acid, and butyric acid, were also enhanced in a dose-dependent manner to help restore gut microbiota equilibrium. This study supports using TPP as a viable plant protein-derived dietary resource for alleviating inflammatory bowel disease.
Collapse
Affiliation(s)
- Huifang Ge
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, People's Republic of China; Joint Research Center for Food Nutrition and Health of IHM, Hefei, Anhui 230036, People's Republic of China
| | - Fengxue Qi
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, People's Republic of China
| | - Ziyi Shen
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, People's Republic of China
| | - Hongyan Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, People's Republic of China; Joint Research Center for Food Nutrition and Health of IHM, Hefei, Anhui 230036, People's Republic of China
| | - Shangliang Zhu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, People's Republic of China
| | - Simeng Zhou
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, People's Republic of China
| | - Zhongwen Xie
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, People's Republic of China; Joint Research Center for Food Nutrition and Health of IHM, Hefei, Anhui 230036, People's Republic of China.
| | - Daxiang Li
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, People's Republic of China; Joint Research Center for Food Nutrition and Health of IHM, Hefei, Anhui 230036, People's Republic of China.
| |
Collapse
|
3
|
Xu J, Xie L, Fan R, Shi X, Xu W, Dong K, Ma D, Yan Y, Zhang S, Sun N, Huang G, Gao M, Yu X, Wang M, Wang F, Chen J, Tao J, Yang Y. The role of dietary inflammatory index in metabolic diseases: the associations, mechanisms, and treatments. Eur J Clin Nutr 2024:10.1038/s41430-024-01525-6. [PMID: 39433856 DOI: 10.1038/s41430-024-01525-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
In recent years, the prevalence of metabolic diseases has increased significantly, posing a serious threat to global health. Chronic low-grade inflammation is implicated in the development of most metabolic diseases, such as type 2 diabetes mellitus (T2DM), obesity, dyslipidemia, and cardiovascular disease, serving as a link between diet and these conditions. Increasing attention has been directly toward dietary inflammatory patterns that may prevent or ameliorate metabolic diseases. The Dietary Inflammatory Index (DII) was developed to assess the inflammatory potential of dietary intake. Consequently, a growing body of research has examined the associations between the DII and the risk of several metabolic diseases. In this review, we explore the current scientific literature on the relationships between the DII, T2DM, obesity, and dyslipidemia. It summarizes recent findings and explore potential underlying mechanisms from two aspects: the interaction between diet and inflammation, and the link between inflammation and metabolic diseases. Furthermore, this review discusses the therapeutic strategies, including dietary modifications, prebiotics, and probiotics, and discusses the application of the DII in metabolic diseases, as well as future research directions.
Collapse
Affiliation(s)
- Jialu Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Lei Xie
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Rongping Fan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Xiaoli Shi
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Weijie Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Kun Dong
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Delin Ma
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Yongli Yan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Shujun Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Nan Sun
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guomin Huang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Min Gao
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuefeng Yu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Mei Wang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Fen Wang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Juan Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Tao
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China.
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China.
| |
Collapse
|
4
|
Sasidharan Pillai S, Gagnon CA, Foster C, Ashraf AP. Exploring the Gut Microbiota: Key Insights Into Its Role in Obesity, Metabolic Syndrome, and Type 2 Diabetes. J Clin Endocrinol Metab 2024; 109:2709-2719. [PMID: 39040013 PMCID: PMC11479700 DOI: 10.1210/clinem/dgae499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/22/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
The gut microbiota (GM), comprising trillions of microorganisms in the gastrointestinal tract, is a key player in the development of obesity and related metabolic disorders, such as type 2 diabetes (T2D), metabolic syndrome (MS), and cardiovascular diseases. This mini-review delves into the intricate roles and mechanisms of the GM in these conditions, offering insights into potential therapeutic strategies targeting the microbiota. The review elucidates the diversity and development of the human GM, highlighting its pivotal functions in host physiology, including nutrient absorption, immune regulation, and energy metabolism. Studies show that GM dysbiosis is linked to increased energy extraction, altered metabolic pathways, and inflammation, contributing to obesity, MS, and T2D. The interplay between dietary habits and GM composition is explored, underscoring the influence of diet on microbial diversity and metabolic functions. Additionally, the review addresses the impact of common medications and therapeutic interventions like fecal microbiota transplantation on GM composition. The evidence so far advocates for further research to delineate the therapeutic potential of GM modulation in mitigating obesity and metabolic diseases, emphasizing the necessity of clinical trials to establish effective and sustainable treatment protocols.
Collapse
Affiliation(s)
- Sabitha Sasidharan Pillai
- Center for Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Charles A Gagnon
- University of Alabama at Birmingham Marnix E. Heersink School of Medicine, Birmingham, AL 35294, USA
| | - Christy Foster
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ambika P Ashraf
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
5
|
Lei M, Li Y, Li J, Liu J, Dai Z, Chen R, Zhu H. Low Testosterone and High Leptin Activate PPAR Signaling to Induce Adipogenesis and Promote Fat Deposition in Caponized Ganders. Int J Mol Sci 2024; 25:8686. [PMID: 39201373 PMCID: PMC11354323 DOI: 10.3390/ijms25168686] [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: 06/18/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Low or insufficient testosterone levels caused by caponization promote fat deposition in animals. However, the molecular mechanism of fat deposition in caponized animals remains unclear. This study aimed to investigate the metabolomics and transcriptomic profiles of adipose tissues and study the effect of testosterone and leptin on the proliferation of adipocytes. We observed a significant enlargement in the areas of adipocytes in the abdominal fat tissues in capon, as well as increased luciferase activity of the serum leptin and a sharp decrease in the serum testosterone in caponized gander. Metabolomics and transcriptomic results revealed differentially expressed genes and differentially expressed metabolites with enhanced PARR signal pathway. The mRNA levels of peroxisome proliferators-activated receptor γ, fatty acid synthase, and suppressor of cytokine signaling 3 in goose primary pre-adipocytes were significantly upregulated with high leptin treatment and decreased significantly with increasing testosterone dose. Hence, reduced testosterone and increased leptin levels after caponization possibly promoted adipocytes proliferation and abdominal fat deposition by altering the expression of PPAR pathway related genes in caponized ganders. This study provides a new direction for the mechanism through which testosterone regulates the biological function of leptin and fat deposition in male animals.
Collapse
Affiliation(s)
- Mingming Lei
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (Y.L.); (J.L.); (J.L.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Jiangsu Province Engineering Research Center of Precision Animal Breeding, Nanjing 210014, China
| | - Yaxin Li
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (Y.L.); (J.L.); (J.L.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Jiangsu Province Engineering Research Center of Precision Animal Breeding, Nanjing 210014, China
| | - Jiaying Li
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (Y.L.); (J.L.); (J.L.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Jiangsu Province Engineering Research Center of Precision Animal Breeding, Nanjing 210014, China
| | - Jie Liu
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (Y.L.); (J.L.); (J.L.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Jiangsu Province Engineering Research Center of Precision Animal Breeding, Nanjing 210014, China
| | - Zichun Dai
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (Y.L.); (J.L.); (J.L.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Jiangsu Province Engineering Research Center of Precision Animal Breeding, Nanjing 210014, China
| | - Rong Chen
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (Y.L.); (J.L.); (J.L.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Jiangsu Province Engineering Research Center of Precision Animal Breeding, Nanjing 210014, China
| | - Huanxi Zhu
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (M.L.); (Y.L.); (J.L.); (J.L.); (Z.D.); (R.C.)
- Key Laboratory of Crop and Livestock Integration, Ministry of Agriculture, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Jiangsu Province Engineering Research Center of Precision Animal Breeding, Nanjing 210014, China
| |
Collapse
|
6
|
Zhu A, Luo N, Sun L, Zhou X, Chen S, Huang Z, Mao X, Li K. Mulberry and Hippophae-based solid beverage attenuate hyperlipidemia and hepatic steatosis via adipose tissue-liver axis. Food Sci Nutr 2024; 12:5052-5064. [PMID: 39055214 PMCID: PMC11266884 DOI: 10.1002/fsn3.4155] [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: 11/02/2023] [Revised: 03/17/2024] [Accepted: 03/23/2024] [Indexed: 07/27/2024] Open
Abstract
Dyslipidemia and hepatic steatosis are the characteristics of the initial stage of nonalcohol fatty liver disease (NAFLD), which can be reversed by lifestyle intervention, including dietary supplementation. However, such commercial dietary supplements with solid scientific evidence and in particular clear mechanistic elucidation are scarce. Here, the health benefits of MHP, a commercial mulberry and Hippophae-based solid beverage, were evaluated in NAFLD rat model and the underlying molecular mechanisms were investigated. Histopathologic examination of liver and white adipose tissue found that MHP supplementation reduced hepatic lipid accumulation and adipocyte hypertrophy. Serum biochemical results confirmed that MHP effectively ameliorated dyslipidemia and decreased circulation-free fatty acid level. RNA-Seq-based transcriptomic analysis showed that MHP-regulated genes are involved in the inhibition of lipolysis of adipose tissue and thus may contribute to the reduction of hepatic ectopic lipid deposition. Furthermore, MHP upregulated ACSL1-CPT1a-CPT2 pathway, a canonical pathway that regulated mitochondrial fatty acid metabolism, and promoted liver and adipose tissue fatty acid β-oxidation. These results suggest that adipose tissue-liver crosstalk may play a key role in maintaining glucose and lipid metabolic hemostasis. In addition, MHP can also ameliorate chronic inflammation through regulating the secretion of adipokines. Our study demonstrates that MHP is able to improve dyslipidemia and hepatic steatosis through crosstalk between adipose tissue and liver and also presents transcriptomic evidence to support the underlying mechanisms of action, providing solid evidence for its health claims.
Collapse
Affiliation(s)
- An‐Qi Zhu
- Institute of Chinese Medicinal SciencesGuangdong Pharmaceutical UniversityGuangzhouChina
| | - Nin Luo
- Institute of Chinese Medicinal SciencesGuangdong Pharmaceutical UniversityGuangzhouChina
| | - Ling‐Yue Sun
- Institute of Chinese Medicinal SciencesGuangdong Pharmaceutical UniversityGuangzhouChina
| | - Xiao‐Ting Zhou
- Institute of Chinese Medicinal SciencesGuangdong Pharmaceutical UniversityGuangzhouChina
| | - Shi‐Sheng Chen
- Perfect Life & Health InstituteZhongshanGuangdongChina
- Perfect (Guangdong) Co., Ltd.ZhongshanChina
| | - Zebo Huang
- School of Food Science and EngineeringSouth China University of TechnologyGuangzhouChina
| | - Xin‐Liang Mao
- Perfect Life & Health InstituteZhongshanGuangdongChina
- Perfect (Guangdong) Co., Ltd.ZhongshanChina
| | - Kun‐Ping Li
- Institute of Chinese Medicinal SciencesGuangdong Pharmaceutical UniversityGuangzhouChina
| |
Collapse
|
7
|
Li Y, Qi X, Wang Q, He Y, Li Z, Cen X, Wei L. Comprehensive analysis of key host gene-microbe networks in the cecum tissues of the obese rabbits induced by a high-fat diet. Front Cell Infect Microbiol 2024; 14:1407051. [PMID: 38947127 PMCID: PMC11211605 DOI: 10.3389/fcimb.2024.1407051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/24/2024] [Indexed: 07/02/2024] Open
Abstract
The Cecum is a key site for cellulose digestion in nutrient metabolism of intestine, but its mechanisms of microbial and gene interactions has not been fully elucidated during pathogenesis of obesity. Therefore, the cecum tissues of the New Zealand rabbits and their contents between the high-fat diet-induced group (Ob) and control group (Co) were collected and analyzed using multi-omics. The metagenomic analysis indicated that the relative abundances of Corallococcus_sp._CAG:1435 and Flavobacteriales bacterium species were significantly lower, while those of Akkermansia glycaniphila, Clostridium_sp._CAG:793, Mycoplasma_sp._CAG:776, Mycoplasma_sp._CAG:472, Clostridium_sp._CAG:609, Akkermansia_sp._KLE1605, Clostridium_sp._CAG:508, and Firmicutes_bacterium_CAG:460 species were significantly higher in the Ob as compared to those in Co. Transcriptomic sequencing results showed that the differentially upregulated genes were mainly enriched in pathways, including calcium signaling pathway, PI3K-Akt signaling pathway, and Wnt signaling pathway, while the differentially downregulated genes were mainly enriched in pathways of NF-kappaB signaling pathway and T cell receptor signaling pathway. The comparative analysis of metabolites showed that the glycine, serine, and threonine metabolism and cysteine and methionine metabolism were the important metabolic pathways between the two groups. The combined analysis showed that CAMK1, IGFBP6, and IGFBP4 genes were highly correlated with Clostridium_sp._CAG:793, and Akkermansia_glycaniphila species. Thus, the preliminary study elucidated the microbial and gene interactions in cecum of obese rabbit and provided a basis for further studies in intestinal intervention for human obesity.
Collapse
Affiliation(s)
- Yanhong Li
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, Guizhou, China
| | - Qinrong Wang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yan He
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhupeng Li
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xi Cen
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang, Guizhou, China
| | - Limin Wei
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing, China
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Zhu AQ, Luo N, Zhou XT, Yuan M, Zhang CM, Pan TL, Li KP. Transcriptomic insights into the lipotoxicity of high-fat high-fructose diet in rat and mouse. J Nutr Biochem 2024; 128:109626. [PMID: 38527560 DOI: 10.1016/j.jnutbio.2024.109626] [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: 09/05/2023] [Revised: 02/23/2024] [Accepted: 03/21/2024] [Indexed: 03/27/2024]
Abstract
Along with the increasing prevalence of obesity worldwide, the deleterious effects of high-calorie diet are gradually recognized through more and more epidemiological studies. However, the concealed and chronic causality whitewashes its unhealthy character. Given an ingenious mechanism orchestrates the metabolic adaptation to high-fat high-fructose (HFF) diet and connive its lipotoxicity, in this study, an experimental rat/mouse model of obesity was induced and a comparative transcriptomic analysis was performed to probe the mystery. Our results demonstrated that HFF diet consumption altered the transcriptomic pattern as well as different high-calorie diet fed rat/mouse manifested distinct hepatic transcriptome. Validation with RT-qPCR and Western blotting confirmed that SREBP1-FASN involved in de novo lipogenesis partly mediated metabolic self-adaption. Moreover, hepatic ACSL1-CPT1A-CPT2 pathway involved in fatty acids β-oxidation, played a key role in the metabolic adaption to HFF. Collectively, our findings enrich the knowledge of the chronic adaptation mechanisms and also shed light on future investigations. Meanwhile, our results also suggest that efforts to restore the fatty acids metabolic fate could be a promising avenue to fight against obesity and associated steatosis and insulin resistance challenged by HFF diet.
Collapse
Affiliation(s)
- An-Qi Zhu
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ning Luo
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiao-Ting Zhou
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Min Yuan
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chu-Mei Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tian-Ling Pan
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China
| | - Kun-Ping Li
- Institute of Chinese Medicinal Sciences, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, China.; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China.
| |
Collapse
|
9
|
Xu J, Zhao Y, Tyler Mertens R, Ding Y, Xiao P. Sweet regulation - The emerging immunoregulatory roles of hexoses. J Adv Res 2024:S2090-1232(24)00157-7. [PMID: 38631430 DOI: 10.1016/j.jare.2024.04.014] [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: 10/04/2023] [Revised: 03/20/2024] [Accepted: 04/13/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND It is widely acknowledged that dietary habits have profound impacts on human health and diseases. As the most important sweeteners and energy sources in human diets, hexoses take part in a broad range of physiopathological processes. In recent years, emerging evidence has uncovered the crucial roles of hexoses, such as glucose, fructose, mannose, and galactose, in controlling the differentiation or function of immune cells. AIM OF REVIEW Herein, we reviewed the latest research progresses in the hexose-mediated modulation of immune responses, provided in-depth analyses of the underlying mechanisms, and discussed the unresolved issues in this field. KEY SCIENTIFIC CONCEPTS OF REVIEW Owing to their immunoregulatory effects, hexoses affect the onset and progression of various types of immune disorders, including inflammatory diseases, autoimmune diseases, and tumor immune evasion. Thus, targeting hexose metabolism is becoming a promising strategy for reversing immune abnormalities in diseases.
Collapse
Affiliation(s)
- Junjie Xu
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuening Zhao
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Yimin Ding
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Xiao
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China; The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
10
|
Zhou XT, Zhu AQ, Li XM, Sun LY, Yan JG, Luo N, Chen SS, Huang Z, Mao XL, Li KP. Mulberry and Hippophae-based solid beverage promotes weight loss in rats by antagonizing white adipose tissue PPARγ and FGFR1 signaling. Front Endocrinol (Lausanne) 2024; 15:1344262. [PMID: 38559696 PMCID: PMC10978776 DOI: 10.3389/fendo.2024.1344262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Obesity, a multifactorial disease with many complications, has become a global epidemic. Weight management, including dietary supplementation, has been confirmed to provide relevant health benefits. However, experimental evidence and mechanistic elucidation of dietary supplements in this regard are limited. Here, the weight loss efficacy of MHP, a commercial solid beverage consisting of mulberry leaf aqueous extract and Hippophae protein peptides, was evaluated in a high-fat high-fructose (HFF) diet-induced rat model of obesity. Body component analysis and histopathologic examination confirmed that MHP was effective to facilitate weight loss and adiposity decrease. Pathway enrichment analysis with differential metabolites generated by serum metabolomic profiling suggests that PPAR signal pathway was significantly altered when the rats were challenged by HFF diet but it was rectified after MHP intervention. RNA-Seq based transcriptome data also indicates that MHP intervention rectified the alterations of white adipose tissue mRNA expressions in HFF-induced obese rats. Integrated omics reveals that the efficacy of MHP against obesogenic adipogenesis was potentially associated with its regulation of PPARγ and FGFR1 signaling pathway. Collectively, our findings suggest that MHP could improve obesity, providing an insight into the use of MHP in body weight management.
Collapse
Affiliation(s)
- Xiao-Ting Zhou
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - An-Qi Zhu
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiao-Min Li
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Ling-Yue Sun
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jian-Gang Yan
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Nin Luo
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shi-Sheng Chen
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Zebo Huang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Xin-Liang Mao
- Research & Development Division, Perfect Life & Health Institute, Zhongshan, China
- Research & Development Division, Perfect (Guangdong) Co., Ltd., Zhongshan, China
| | - Kun-Ping Li
- Key Laboratory of Glucolipid Metabolic Disorders, Ministry of Education of China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
11
|
Mustika S, Santosaningsih D, Handayani D, Rudijanto A. Impact of multiple different high-fat diets on metabolism, inflammatory markers, dysbiosis, and liver histology: study on NASH rat model induced diet. F1000Res 2023; 12:180. [PMID: 39229607 PMCID: PMC11369591 DOI: 10.12688/f1000research.129645.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 09/05/2024] Open
Abstract
Background The spectrum of non-alcoholic fatty liver disease (NAFLD), known as non-alcoholic steatohepatitis (NASH), can lead to advanced liver disease. It is known that a variety of diets play a significant role in the development of NAFLD/NASH. The goal of this study was to determine the most appropriate composition of diet to induce NASH in an animal model. Methods This research used Rattus norvegicus strain Wistar (n=27), which were divided into four groups and given each diet for 12 weeks: normal diet (ND, n=7), high-fat diet (HFD, n=6), western diet (WD, n=7), and high-fat-high-fructose diet (HFHFD, n=7). Subjects were monitored for changes in body weight. Blood samples were collected for biochemical analysis, including low-density lipoprotein (LDL), triglyceride, alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), hepatic lipase, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and lipopolysaccharide (LPS). Fecal samples were taken for short-chain fatty acid (SCFA) analysis. Liver histology was assessed using NAS (NAFLD activity score). A statistical comparison test was carried out using the one-way ANOVA or Kruskal-Wallis test. Results The highest average body weight was observed in the WD group (346.14 g). Liver enzymes, LDL, triglyceride, propionic acid, and acetic acid did not show significantly differences among the groups. TNF-α, IL-6, and hepatic lipase were significant (p = 0.000; p = 0.000; p = 0.004) and the highest level recorded in the HFD group. Butyrate acid level also showed significances (p = 0.021) with the lowest concentration seen in the HFHFD group (4.77 mMol/g). Only WD and HFHFD had a NAS ≥ 5 (14% and 14%). The highest percentage of borderline NAS was found in WD (57%). Conclusions WD feeding is the most appropriate diet type to induce NASH in rats as it influences metabolic, inflammatory, dysbiosis, and liver histology of rats.
Collapse
Affiliation(s)
- Syifa Mustika
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Jl. Veteran, 65145, Indonesia
| | - Dewi Santosaningsih
- Department of Clinical Microbiology, Faculty of Medicine, Universitas Brawijaya, Malang, Jl. Veteran, 65145, Indonesia
| | - Dian Handayani
- Department of Nutrition, Faculty of Health Science, Universitas Brawijaya, Malang, Jl. Veteran, 65145, Indonesia
| | - Achmad Rudijanto
- Endocrine Metabolic & Diabetes Division, Department of Internal Medicine, Faculty of Medicine, Universitas Brawijaya - Dr Saiful Anwar Hospital, Malang, Jl. Veteran, 65145, Indonesia
| |
Collapse
|
12
|
Wang Z, Li Q, Huang H, Liu J, Wang J, Chen Y, Huang S, Luo X, Zheng Z. Distribution and potential ecological risks of microplastics in Zhushan Bay, China. CHEMOSPHERE 2023:139024. [PMID: 37247671 DOI: 10.1016/j.chemosphere.2023.139024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 05/31/2023]
Abstract
The interaction between microplastics (MPs) and microorganisms may alter the distribution of antibiotic resistance genes (ARGs) in water and increase the ecological risk of drinking water sources. To investigate the characteristics of MPs geographical distribution and its potential ecological risk in typical urban water, this study was conducted in Zhushan Bay, and we carried out a combination of tests to analyze the distribution of MPs and the migration changes of their surface microbial community composition and ARGs in different media by 16S rRNA gene high-throughput sequencing, non-targeted metabolomics and qPCR genomics in the near-shore (I), middle area (Ⅱ) and near-lake (Ⅲ) of Zhushan Bay. The results showed that MPs in fibrous form were dominant in the aquatic environment of Zhushan Bay; Polyurethane (PU) and Silicone were the main MPs types in Zhushan Bay. The abundance of MPs in the water of Zhushan Bay was winter > summer > autumn > spring; and in the sediment was winter > summer > autumn > spring, respectively. The distribution results of MPs in geographical location are as follows: In the water I > Ⅱ > Ⅲ, sediment exhibited Ⅱ > Ⅲ > I. The results indicate that physicochemical factors will affect the geographical distribution of MPs and their surface microbial community composition in the aquatic environment of Zhushan Bay. More cooperative behaviors and increased metabolically important pathways occurred in the microbial network on water-MPs compared to sediment-MPs. However, the microbial community in the sediment-MPs was more stable and had higher abundance of mobile genetic elements (MGEs). A total of 362 differential metabolites were detected, of which 193 were up-regulated and 19 down-regulated differential metabolites. blaTEM, Sul, and inti1 were prevalent in both the water and sediments of Zhushan Bay. Sul1 was most contaminated in ARGs. This study provides the latest field data and insights into MPs pollution in key aquatic environments.
Collapse
Affiliation(s)
- Zhikai Wang
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Qihui Li
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China; School of Ecological and Environment, Chengdu University of Technology, Chengdu, 610059, China
| | - Haiqing Huang
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Jing Liu
- School of Ecological and Environment, Chengdu University of Technology, Chengdu, 610059, China
| | - Jie Wang
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Yican Chen
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Suzhen Huang
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Xingzhang Luo
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China
| | - Zheng Zheng
- Department of Environmental Science and Engineering, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
13
|
Liu J, Ding H, Yan C, He Z, Zhu H, Ma KY. Effect of tea catechins on gut microbiota in high fat diet-induced obese mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:2436-2445. [PMID: 36715435 DOI: 10.1002/jsfa.12476] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/06/2023] [Accepted: 01/30/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Tea catechins have been shown to have beneficial effects on the alleviation of obesity, the prevention of diabetes, and the amelioration of metabolic syndrome. The purpose of the present work is to explore the underlying mechanisms linking the intestinal microbiota and anti-obesity benefits of green tea, oolong tea, and black tea catechins in C57BL/6J mice fed with a high-fat diet (HFD). RESULTS The results indicated that, after the dietary intake of three tea catechins, obesity and low-grade inflammation were significantly alleviated. Hepatic steatosis was prevented, and this was accompanied by the upregulation of the mRNA and protein expressions of hepatic peroxisome proliferator-activated receptor α (PPARα). Metagenomic analysis of fecal samples suggested that the three tea catechins similarly changed the microbiota in terms of overall structure, composition, and protein functions by regulating the metabolites, facilitating the generation of short-chain fatty acids (SCFAs), and repressing lipopolysaccharides. CONCLUSION The anti-obese properties of three tea catechins were partially mediated by their positive effect on gut microbiota, hepatic steatosis alleviation, and anti-inflammatory activity. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jianhui Liu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Huafang Ding
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Chi Yan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, China
| | - Zouyan He
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, China
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Hanyue Zhu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, China
- School of Food Science and Engineering / South China Food Safety Research Center, Foshan University, Foshan, China
| | - Ka Ying Ma
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, China
| |
Collapse
|
14
|
Xu Y, Huang X, Huangfu B, Hu Y, Xu J, Gao R, Huang K, He X. Sulforaphane Ameliorates Nonalcoholic Fatty Liver Disease Induced by High-Fat and High-Fructose Diet via LPS/TLR4 in the Gut-Liver Axis. Nutrients 2023; 15:nu15030743. [PMID: 36771448 PMCID: PMC9920698 DOI: 10.3390/nu15030743] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
The gut-liver axis has emerged as a key player in the progression of non-alcoholic fatty liver disease (NAFLD). Sulforaphane (SFN) is a bioactive compound found in cruciferous vegetables; however, it has not been reported whether SFN improves NAFLD via the gut-liver axis. C57BL/6 mice were fed a high-fat and high-fructose (HFHFr) diet, with or without SFN gavage at doses of 15 and 30 mg·kg-1 body weight for 12 weeks. The results showed that SFN reduced weight gain, hepatic inflammation, and steatosis in HFHFr mice. SFN altered the composition of gut microbes. Moreover, SFN enhanced the intestinal tight junction protein ZO-1, reduced serum LPS, and inhibited LPS/TLR4 and ERS pathways to reduce intestinal inflammation. As a result, SFN protected the intestinal integrity and declined the gut-derived LPS translocations to the liver in HFHFr diet-induced mice. SFN decreased the liver LPS levels and inhibited the LPS/TLR4 pathway activations, thus inhibiting the pro-inflammatory cytokines. Notably, Spearman correlation analysis showed that the protective effect of SFN on intestinal barrier integrity and its anti-inflammatory effect on the liver was associated with improved intestinal dysbiosis. Above all, dietary intervention with SFN attenuates NAFLD through the gut-liver axis.
Collapse
Affiliation(s)
- Ye Xu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xianghui Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Henan Shuanghui Investment and Development Co., Ltd., Luohe 462000, China
| | - Bingxin Huangfu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yanzhou Hu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Jia Xu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ruxin Gao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
| | - Xiaoyun He
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
- Correspondence:
| |
Collapse
|
15
|
Zou T, Xie F, Liang P, Chen J, Wang Z, Du M, You J. Polysaccharide-rich fractions from Enteromorpha prolifera improve hepatic steatosis and gut barrier integrity in high-fat diet-induced obese mice linking to modulation of gut microbiota. Biomed Pharmacother 2023; 157:114034. [PMID: 36434956 DOI: 10.1016/j.biopha.2022.114034] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
Polysaccharides from Enteromorpha prolifera (EP) possess important benefits in the management of obesity and associated metabolic diseases, but to date, the underlying mechanism linking this alleviative effect of EP to gut microbiota remains obscure. This study aimed to investigate the effects of EP in improving lipid metabolism disorders and intestinal barrier disruption in mice with high-fat diet (HFD), and its association with modulation of gut microbiota. C57BL/6 mice were fed a control diet or a HFD with or without 5% EP for 12 weeks. Factors related to lipid metabolism, insulin signaling and intestinal barrier integrity, as well as the involvement of gut microbiota and metabolites, were measured. EP supplementation reduced HFD-induced adiposity and mitigated insulin resistance, hepatic steatosis and elevation of serum lipopolysaccharides (LPS). HFD impaired intestinal barrier integrity while improved due to EP. Moreover, EP administration ameliorated HFD-induced gut dysbiosis, as revealed by the increased short-chain fatty acid (SCFA)-producing bacteria (e.g., Bacteroides, Parabacteroides, Alloprevotella, and Ruminococcus) and gut barrier-protective Akkermansia muciniphila and decreased endotoxin-producing bacteria (e.g., Desulfovibrionaceae and Bilophila), accompanied by enrichment in intestinal SCFA content and reduction in circulating LPS level. The change of dominant bacterial genera is significantly correlated with improved metabolic profiles and intestinal permeability induced by EP. In conclusion, our results indicate that EP can attenuate HFD-induced metabolic disorders along with restoration of gut barrier integrity and lowering of circulating endotoxin, and these improvements are associated with modulation of gut microbiota composition and related metabolites. These data deepen mechanistic understanding of the anti-obesity and metabolic improving effects of EP.
Collapse
Affiliation(s)
- Tiande Zou
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, Jiangxi, China
| | - Fei Xie
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, Jiangxi, China
| | - Pengbo Liang
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, Jiangxi, China
| | - Jun Chen
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, Jiangxi, China
| | - Zirui Wang
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, Jiangxi, China
| | - Min Du
- Laboratory of Nutrigenomics and Growth Biology, Department of Animal Sciences, Washington State University, Pullman, WA 99164, USA
| | - Jinming You
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang 330045, Jiangxi, China.
| |
Collapse
|
16
|
Zhang Y, Liu Y, Ma H, Sun M, Wang X, Jin S, Yuan X. Insufficient or excessive dietary carbohydrates affect gut health through change in gut microbiota and regulation of gene expression of gut epithelial cells in grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2023; 132:108442. [PMID: 36410648 DOI: 10.1016/j.fsi.2022.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 06/16/2023]
Abstract
Dietary carbohydrate levels can affect gut health, but the roles played by gut microbiota and gut epithelial cells, and their interactions remain unclear. In this experiment, we investigated gut health, gut microbiota, and the gene expression profiles of gut epithelial cells in grass carp consuming diets with different carbohydrate levels. Compared to the moderate-carbohydrate diet, low-carbohydrate diet significantly increased the relative abundance of pathogenic bacteria (Ralstonia and Elizabethkingia) and decreased the abundance of metabolism in cofactors and vitamins, implying a dysregulated gut microbiota and compromised metabolic function. Moreover, low-carbohydrate diet inhibited the expression levels of key genes in autophagy-related pathways in gut epithelial cells, which might directly lead to reduced clearance of defective organelles and pathogenic microorganisms. These aforementioned factors may be responsible for the imperfect organization of the intestinal tract. High-carbohydrate diet also significantly increased the abundance of pathogenic bacteria (Flavobacterium), which directly contributed to a decrease in the abundance of immune system of the microbiota. Furthermore, the active pathways of staphylococcus aureus infection and complement and coagulation cascades, as well as the inhibition of the glutathione metabolism pathway were observed. Above results implied that high-carbohydrate diet might ultimately cause severe gut damage by affecting immune function of microbiota, mentioned immune-related pathways, and the antioxidant capacity. Finally, the correlation network diagram revealed strong correlations of the differentially immune-related gene major histocompatibility complex class I antigen (MR1) with Enhydrobacter and Ruminococcus_gnavus_group in low-carbohydrate diet group, and Arenimonas in high-carbohydrate diet group, respectively, suggesting that MR1 might be a central target for immune responses in gut epithelial cells induced by gut microbiota at different levels of dietary carbohydrate. All these results provided insight in the development of antagonistic probiotics and target genes to improve the utilization of carbohydrate.
Collapse
Affiliation(s)
- Yanpeng Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Yucheng Liu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Huan Ma
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Manjie Sun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Xin Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Shengzhen Jin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, China
| | - Xiaochen Yuan
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, China.
| |
Collapse
|
17
|
Shi J, Qiu H, Xu Q, Ma Y, Ye T, Kuang Z, Qu N, Kan C, Hou N, Han F, Sun X. Integrated multi-omics analyses reveal effects of empagliflozin on intestinal homeostasis in high-fat-diet mice. iScience 2022; 26:105816. [PMID: 36636340 PMCID: PMC9830204 DOI: 10.1016/j.isci.2022.105816] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/25/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Obesity has become a global epidemic, associated with several chronic complications. The intestinal microbiome is a critical regulator of metabolic homeostasis and obesity. Empagliflozin, a sodium-glucose cotransporter 2 (SGLT2) inhibitor, has putative anti-obesity effects. In this study, we used multi-omics analysis to determine whether empagliflozin regulates metabolism in an obese host through the intestinal microbiota. Compared with obese mice, the empagliflozin-treated mice had a higher species diversity of gut microbiota, characterized by a reduction in the Firmicutes/Bacteroides ratio. Metabolomic analysis unambiguously identified 1,065 small molecules with empagliflozin affecting metabolites mainly enriched in amino acid metabolism, such as tryptophan metabolism. RNA sequencing results showed that immunoglobulin A and peroxisome proliferator-activated receptor signaling pathways in the intestinal immune network were activated after empagliflozin treatment. This integrative analysis highlighted that empagliflozin maintains intestinal homeostasis by modulating gut microbiota diversity and tryptophan metabolism. This will inform the development of therapies for obesity based on host-microbe interactions.
Collapse
Affiliation(s)
- Junfeng Shi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Hongyan Qiu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Qian Xu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yuting Ma
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Tongtong Ye
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zengguang Kuang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Na Qu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China,Department of Pathology, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Corresponding author
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, 2428 Yuhe Road, Weifang, Shandong 261031, China,Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China,Corresponding author
| |
Collapse
|
18
|
Purification Process and In Vitro and In Vivo Bioactivity Evaluation of Pectolinarin and Linarin from Cirsium japonicum. Molecules 2022; 27:molecules27248695. [PMID: 36557828 PMCID: PMC9780979 DOI: 10.3390/molecules27248695] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Pectolinarin and linarin are two major flavone O-glycosides of Cirsium japonicum, which has been used for thousands of years in traditional Chinese medicine. Pharmacological research on pectolinarin and linarin is meaningful and necessary. Here, a process for the purification of pectolinarin and linarin from C. japonicum was established using macroporous resin enrichment followed by prep-HPLC separation. The results show the purity of pectolinarin and linarin reached 97.39% and 96.65%, respectively. The in vitro bioactivities result shows the ORAC values of pectolinarin and linarin are 4543 and 1441 µmol TE/g, respectively, meanwhile their inhibition rate of BSA-MGO-derived AGEs is 63.58% and 19.31% at 2 mg/mL, which is 56.03% and 30.73% in the BSA-fructose system, respectively. The COX-2 inhibition rate at 50 µg/mL of linarin and pectolinarin reached 55.35% and 40.40%, respectively. Furthermore, the in vivo bioassay combining of histopathologic evaluation and biochemical analysis of liver glutamic oxaloacetic transaminase, serum creatinine and TNF-α show pectolinarin can alleviate lipopolysaccharide (LPS)-induced acute liver and kidney injury in mice. Metabolomics analysis shows that pectolinarin attenuates LPS-challenged liver and kidney stress through regulating the arachidonic acid metabolism and glutathione synthesis pathways. Collectively, our work presents a solid process for pectolinarin and linarin purification and has discovered a promising natural therapeutic agent-pectolinarin.
Collapse
|
19
|
Jia W, Wu X, Shi L. Hydrocortisone-Containing Animal-Derived Food Intake Affects Lipid Nutrients Utilization. Mol Nutr Food Res 2022; 66:e2200487. [PMID: 36261391 DOI: 10.1002/mnfr.202200487] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/27/2022] [Indexed: 01/18/2023]
Abstract
SCOPE As the tremendous increases in consumption of animal-derived food, endogenous hydrocortisone migrating along the food chain to organism arouses extensive attention. This study aims to investigate the cumulative impacts of dietary hydrocortisone intake and mechanistic understanding on metabolism of lipid nutrients. METHODS AND RESULTS A total of 120 porcine muscles samples with different concentrations of hydrocortisone are collected at three time points. An operational food chain simulation framework is constructed and 175 lipid molecules are identified by UHPLC-Q-Orbitrap HRMS. Compared to the control group, 66 lipid molecules are significantly different, including 17 triglycerides and 31 glycerophospholipids. Integrated analyses of lipidomics and proteomics indicate that hydrocortisone promotes adipose triglyceride lipase and hormone sensitive lipase activity to precondition for triglycerides hydrolysis. Quantitative lipidomics analysis shows the presence of hydrocortisone decreases the concentration of docosahexaenoic acid (3.66 ± 0.15-3.09 ± 0.12 mg kg-1 ) and eicosapentanoic acid (0.54 ± 0.09-0.48 ± 0.06 mg kg-1 ). A noteworthy increase of most saturated triglycerides concentration with the prolonging of time is observed. CONCLUSIONS Hydrocortisone originating from animal-derived food induces glycerophospholipids degradation and triglycerides hydrolysis through promoting adipose triglyceride lipase, hormone sensitive lipase, and phosphoglycerate kinase activity and further intervenes lipid nutrients utilization.
Collapse
Affiliation(s)
- Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.,Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an, 710021, China
| | - Xixuan Wu
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Lin Shi
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| |
Collapse
|
20
|
Zhen Y, Chen Y, Ge L, Wei W, Wang Y, Hu L, Loor JJ, Wang M, Yin J. The Short-Day Cycle Induces Intestinal Epithelial Purine Metabolism Imbalance and Hepatic Disfunctions in Antibiotic-Mediated Gut Microbiota Perturbation Mice. Int J Mol Sci 2022; 23:ijms23116008. [PMID: 35682688 PMCID: PMC9181120 DOI: 10.3390/ijms23116008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/14/2022] [Accepted: 05/24/2022] [Indexed: 11/28/2022] Open
Abstract
Intestinal microbiota dysbiosis is related to many metabolic diseases in human health. Meanwhile, as an irregular environmental light–dark (LD) cycle, short day (SD) may induce host circadian rhythm disturbances and worsen the risks of gut dysbiosis. Herein, we investigated how LD cycles regulate intestinal metabolism upon the destruction of gut microbes with antibiotic treatments. The growth indices, serum parameters, concentrations of short-chain fatty acids (SCFAs), and relative abundance of intestinal microbes were measured after euthanasia; intestinal contents, epithelial metabolomics, and hepatic transcriptome sequencing were also assessed. Compared with a normal LD cycle (NLD), SD increased the body weight, spleen weight, and serum concentration of aspartate aminotransferase, while it decreased high-density lipoprotein. Meanwhile, SD increased the relative abundance of the Bacteroidetes phylum while it decreased the Firmicutes phylum in the gut of ABX mice, thus leading to a disorder of SCFA metabolism. Metabolomics data revealed that SD exposure altered gut microbial metabolism in ABX mice, which also displayed more serious alterations in the gut epithelium. In addition, most differentially expressed metabolites were decreased, especially the purine metabolism pathway in epithelial tissue. This response was mainly due to the down-regulation of adenine, inosine, deoxyguanosine, adenylsuccinic acid, hypoxanthine, GDP, IMP, GMP, and AMP. Finally, the transcriptome data also indicated that SD has some negative effects on hepatic metabolism and endocrine, digestive, and disease processes. Overall, SD induced an epithelial and hepatic purine metabolism pathway imbalance in ABX mice, as well as the gut microbes and their metabolites, all of which could contribute to host metabolism and digestion, endocrine system disorders, and may even cause diseases in the host.
Collapse
Affiliation(s)
- Yongkang Zhen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Z.); (Y.C.); (L.G.); (W.W.); (Y.W.); (L.H.)
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi 832000, China
| | - Yifei Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Z.); (Y.C.); (L.G.); (W.W.); (Y.W.); (L.H.)
| | - Ling Ge
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Z.); (Y.C.); (L.G.); (W.W.); (Y.W.); (L.H.)
| | - Wenjun Wei
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Z.); (Y.C.); (L.G.); (W.W.); (Y.W.); (L.H.)
| | - Yusu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Z.); (Y.C.); (L.G.); (W.W.); (Y.W.); (L.H.)
| | - Liangyu Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Z.); (Y.C.); (L.G.); (W.W.); (Y.W.); (L.H.)
- Human and Animal Physiology, Wageningen University & Research, 6708 WD Wageningen, The Netherlands
| | - Juan J. Loor
- Mammalian Nutrition Physiology Genomics, Department of Animal Sciences, Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA;
| | - Mengzhi Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Y.Z.); (Y.C.); (L.G.); (W.W.); (Y.W.); (L.H.)
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi 832000, China
- Correspondence: (M.W.); (J.Y.)
| | - Junliang Yin
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Sciences, Shihezi 832000, China
- Correspondence: (M.W.); (J.Y.)
| |
Collapse
|