1
|
Wang T, Liu S, Shen W, Liu J, Liu Y, Li Y, Zhang F, Li T, Zhang X, Tian W, Zhang J, Ma J, Guo Y, Mi X, Lin Y, Hu Q, Zhang X, Liu J, Wang H. α-linolenic acid mitigates microglia-mediated neuroinflammation of schizophrenia in mice by suppressing the NF-κB/NLRP3 pathway via binding GPR120-β-arrestin 2. Int Immunopharmacol 2024; 142:113047. [PMID: 39236458 DOI: 10.1016/j.intimp.2024.113047] [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: 07/12/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Schizophrenia (SCZ) is a heterogeneous psychiatric disorder that is poorly treated by current therapies. Emerging evidence indicates that SCZ is closely correlated with a persistent neuroinflammation. α-linolenic acid (ALA) is highly concentrated in the brain and represents a modulator of the immune system by decreasing the inflammatory response in chronic metabolic diseases. This study was first designed to investigate the potential role of dietary ALA on cognitive function and neuroinflammation in mice with SCZ. METHODS In vivo, after 2 weeks of modeling, mice were treated with dietary ALA treatment for 6 weeks. In vitro, inflammation model was created using lipopolysaccharide as an inducer in BV2 microglial cells. RESULTS Our results demonstrated that ALA alleviated cognitive impairment and enhanced synaptic plasticity in mice with SCZ. Moreover, ALA mitigated systematic and cerebral inflammation through elevating IL-10 and inhibiting IL-1β, IL-6, IL-18 and TNF-α. Furthermore, ALA notably inhibited microglia and pro-inflammatory monocytes, as well as microglial activation andpolarization. Mechanistically, ALA up-regulated the expressions of G protein coupled receptor (GPR) 120 and associated β-inhibitor protein 2 (β-arrestin2), accompanied by observable weakened levels of transforming growth factor-β activated kinase 1 (TAK1), NF-κB p65, cysteine proteinase-1 (caspase-1), pro-caspase-1, associated speck-like protein (ASC) and NLRP3. In vitro, ALA directly restrained the inflammation of microglia by decreasing the levels of pro-inflammatory factors and regulating microglial polarization via GPR120-NF-κB/NLRP3inflammasome signaling pathway, whereas AH7614 definitely eliminated this anti-inflammatory effect of ALA. CONCLUSION Dietary ALA ameliorates microglia-mediated neuroinflammation by suppressing the NF-κB/NLRP3 pathway via binding GPR120-β-arrestin2.
Collapse
Affiliation(s)
- Ting Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| | - Shudan Liu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| | - Wenke Shen
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| | - Jian Liu
- Clinical Medical College, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Yuanyuan Liu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| | - Yiwei Li
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| | - Feng Zhang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| | - Ting Li
- Clinical Medical College, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Xiaoxu Zhang
- General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Wenyan Tian
- Clinical Medical College, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Jiani Zhang
- Clinical Medical College, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Junbai Ma
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| | - Yamei Guo
- General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Xiaojuan Mi
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| | - Yuan Lin
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| | - Qikuan Hu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| | - Xiaoxia Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Juan Liu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China; General Hospital of Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Hao Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004 Ningxia, China.
| |
Collapse
|
2
|
Šardzíková S, Gajewska M, Gałka N, Štefánek M, Baláž A, Garaiová M, Holič R, Świderek W, Šoltys K. Can longer lifespan be associated with gut microbiota involvement in lipid metabolism? FEMS Microbiol Ecol 2024; 100:fiae135. [PMID: 39354675 PMCID: PMC11503954 DOI: 10.1093/femsec/fiae135] [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: 12/01/2023] [Revised: 08/25/2024] [Accepted: 09/30/2024] [Indexed: 10/03/2024] Open
Abstract
Biological aging is linked to altered body composition and reduced neuroactive steroid hormones like dehydroepiandrosterone sulfate (DHEAS), which can stimulate the GABA signaling pathway via gut microbiota. Our study examined the association of gut microbiota with lifespan in mice through comprehensive analysis of its composition and functional involvement in cholesterol sulfate, a precursor of DHEAS, metabolism. We used 16S rRNA and metagenomic sequencing, followed by metabolic pathway prediction and thin layer chromatography and MALDI-TOF cholesterol sulfate identification. Significant increases in bacteria such as Bacteroides, typical for long-lived and Odoribacter and Colidextribacter, specific for short-lived mice were detected. Furthermore, for males (Rikenella and Alloprevotella) and females (Lactobacillus and Bacteroides), specific bacterial groups emerged as predictors (AUC = 1), highlighting sex-specific patterns. Long-lived mice showed a strong correlation of Bacteroides (0.918) with lipid and steroid hormone metabolism, while a negative correlation of GABAergic synapse with body weight (-0.589). We found that several Bacteroides species harboring the sulfotransferase gene and gene cluster for sulfonate donor synthesis are involved in converting cholesterol to cholesterol sulfate, significantly higher in the feces of long-lived individuals. Overall, we suggest that increased involvement of gut bacteria, mainly Bacteroides spp., in cholesterol sulfate synthesis could ameliorate aging through lipid metabolism.
Collapse
Affiliation(s)
- Sára Šardzíková
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| | - Marta Gajewska
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Norbert Gałka
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Matúš Štefánek
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| | - Andrej Baláž
- Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava, 842 48 Bratislava, Slovakia
| | - Martina Garaiová
- Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska Cesta 9, 84005 Bratislava, Slovakia
| | - Roman Holič
- Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska Cesta 9, 84005 Bratislava, Slovakia
| | - Wiesław Świderek
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Katarína Šoltys
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| |
Collapse
|
3
|
Zhang L, Yang Z, Zhang L, Wei Y, Wan L. Causal effect of gut microbiota on juvenile idiopathic arthritis: A two-sample Mendelian a randomization study. J Cell Mol Med 2024; 28:e70183. [PMID: 39473264 PMCID: PMC11522359 DOI: 10.1111/jcmm.70183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 09/03/2024] [Accepted: 10/19/2024] [Indexed: 11/02/2024] Open
Abstract
There is increasing evidence of a significant association between the gut microbiome and juvenile idiopathic arthritis (JIA). However, whether this association is causal remains to be determined. This study was a two-sample Mendelian randomization (MR) study using publicly available genome-wide association study (GWAS) summary data to investigate the causal relationship between the gut microbiome and JIA. We used summary data on gut flora and JIA obtained from genome-wide association studies (GWAS) from MiBioGen and NHGRI-EBI, using inverse variance weighting as the main method to analyse causality in the TSMR causality analysis. To check the stability of the TSMR results, we performed several sensitivity analyses and assessed the presence of reverse causality through a reverse TSMR analysis. We calculated the degree of sample overlap where applicable. The current TSMR analyses identified four bacterial taxa associated with JIA. Specifically, two bacteria, Catenibacterium (p = 2 × 10-2) and Holdemania (p = 4 × 10-2), were negatively associated with the risk of developing JIA, suggesting a protective effect, while Olsenella (p = 1 × 10-2) and Rikenellaceae (RC9gutgroup) (p = 1 × 10-2) were positively associated with the risk of JIA, suggesting that these two bacteria may be risk factors for JIA. However, the results for Catenibacterium and Holdemania should be interpreted with caution due to instability observed in 'leave-one-out' sensitivity analyses. Reverse TSMR analyses found no evidence of reverse causality between JIA and gut flora. Our confirmation of a causal relationship between gut flora and JIA provides an innovative perspective for the study of JIA: targeting and modulating dysregulation of specific bacterial taxa to prevent and treat JIA.
Collapse
Affiliation(s)
- Lian Zhang
- Shenzhen Children's HospitalShenzhenChina
| | - Zhihua Yang
- Department of Internal Medicine V Hematology Oncology RheumatologyHeidelberg University HospitalHeidelbergGermany
| | - LuLu Zhang
- Shenzhen Children's HospitalShenzhenChina
| | - Yanwen Wei
- Shenzhen Hospital of Integrated Traditional Chinese and Wsestern MedicineShenzhenChina
| | | |
Collapse
|
4
|
Zhang G, Zhang H, Dong R, Zhao H, Li J, Yue W, Ma Z. Oleanolic acid attenuates obesity through modulating the lipid metabolism in high-fat diet-fed mice. Food Sci Nutr 2024; 12:8243-8254. [PMID: 39479652 PMCID: PMC11521747 DOI: 10.1002/fsn3.4408] [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: 05/08/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 11/02/2024] Open
Abstract
As a natural pentacyclic triterpenoid, oleanolic acid has hepatoprotective, anti-inflammatory, and antioxidant activities. This work performed the in vitro experiments and animal assay to explore whether oleanolic acid alleviates lipid accumulation induced by high-fat diet by mediating PPARγ. Oil red O staining showed that oleanolic acid can reduce lipid accumulation in HepG2 cells, which were treated with oleic acid and palmitic acid. Immunofluorescence, western blot analysis, and RT-qPCR showed that oleanolic acid could promote nuclear translocation of PPARγ and reduce the expression level of PPARγ, C/EBP-β, and SREBP-1c. The results of in vivo experiments indicated that dietary intervention with oleanolic acid can effectively improve the fat accumulation in liver tissue and attenuate the level of IL-6 and TNF-α in serum caused by high-fat diet. Meanwhile, oleanolic acid did not cause lesions in vital organs at the experimental concentrations. In addition, the computer simulation indicated that oleanolic acid could directly bind to PPARγ with a reasonable and stable docking conformation. The above research results can provide new evidence for oleanolic acid to prevent nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Guangjie Zhang
- School of Biology and Food EngineeringAnyang Institute of TechnologyAnyangChina
| | - Huiying Zhang
- Department of Thoracic SurgeryQilu Hospital of Shandong UniversityJinanChina
| | - Ruiyi Dong
- College of Physical EducationHunan Normal UniversityChangshaChina
| | - Hongmei Zhao
- School of Biology and Food EngineeringAnyang Institute of TechnologyAnyangChina
| | - Junfeng Li
- College of Food Science and EngineeringJilin UniversityChangchunChina
| | - Weiming Yue
- Department of Thoracic SurgeryQilu Hospital of Shandong UniversityJinanChina
| | - Zheng Ma
- Department of Thoracic SurgeryQilu Hospital of Shandong UniversityJinanChina
| |
Collapse
|
5
|
Cao Y, Yang Y, Liang Z, Guo W, Lv X, Ni L, Chen Y. Synthesis of Ganoderic Acids Loaded Zein-Chitosan Nanoparticles and Evaluation of Their Hepatoprotective Effect on Mice Given Excessive Alcohol. Foods 2024; 13:2760. [PMID: 39272525 PMCID: PMC11394847 DOI: 10.3390/foods13172760] [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: 07/22/2024] [Revised: 08/24/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
Ganoderma lucidum, used in East Asia for its health benefits, contains ganoderic acids (GA) which have various pharmacological activities but are limited by poor water solubility and low oral bioaccessibility. This study synthesized and characterized ganoderic acids loaded zein-chitosan nanoparticles (GA-NPs), and investigated its advantages in alleviating alcoholic liver injury (ALI) in mice model. The GA-NPs demonstrated high encapsulation efficiency (92.68%), small particle size (177.20 nm), and a +29.53 mV zeta potential. The experimental results of alcohol-induced liver injury mouse model showed that GA-NPs significantly improved liver metabolic function, reduced alcohol-induced liver oxidative stress in liver by decreasing lactate dehydrogenase activity and malondialdehyde level, while increasing the activities of liver antioxidant enzymes and alcohol dehydrogenase. Moreover, GA-NPs were favorable to ameliorate intestinal microbiota dysbiosis in mice exposed to alcohol by increasing the proportion of probiotics such as Romboutsia, Faecalibaculum, Bifidobacterium and Turicibacter, etc., which were highly correlated with the improvement of liver function. Furthermore, GA-NPs modulated the mRNA expression related to ethanol metabolism, oxidative stress and lipid metabolism. Conclusively, this study revealed that GA-NPs have stronger hepatoprotective effects than non-encapsulated ganoderic acids on alleviating ALI by regulating intestinal microbiota and liver metabolism.
Collapse
Affiliation(s)
- Yingjia Cao
- Department of Hepatopancreatobiliary Surgery, Fujian Research Institute of Abdominal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang 362200, China
| | - Yuheng Yang
- Department of Hepatopancreatobiliary Surgery, Fujian Research Institute of Abdominal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- The First Affiliated Hospital of Fujian Medical University, Fujian Medical University, Fuzhou 350004, China
| | - Zihua Liang
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang 362200, China
| | - Weiling Guo
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang 362200, China
| | - Xucong Lv
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang 362200, China
| | - Li Ni
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China
- Food Nutrition and Health Research Center, School of Advanced Manufacturing, Fuzhou University, Jinjiang 362200, China
| | - Youting Chen
- Department of Hepatopancreatobiliary Surgery, Fujian Research Institute of Abdominal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
- Department of Hepatopancreatobiliary Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| |
Collapse
|
6
|
Fu Y, Wang Q, Tang Z, Liu G, Guan G, Lyu J. Cordycepin Ameliorates High Fat Diet-Induced Obesity by Modulating Endogenous Metabolism and Gut Microbiota Dysbiosis. Nutrients 2024; 16:2859. [PMID: 39275176 PMCID: PMC11396883 DOI: 10.3390/nu16172859] [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: 07/16/2024] [Revised: 08/14/2024] [Accepted: 08/24/2024] [Indexed: 09/16/2024] Open
Abstract
BACKGROUND Numerous metabolic illnesses have obesity as a risk factor. The composition of the gut microbiota and endogenous metabolism are important factors in the onset and progression of obesity. Recent research indicates that cordycepin (CRD), derived from fungi, exhibits anti-inflammatory and antioxidant properties, showing potential in combating obesity. However, further investigation is required to delineate its precise impacts on endogenous metabolism and gut microbiota. METHODS In this work, male C57BL/6J mice were used as models of obesity caused by a high-fat diet (HFD) and given CRD. Mice's colon, liver, and adipose tissues were stained with H&E. Serum metabolome analysis and 16S rRNA sequencing elucidated the effects of CRD on HFD-induced obese mice and identified potential mediators for its anti-obesity effects. RESULTS CRD intervention alleviated HFD-induced intestinal inflammation, improved blood glucose levels, and reduced fat accumulation. Furthermore, CRD supplementation demonstrated the ability to modulate endogenous metabolic disorders by regulating the levels of key metabolites, including DL-2-aminooctanoic acid, inositol, and 6-deoxyfagomine. CRD influenced the abundance of important microbiota such as Parasutterella, Alloprevotella, Prevotellaceae_NK3B31_group, Alistipes, unclassified_Clostridia_vadinBB60_group, and unclassified_Muribaculaceae, ultimately leading to the modulation of endogenous metabolism and the amelioration of gut microbiota disorders. CONCLUSIONS According to our research, CRD therapies show promise in regulating fat accumulation and stabilizing blood glucose levels. Furthermore, through the modulation of gut microbiota composition and key metabolites, CRD interventions have the dual capacity to prevent and ameliorate obesity.
Collapse
Affiliation(s)
- Yifeng Fu
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Qiangfeng Wang
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Zihan Tang
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Gang Liu
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Guiping Guan
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Jin Lyu
- Department of Pathology, The First People's Hospital of Foshan, Foshan 528000, China
| |
Collapse
|
7
|
Wang Y, Zhang Y, Wang Q, Fan Y, Li W, Liu M, Zhang X, Zhou W, Wang M, Jiang S, Shang E, Duan J. Multi-omics combined to explore the purging mechanism of Rhei Radix et Rhizoma and Magnoliae Officinalis Cortex. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1243:124218. [PMID: 38959707 DOI: 10.1016/j.jchromb.2024.124218] [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: 04/24/2024] [Revised: 06/04/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Rhei Radix et Rhizoma and Magnoliae Officinalis Cortex have been used together to treat constipation in the clinical practices for more than 2000 years. Nonetheless, their compatibility mechanism is still unclear. In this study, the amelioration of Rhei Radix et Rhizoma combined with Magnoliae Officinalis Cortex on constipation was systematically and comprehensively evaluated. The results showed that their compatibility could markedly shorten gastrointestinal transport time, increase fecal water content and frequency of defecation, improve gastrointestinal hormone disorders and protect colon tissue of constipation rats compared with the single drug. Furthermore, according to 16S rRNA sequencing in conjunction with UPLC-Q-TOF/MS, the combination of two herbal medications could greatly raise the number of salutary bacteria (Lachnospiraceae, Romboutsia and Subdoligranulum) while decreasing the abundance of pathogenic bacteria (Erysipelatoclostridiaceae). And two herb drugs could markedly improve the disorder of fecal metabolic profiles. A total of 7 different metabolites associated with constipation were remarkably shifted by the compatibility of two herbs, which were mainly related to arachidonic acid metabolism, alpha-linolenic acid metabolism, unsaturated fatty acid biosynthesis and other metabolic ways. Thus, the regulation of intestinal microbiome and its metabolism could be a potential target for Rhei Radix et Rhizoma and Magnoliae Officinalis Cortex herb pair to treat constipation. Furthermore, the multi-omics approach utilized in this study, which integrated the microbiome and metabolome, had potential for investigating the mechanism of traditional Chinese medicines.
Collapse
Affiliation(s)
- Yu Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Yun Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Quyi Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Yuwen Fan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Wenwen Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Meijuan Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Xiaoxiao Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Wenwen Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Mingyang Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China.
| | - Erxin Shang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China.
| |
Collapse
|
8
|
Li C, Liu Z, Wei W, Chen C, Zhang L, Wang Y, Zhou B, Liu L, Li X, Zhao C. Exploring the Regulatory Effect of LPJZ-658 on Copper Deficiency Combined with Sugar-Induced MASLD in Middle-Aged Mice Based on Multi-Omics Analysis. Nutrients 2024; 16:2010. [PMID: 38999758 PMCID: PMC11243161 DOI: 10.3390/nu16132010] [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: 05/05/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 07/14/2024] Open
Abstract
Globally, metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed nonalcoholic fatty liver disease (NAFLD), is one of the most common liver disorders and is strongly associated with copper deficiency. To explore the potential effects and mechanisms of Lactiplantibacillus plantarum LPJZ-658, copper deficiency combined with a high-sugar diet-induced MASLD mouse model was utilized in this study. We fed 40-week-old (middle-aged) male C57BL/6 mice a copper-deficient and high-sugar diet for 16 weeks (CuDS), with supplementary LPJZ-658 for the last 6 weeks (CuDS + LPJZ-658). In this study, we measured body weight, liver weight, and serum biochemical markers. Lipid accumulation, histology, lipidomics, and sphingolipid metabolism-related enzyme expression were investigated to analyze liver function. Untargeted metabolomics was used to analyze the serum and the composition and abundance of intestinal flora. In addition, the correlation between differential liver lipid profiles, serum metabolites, and gut flora at the genus level was measured. The results show that LPJZ-658 significantly improves abnormal liver function and hepatic steatosis. The lipidomics analyses and metabolic pathway analysis identified sphingolipid, retinol, and glycerophospholipid metabolism as the most relevant metabolic pathways that characterized liver lipid dysregulation in the CuDS group. Consistently, RT-qPCR analyses revealed that the enzymes catalyzing sphingolipid metabolism that were significantly upregulated in the CuDS group were downregulated by the LPJZ-658 treatment. In addition, the serum metabolomics results indicated that the linoleic acid, taurine and hypotaurine, and ascorbate and aldarate metabolism pathways were associated with CuDS-induced MASLD. Notably, we found that treatment with LPJZ-658 partially reversed the changes in the differential serum metabolites. Finally, LPJZ-658 effectively regulated intestinal flora abnormalities and was significantly correlated with differential hepatic lipid species and serum metabolites. In conclusion, we elucidated the function and potential mechanisms of LPJZ-658 in alleviating copper deficiency combined with sugar-induced middle-aged MASLD and hope this will provide possible treatment strategies for improving MASLD.
Collapse
Affiliation(s)
- Chunhua Li
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| | - Ziqi Liu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| | - Wei Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China;
| | - Chen Chen
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| | - Lichun Zhang
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| | - Yang Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China;
| | - Bo Zhou
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Changchun Veterinary Research Institute, Chinese Academy of Medical Sciences, Changchun 130122, China;
| | - Liming Liu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| | - Xiao Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Changchun Veterinary Research Institute, Chinese Academy of Medical Sciences, Changchun 130122, China;
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Cuiqing Zhao
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin City 132101, China; (C.L.); (Z.L.); (C.C.); (L.Z.); (L.L.)
| |
Collapse
|
9
|
Guo X, Wang R, Chen R, Zhang Z, Wang J, Liu X. Gut microbiota and serum metabolite signatures along the colorectal adenoma-carcinoma sequence: Implications for early detection and intervention. Clin Chim Acta 2024; 560:119732. [PMID: 38772522 DOI: 10.1016/j.cca.2024.119732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/11/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024]
Abstract
AIM Our study focuses on the microbial and metabolomic profile changes during the adenoma stage, as adenomas can be considered potential precursors to colorectal cancer through the adenoma-carcinoma sequence. Identifying possible intervention targets at this stage may aid in preventing the progression of colorectal adenoma (CRA) to malignant lesions. Furthermore, we evaluate the efficacy of combined microbial and metabolite biomarkers in detecting CRA. METHODS Fecal metagenomic and serum metabolomic analyses were performed for the discovery of alterations of gut microbiome and metabolites in CRA patients (n = 26), Colorectal cancer (CRC) patients (n = 19), Familial Adenomatous Polyposis (FAP) patients (n = 10), and healthy controls (n = 20). Finally, analyzing the associations between gut microbes and metabolites was performed by a Receiver Operating Characteristic (ROC) curve. RESULTS Our analysis present that CRA patients differ significantly in gut microflora and serum metabolites compared with healthy controls, especially for Lachnospiraceae and Parasutterella. Its main metabolite, butyric acid, concentrations were raised in CRA patients compared with the healthy controls, indicating its role as a promoter of colorectal tumorigenesis. α-Linolenic acid and lysophosphatidylcholine represented the other healthy metabolite for CRA. Combining five microbial and five metabolite biomarkers, we differentiated CRA from CRC with an Area Under the Curve (AUC) of 0.85 out of this performance vastly superior to the specificity recorded by traditional markers CEA and CA199 in such differentiation of these conditions. CONCLUSIONS The study underlines significant microbial and metabolic alterations in CRA with a novel insight into screening and early intervention of its tumorigenesis.
Collapse
Affiliation(s)
- Xiaodong Guo
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, ShangHai 200437, China.
| | - Ruoyao Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, ShangHai 200437, China
| | - Rui Chen
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, ShangHai 200437, China
| | - Zhongxiao Zhang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, No.1111, XianXia Road, Shanghai 200336, China.
| | - Jingxia Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, ShangHai 200437, China
| | - Xuan Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
10
|
Garrison EC, Brown AMV, Salazar MM, Barr B, Moustaid-Moussa N, Gollahon LS. Microbiome Taxonomic and Functional Differences in C3H/HeJ Mice Fed a Long-Term High-Fat Diet with Beef Protein ± Ammonium Hydroxide Supplementation. Nutrients 2024; 16:1613. [PMID: 38892546 PMCID: PMC11174526 DOI: 10.3390/nu16111613] [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: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Studies have suggested that alkalinized foods may reduce the effects of the acidogenic Western diet in promoting obesity, metabolic syndrome, type 2 diabetes, cancer, and coronary heart disease. Indeed, a recent study in mice fed a high-fat diet containing dietary beef supplemented with ammonium hydroxide showed improvement in a suite of metabolic outcomes. However, the effects of dietary protein ammonium supplementation on the microbiome remain unknown. In this study, the effects of ammonium supplementation on beef protein towards microbiome taxa and function in a high-fat diet were analyzed. Fecal microbiomes were characterized using a shotgun metagenomic approach for 16-month-old male and female mice after long-term diet treatments. The results for ammoniated diets showed that several bacteria known to be associated with health benefits increased significantly, including Romboutsia, Oscillospiraceae, and Lactococcus cremoris. The beneficial mucin-degrader Akkermansia was especially abundant, with a high prevalence (~86%) in females. Concurrently, the phyla Actinomycetota (Actinobacteria) and Bacteroidota (Bacteroidetes) were significantly reduced. While sex was a confounding factor affecting microbiome responses to ammonium supplementation in dietary protein, it is worth noting that several putatively beneficial microbiome functions increased with ammonium supplementation, such as glycine betaine transport, xenobiotic detoxification, enhanced defense, and others. Conversely, many disease-associated microbiome functions reduced. Importantly, modifying protein pH alone via ammonium supplementation induced beneficial microbiota changes. Taken together, these results suggest that ammonium-supplemented proteins may mediate some negative microbiome-associated effects of high-fat/Western diets.
Collapse
Affiliation(s)
- Emily C. Garrison
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA; (E.C.G.); (A.M.V.B.); (M.M.S.); (B.B.)
| | - Amanda M. V. Brown
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA; (E.C.G.); (A.M.V.B.); (M.M.S.); (B.B.)
| | - McKinlee M. Salazar
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA; (E.C.G.); (A.M.V.B.); (M.M.S.); (B.B.)
| | - Benjamin Barr
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA; (E.C.G.); (A.M.V.B.); (M.M.S.); (B.B.)
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA;
- Obesity Research Institute, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA
| | - Lauren S. Gollahon
- Department of Biological Sciences, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA; (E.C.G.); (A.M.V.B.); (M.M.S.); (B.B.)
- Obesity Research Institute, Texas Tech University, 2500 Broadway, Lubbock, TX 79409, USA
| |
Collapse
|
11
|
Zhang Y, Tang N, Zhou H, Zhu Y. The role of microbial metabolites in endocrine tumorigenesis: From the mechanistic insights to potential therapeutic biomarkers. Biomed Pharmacother 2024; 172:116218. [PMID: 38308969 DOI: 10.1016/j.biopha.2024.116218] [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: 09/26/2023] [Revised: 12/28/2023] [Accepted: 01/22/2024] [Indexed: 02/05/2024] Open
Abstract
Microbial metabolites have been indicated to communicate with the host's endocrine system, regulating hormone production, immune-endocrine communications, and interactions along the gut-brain axis, eventually affecting the occurrence of endocrine cancer. Furthermore, microbiota metabolites such as short-chain fatty acids (SCFAs) have been found to affect the tumor microenvironment and boost immunity against tumors. SCFAs, including butyrate and acetate, have been demonstrated to exert anti-proliferative and anti-protective activity on pancreatic cancer cells. The employing of microbial metabolic products in conjunction with radiation and chemotherapy has shown promising outcomes in terms of reducing treatment side effects and boosting effectiveness. Certain metabolites, such as valerate and butyrate, have been made known to improve the efficiency of CAR T-cell treatment, whilst others, such as indole-derived tryptophan metabolites, have been shown to inhibit tumor immunity. This review explores the intricate interplay between microbial metabolites and endocrine tumorigenesis, spanning mechanistic insights to the discovery of potential therapeutic biomarkers.
Collapse
Affiliation(s)
- Yiyi Zhang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Nie Tang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Hui Zhou
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| | - Ying Zhu
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| |
Collapse
|
12
|
Wang J, Hu Z, Xu Q, Shi Y, Cao X, Ma Y, Wang M, Zhang C, Luo X, Lin F, Li X, Duan Y, Cai H. Gut microbiome-based noninvasive diagnostic model to predict acute coronary syndromes. Front Cell Infect Microbiol 2024; 13:1305375. [PMID: 38298920 PMCID: PMC10829574 DOI: 10.3389/fcimb.2023.1305375] [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: 10/19/2023] [Accepted: 12/26/2023] [Indexed: 02/02/2024] Open
Abstract
Background Previous studies have shown that alterations in the gut microbiota are closely associated with Acute Coronary Syndrome (ACS) development. However, the value of gut microbiota for early diagnosis of ACS remains understudied. Methods We recruited 66 volunteers, including 29 patients with a first diagnosis of ACS and 37 healthy volunteers during the same period, collected their fecal samples, and sequenced the V4 region of the 16S rRNA gene. Functional prediction of the microbiota was performed using PICRUSt2. Subsequently, we constructed a nomogram and corresponding webpage based on microbial markers to assist in the diagnosis of ACS. The diagnostic performance and usefulness of the model were analyzed using boostrap internal validation, calibration curves, and decision curve analysis (DCA). Results Compared to that of healthy controls, the diversity and composition of microbial community of patients with ACS was markedly abnormal. Potentially pathogenic genera such as Streptococcus and Acinetobacter were significantly increased in the ACS group, whereas certain SCFA-producing genera such as Blautia and Agathobacter were depleted. In addition, in the correlation analysis with clinical indicators, the microbiota was observed to be associated with the level of inflammation and severity of coronary atherosclerosis. Finally, a diagnostic model for ACS based on gut microbiota and clinical variables was developed with an area under the receiver operating characteristic (ROC) curve (AUC) of 0.963 (95% CI: 0.925-1) and an AUC value of 0.948 (95% CI: 0.549-0.641) for bootstrap internal validation. The calibration curves of the model show good consistency between the actual and predicted probabilities. The DCA showed that the model had a high net clinical benefit for clinical applications. Conclusion Our study is the first to characterize the composition and function of the gut microbiota in patients with ACS and healthy populations in Southwest China and demonstrates the potential effect of the microbiota as a non-invasive marker for the early diagnosis of ACS.
Collapse
Affiliation(s)
- Jincheng Wang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhao Hu
- Department of Geriatric Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qiuyue Xu
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Yunnan Key Laboratory of Laboratory Medicine, Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming, China
| | - Yunke Shi
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xingyu Cao
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yiming Ma
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mingqiang Wang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chaoyue Zhang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiang Luo
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fanru Lin
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xianbin Li
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yong Duan
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Yunnan Key Laboratory of Laboratory Medicine, Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming, China
| | - Hongyan Cai
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
13
|
Bourragat A, Escoula Q, Bellenger S, Zemb O, Beaumont M, Chaumonnot K, Farine JP, Jacotot E, Bonnotte A, Avoscan L, Lherminier J, Luo K, Narce M, Bellenger J. The transplantation of the gut microbiome of fat-1 mice protects against colonic mucus layer disruption and endoplasmic reticulum stress induced by high fat diet. Gut Microbes 2024; 16:2356270. [PMID: 38797998 PMCID: PMC11135845 DOI: 10.1080/19490976.2024.2356270] [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: 06/06/2023] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
High-fat diets alter gut barrier integrity, leading to endotoxemia by impacting epithelial functions and inducing endoplasmic reticulum (ER) stress in intestinal secretory goblet cells. Indeed, ER stress, which is an important contributor to many chronic diseases such as obesity and obesity-related disorders, leads to altered synthesis and secretion of mucins that form the protective mucus barrier. In the present study, we investigated the relative contribution of omega-3 polyunsaturated fatty acid (PUFAs)-modified microbiota to alleviating alterations in intestinal mucus layer thickness and preserving gut barrier integrity. Male fat-1 transgenic mice (exhibiting endogenous omega-3 PUFAs tissue enrichment) and wild-type (WT) littermates were fed either an obesogenic high-fat diet (HFD) or a control diet. Unlike WT mice, HFD-fed fat-1 mice were protected against mucus layer alterations as well as an ER stress-mediated decrease in mucin expression. Moreover, cecal microbiota transferred from fat-1 to WT mice prevented changes in the colonic mucus layer mainly through colonic ER stress downregulation. These findings highlight a novel feature of the preventive effects of omega-3 fatty acids against intestinal permeability in obesity-related conditions.
Collapse
Affiliation(s)
- Amina Bourragat
- CTM UMR1231, Université de Bourgogne, Dijon, France
- CTM UMR1231, INSERM, Dijon, France
- LipSTIC LabEx, FCS Bourgogne-Franche Comté, Dijon, France
| | - Quentin Escoula
- CTM UMR1231, Université de Bourgogne, Dijon, France
- CTM UMR1231, INSERM, Dijon, France
- LipSTIC LabEx, FCS Bourgogne-Franche Comté, Dijon, France
- Valorex, La Messayais, Combourtillé, France
| | - Sandrine Bellenger
- CTM UMR1231, Université de Bourgogne, Dijon, France
- CTM UMR1231, INSERM, Dijon, France
- LipSTIC LabEx, FCS Bourgogne-Franche Comté, Dijon, France
| | - Olivier Zemb
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Martin Beaumont
- GenPhySE, Université de Toulouse, INRAE, ENVT, Castanet-Tolosan, France
| | - Killian Chaumonnot
- CTM UMR1231, Université de Bourgogne, Dijon, France
- CTM UMR1231, INSERM, Dijon, France
| | - Jean-Pierre Farine
- Centre des Sciences du Goût et de l’Alimentation, UMR6265 CNRS, UMR1324 INRA, Université de Bourgogne, Dijon, France
| | - Emmanuel Jacotot
- L’Institut Agro Dijon, PAM UMR A 02.102, Université de Bourgogne, Dijon, France
| | - Aline Bonnotte
- Agroécologie, L’Institut Agro Dijon, CNRS, INRAE, Plateforme DimaCell, Dijon, France
| | - Laure Avoscan
- Agroécologie, L’Institut Agro Dijon, CNRS, INRAE, Plateforme DimaCell, Dijon, France
| | - Jeanine Lherminier
- Agroécologie, L’Institut Agro Dijon, CNRS, INRAE, Plateforme DimaCell, Dijon, France
| | - Kangjia Luo
- CTM UMR1231, Université de Bourgogne, Dijon, France
- CTM UMR1231, INSERM, Dijon, France
- LipSTIC LabEx, FCS Bourgogne-Franche Comté, Dijon, France
| | - Michel Narce
- CTM UMR1231, Université de Bourgogne, Dijon, France
- CTM UMR1231, INSERM, Dijon, France
- LipSTIC LabEx, FCS Bourgogne-Franche Comté, Dijon, France
| | - Jérôme Bellenger
- CTM UMR1231, Université de Bourgogne, Dijon, France
- CTM UMR1231, INSERM, Dijon, France
- LipSTIC LabEx, FCS Bourgogne-Franche Comté, Dijon, France
| |
Collapse
|
14
|
Feng R, Yang W, Feng W, Huang X, Cen M, Peng G, Wu W, Wang Z, Jing Y, Long T, Liu Y, Li Z, Chang G, Huang K. Time-restricted feeding ameliorates non-alcoholic fatty liver disease through modulating hepatic nicotinamide metabolism via gut microbiota remodeling. Gut Microbes 2024; 16:2390164. [PMID: 39154362 PMCID: PMC11332628 DOI: 10.1080/19490976.2024.2390164] [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: 01/22/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/20/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as a global health concern, lacking specific therapeutic strategies. Time-restricted feeding (TRF) regimen demonstrated beneficial effects in NAFLD; however, the underlying mechanisms remain unclear. In this study, we established a NAFLD mouse model through a high-fat diet (HFD) and implemented the 16:8 TRF regimen for a duration of 6 weeks. We demonstrated that TRF remarkably alleviated hepatic steatosis in HFD mice. Of note, aldehyde oxidase 1 (AOX1), a key enzyme in hepatic nicotinamide (NAM) catabolism, exhibited apparent upregulation in response to HFD, leading to abnormal accumulation of N-Methyl-6-pyridone-3-carboxamide (N-Me-6-PY, also known as 2PY) and N-Methyl-4-pyridone-5-carboxamide (N-Me-4-PY, also known as 4PY), whereas it was almost restored by TRF. Both N-Me-6-PY and N-Me-4-PY promoted de novo lipogenesis and fatty acid uptake capacities in hepatocyte, and aggravated hepatic steatosis in mice either fed chow diet or HFD. In contrast, pharmacological inhibition of AOX1 was sufficient to ameliorate the hepatic steatosis and lipid metabolic dysregulation induced by HFD. Moreover, transplantation of fecal microbiota efficiently mimicked the modulatory effect of TRF on NAM metabolism, thus mitigating hepatic steatosis and lipid metabolic disturbance, suggesting a gut microbiota-dependent manner. In conclusion, our study reveals the intricate relationship between host NAM metabolic modification and gut microbiota remodeling during the amelioration of NAFLD by TRF, providing promising insights into the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Ruijia Feng
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenchao Yang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weiqi Feng
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiuyi Huang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meifeng Cen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guiyan Peng
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenrui Wu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhecun Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yexiang Jing
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ting Long
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunchong Liu
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zilun Li
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guangqi Chang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kan Huang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
15
|
Gao X, Yang W, Li S, Liu S, Yang W, Song S, Sheng J, Zhao Y, Tian Y. Moringa oleifera leaf alleviates functional constipation via regulating the gut microbiota and the enteric nervous system in mice. Front Microbiol 2023; 14:1315402. [PMID: 38192287 PMCID: PMC10773919 DOI: 10.3389/fmicb.2023.1315402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
Moringa oleifera Lam. leaf is not only a new food resource in China, but also a traditional medicinal plant. It is commonly used in the folk to alleviate constipation, but its laxative mechanism is not fully understood. Hence we investigated it in loperamide-induced functional constipation (FC) mice. The results showed that MOAE significantly regulated not only gastrointestinal hormones and neurotransmitters in serum but also important gastrointestinal motility factors in the enteric nervous system (ENS)-interstitial cells of Cajal (ICCs)-smooth muscle cell (SMC) network. Meanwhile, MOAE attenuated intestinal inflammation, increased cecal short-chain fatty acid levels and colonic antimicrobial peptide expression, and improved the impaired intestinal barrier function in loperamide-induced FC mice. In addition, MOAE also increased fecal water content by inhibiting the mRNA expression of colonic aquaporins (Aqp3 and Aqp4) in FC mice. Interestingly and importantly, MOAE affected the intestinal microbiota by inhibiting some key "constipation-causing" microbiota, such as Bacteroidaceae, Clostridiaceae, Bacteroides, and Ruminococcus, and promoting the growth of other important "constipation-curing" microbiota, such as Butyricoccus, Tyzzerella, and Desulfovibrio. These important taxa are significantly associated with a variety of indicators of constipation. These findings suggest that MOAE can promote defecation through its rich chemical composition to modulate the ENS-ICCs-SMCs network and the gut microecosystem.
Collapse
Affiliation(s)
- Xiaoyu Gao
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming, China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Weiqian Yang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Department of Hotel Management, Chongqing Vocational Institute of Tourism, Chongqing, China
| | - Sijin Li
- College of Pu’er Tea West Yunnan University of Applied Sciences, Puer, China
| | - Shuangfeng Liu
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Weixing Yang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Shuang Song
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jun Sheng
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Yan Zhao
- Division of Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yang Tian
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
16
|
Yang J, Liu S, Zhao Q, Li X, Jiang K. Gut microbiota-related metabolite alpha-linolenic acid mitigates intestinal inflammation induced by oral infection with Toxoplasma gondii. MICROBIOME 2023; 11:273. [PMID: 38087373 PMCID: PMC10714487 DOI: 10.1186/s40168-023-01681-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 09/27/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND Oral infection with cysts is the main transmission route of Toxoplasma gondii (T. gondii), which leads to lethal intestinal inflammation. It has been widely recognized that T. gondii infection alters the composition and metabolism of the gut microbiota, thereby affecting the progression of toxoplasmosis. However, the potential mechanisms remain unclear. In our previous study, there was a decrease in the severity of toxoplasmosis after T. gondii α-amylase (α-AMY) was knocked out. Here, we established mouse models of ME49 and Δα-amy cyst infection and then took advantage of 16S rRNA gene sequencing and metabolomics analysis to identify specific gut microbiota-related metabolites that mitigate T. gondii-induced intestinal inflammation and analyzed the underlying mechanism. RESULTS There were significant differences in the intestinal inflammation between ME49 cyst- and Δα-amy cyst-infected mice, and transferring feces from mice infected with Δα-amy cysts into antibiotic-treated mice mitigated colitis caused by T. gondii infection. 16S rRNA gene sequencing showed that the relative abundances of gut bacteria, such as Lactobacillus and Bacteroides, Bifidobacterium, [Prevotella], Paraprevotella and Macellibacteroides, were enriched in mice challenged with Δα-amy cysts. Spearman correlation analysis between gut microbiota and metabolites indicated that some fatty acids, including azelaic acid, suberic acid, alpha-linolenic acid (ALA), and citramalic acid, were highly positively correlated with the identified bacterial genera. Both oral administration of ALA and fecal microbiota transplantation (FMT) decreased the expression of pro-inflammatory cytokines and restrained the MyD88/NF-κB pathway, which mitigated colitis and ultimately improved host survival. Furthermore, transferring feces from mice treated with ALA reshaped the colonization of beneficial bacteria, such as Enterobacteriaceae, Proteobacteria, Shigella, Lactobacillus, and Enterococcus. CONCLUSIONS The present findings demonstrate that the host gut microbiota is closely associated with the severity of T. gondii infection. We provide the first evidence that ALA can alleviate T. gondii-induced colitis by improving the dysregulation of the host gut microbiota and suppressing the production of pro-inflammatory cytokines via the MyD88/NF-κB pathway. Our study provides new insight into the medical application of ALA for the treatment of lethal intestinal inflammation caused by Toxoplasma infection. Video Abstract.
Collapse
Affiliation(s)
- Jing Yang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China
| | - Songhao Liu
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China
| | - Qian Zhao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China
| | - Xiaobing Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China.
| | - Kangfeng Jiang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, Yunnan, China.
| |
Collapse
|
17
|
Han J, Li M, Li X, Liu C, Li XL, Wang K, Qiao R, Yang F, Han X, Li XJ. Effects of microbes in pig farms on occupational exposed persons and the environment. AMB Express 2023; 13:136. [PMID: 38032532 PMCID: PMC10689614 DOI: 10.1186/s13568-023-01631-x] [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: 06/09/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
In terms of pig farming, pig gut microbes have a significant effect on farmers and the farm environment. However, it is still unclear which microbial composition is more likely to contribute to this effect. This study collected a total of 136 samples, including pigs' faeces samples, farmers' faeces samples, samples from individuals who had no contact with any type of farm animal (referred to as 'non-exposed' persons), and environmental dust samples (collected from inside and outside pig houses and the farm) from two pig farms, pig farm A and pig farm B. Whereafter, 16S rRNA sequencing and taxonomic composition analysis were performed. According to the study, compared to non-exposed persons, pig farmers had a significantly higher abundance of 7 genera. In addition, the farmers were grouped according to the duration of their occupational exposure, and it was shown that 4 genera, including Turicibacter, Terrisporobacter, and Clostridium_sensu_stricto_1, exhibited a rise in more frequent contact with pigs. As compared to outside the pig house, the environmental dust has a greater concentration of the 3 bacteria mentioned before. Therefore, these 3 microbes can be considered as co-occurring microbes that may exist both in humans and the environment. Also, the 3 co-occurring microbes are involved in the fermentation and production of short-chain fatty acids and their effectiveness decreased as distance from the farm increased. This study shows that the 3 microbes where pig farmers co-occur with the environment come from pig farms, which provides fresh ideas for preventing the spread of microbial aerosols in pig farms and reducing pollution.
Collapse
Affiliation(s)
- Jinyi Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Mengyu Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xin Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Chuang Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xiu-Ling Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Kejun Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Ruimin Qiao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Feng Yang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xuelei Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China.
| | - Xin-Jian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China.
- Sanya Institute, Hainan Academy of Agricultural Science, Sanya, China.
| |
Collapse
|
18
|
Ren X, Xu J, Xu Y, Wang Q, Huang K, He X. Artemether Attenuates Gut Barrier Dysfunction and Intestinal Flora Imbalance in High-Fat and High-Fructose Diet-Fed Mice. Nutrients 2023; 15:4860. [PMID: 38068719 PMCID: PMC10707945 DOI: 10.3390/nu15234860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Intestinal inflammation is a key determinant of intestinal and systemic health, and when our intestines are damaged, there is disruption of the intestinal barrier, which in turn induces a systemic inflammatory response. However, the etiology and pathogenesis of inflammatory diseases of the intestine are still not fully understood. Artemether (ART), one of the artemisinin derivatives, has been widely used to treat malaria. Nevertheless, the effect of ART on intestinal inflammation remains unclear. The present study intended to elucidate the potential mechanism of ART in diet-induced intestinal injury. A high-fat and high-fructose (HFHF) diet-induced mouse model of intestinal injury was constructed, and the mice were treated with ART to examine their role in intestinal injury. RT-qPCR, Western blotting, immunohistochemical staining, and 16S rRNA gene sequencing were used to investigate the anti-intestinal inflammation effect and mechanism of ART. The results indicated that ART intervention may significantly ameliorate the intestinal flora imbalance caused by the HFHF diet and alleviate intestinal barrier function disorders and inflammatory responses by raising the expression of tight junction proteins ZO-1 and occludin and decreasing the expression of pro-inflammatory factors TNF-α and IL-1β. Moreover, ART intervention restrained HFHF-induced activation of the TLR4/NF-κB p65 pathway in colon tissue, which may be concerned with the potential protective effect of ART on intestinal inflammation. ART might provide new insights into further explaining the mechanism of action of other metabolic diseases caused by intestinal disorders.
Collapse
Affiliation(s)
- Xinxin Ren
- 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
| | - 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
| | - Qin Wang
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, 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
| |
Collapse
|
19
|
Li W, Cheng F, Zhang J, Li C, Yu D, Simayijiang H, Liu H, Li S, Yan J. Changes in Gut Microbiota and Metabolites in Papillary Thyroid Carcinoma Patients Following Radioactive Iodine Therapy. Int J Gen Med 2023; 16:4453-4464. [PMID: 37808207 PMCID: PMC10557971 DOI: 10.2147/ijgm.s433433] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/28/2023] [Indexed: 10/10/2023] Open
Abstract
Purpose Radioactive iodine therapy is administered through oral route, which is accumulated and absorbed in the intestine. However, its effects on the intestine remain unclear. In this study, we investigated the changes in the gut microbiota and metabolites following radioactive iodine therapy. Patients and Methods A total of 76 stool samples from the same 38 patients were collected at the start of radioactive iodine therapy and three days following the therapy. Stool microbiota and metabolites were detected using 16S rRNA gene sequencing and liquid chromatography-mass spectrometry. Results Enterobacteriales, Enterobacteriaceae and Escherichia-Shigella were elevated in most patients (27/38) following the therapy. The levels of 2-hydroxyundec-7-enoylcarnitine were significantly lower, whereas those of 5-dehydroavenasterol, butylisopropylamine, and salsoline-1-carboxylate were higher following the therapy. The relative abundance of Escherichia-Shigella was negatively correlated with 2-hydroxyundec-7-enoylcarnitine level (r2 = -0.661, P = 0.009). Functional pathways were predicted to be involved in amino acid and lipid metabolism following the therapy. Particularly, phenylalanine, linoleic acid, sphingolipid, purine, and alpha-linolenic acid metabolism were the main metabolic pathways. Conclusion Gut microbiota was disturbed following radioactive iodine therapy, with increased Escherichia-Shigella. Processes associated with energy production seems to be impacted following the therapy, with significantly decreased 2-hydroxyundec-7-enoylcarnitine level. Meanwhile, some metabolites and functional pathways may have a positive effect on intestinal homeostasis, and may be related to the repair and promotion of gut recovery following the therapy. This study provides a basic foundation to explore how radioactive iodine affects gut microbiota and metabolites, and how gut function is regulated in response to radioactive iodine therapy.
Collapse
Affiliation(s)
- Wanting Li
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Feng Cheng
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Jun Zhang
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Caihong Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Daijing Yu
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Halimureti Simayijiang
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Haiyan Liu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jiangwei Yan
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| |
Collapse
|
20
|
Xia H, Wang Y, Shi X, Liao W, Wang S, Sui J, Sun G. Beneficial Effects of Dietary Flaxseed Oil through Inflammation Pathways and Gut Microbiota in Streptozotocin-Induced Diabetic Mice. Foods 2023; 12:3229. [PMID: 37685162 PMCID: PMC10487211 DOI: 10.3390/foods12173229] [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: 07/31/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Flaxseed oil (FO) has displayed potential anti-diabetes properties by providing a high content of α-linolenic acid. However, the effects and mechanisms of FO on type 1 diabetes are still unclear. The present study aims to explore the effects of different doses of FO feeding on hepatic inflammation and gut microbiota in streptozotocin-induced diabetic mice. Forty-eight six-week-old C57BL/6J male mice were divided into a control group (CON), a diabetic group (MOD), a diabetes with 7.0% w/w FO feeding group (FO-L), and a diabetes with 10.5% w/w FO feeding group (FO-H) for six weeks. The 7.0% w/w and 10.5% w/w FO feeding groups exhibited potential recovery of the number and size of pancreas tissues. The fasting blood glucose level was significantly decreased only after 4 weeks of feeding with 10.5% w/w FO in diabetic mice. The 10.5% w/w FO feeding group significantly decreased the postprandial blood glucose level of mice in the OGTT test. Hepatic glycogen levels were dramatically upregulated in the mice fed with both 7.0% w/w and 10.5% w/w FO. FO feeding significantly attenuated hepatic LPS, TNF-α, and IL-1β levels. In addition, we observed that 7.0% w/w and 10.5% w/w FO feedings notably downregulated hepatic gene and protein expressions of TLR4, MyD88, and P65. Furthermore, only 10.5% FO regulated fecal microbiota by increasing the relative abundance of the Bacteroidetes phylum, Lactococcus family, and Muribaculaceae and Streptococcaceae family and genus in streptozotocin-induced diabetic mice. Therefore, we conclude that FO feeding plays a role in anti-inflammation via the regulation of hepatic LPS/TLR4/MyD88 pathways and gut microbiota. In addition, different doses of FO supplementation may exhibit varying mechanisms in streptozotocin-induced mice.
Collapse
Affiliation(s)
- Hui Xia
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China; (H.X.); (Y.W.); (X.S.); (W.L.); (S.W.); (J.S.)
| | - Ying Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China; (H.X.); (Y.W.); (X.S.); (W.L.); (S.W.); (J.S.)
| | - Xiangling Shi
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China; (H.X.); (Y.W.); (X.S.); (W.L.); (S.W.); (J.S.)
| | - Wang Liao
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China; (H.X.); (Y.W.); (X.S.); (W.L.); (S.W.); (J.S.)
| | - Shaokang Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China; (H.X.); (Y.W.); (X.S.); (W.L.); (S.W.); (J.S.)
| | - Jing Sui
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China; (H.X.); (Y.W.); (X.S.); (W.L.); (S.W.); (J.S.)
- Research Institute for Environment and Health, Nanjing University of Information Science and Technology, Nanjing 211544, China
| | - Guiju Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China; (H.X.); (Y.W.); (X.S.); (W.L.); (S.W.); (J.S.)
| |
Collapse
|
21
|
Li X, Huang G, Zhang Y, Ren Y, Zhang R, Zhu W, Yu K. Succinate signaling attenuates high-fat diet-induced metabolic disturbance and intestinal barrier dysfunction. Pharmacol Res 2023; 194:106865. [PMID: 37482326 DOI: 10.1016/j.phrs.2023.106865] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 07/02/2023] [Accepted: 07/21/2023] [Indexed: 07/25/2023]
Abstract
Succinate is a vital signaling metabolite produced by the host and gut microbiota. Succinate has been shown to regulate host metabolic homeostasis and inhibit obesity-associated inflammation in macrophages by engaging its cognate receptor, SUCNR1. However, the contribution of the succinate-SUCNR1 axis to intestinal barrier dysfunction in obesity remains unclear. In the present study, we explored the effects of succinate-SUCNR1 signaling on high-fat diet (HFD)-induced intestinal barrier dysfunction. Using a SUCNR1-deficient mouse model under HFD feeding conditions, we identified the effects of succinate-SUCNR1 axis on obesity-associated intestinal barrier impairment. Our results showed that HFD administration decreased goblet cell numbers and mucus production, promoted intestinal pro-inflammatory responses, induced gut microbiota composition imbalance, increased intestinal permeability, and caused mucosal barrier dysfunction. Dietary succinate supplementation was sufficient to activate a type 2 immune response, trigger the differentiation of barrier-promoting goblet cells, suppress intestinal inflammation, restore HFD-induced mucosal barrier impairment and intestinal dysbiosis, and eventually exert anti-obesity effects. However, SUNNR1-deficient mice failed to improve the intestinal barrier function and metabolic phenotype in HFD mice. Our data indicate the protective role of the succinate-SUCNR1 axis in HFD-induced intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Xuan Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Guowen Huang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Yanan Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuting Ren
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Ruofan Zhang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Kaifan Yu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
22
|
Liu S, Deng X, Li Z, Zhou W, Wang G, Zhan J, Hu B. Environmental cadmium exposure alters the internal microbiota and metabolome of Sprague-Dawley rats. Front Vet Sci 2023; 10:1219729. [PMID: 37565077 PMCID: PMC10410080 DOI: 10.3389/fvets.2023.1219729] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023] Open
Abstract
Cadmium (Cd) is a toxic element that can negatively affect both humans and animals. It enters the human and animal bodies through the respiratory and digestive tracts, following which it tends to accumulate in different organs, thereby seriously affecting human and animal health, as well as hampering social and economic development. Cd exposure can alter the composition of intestinal microbiota. In addition, it can damage the peripheral organs by causing the translocation of intestinal microbiota. However, the relationship between translocation-induced changes in the composition of microbiome in the blood and metabolic changes remains unclear. In the present study, we investigated the effects of Cd exposure on microbiota and serum metabolism in rats by omics analysis. The results demonstrated that Cd exposure disrupted the balance between the blood and intestinal flora in Sprague-Dawley (SD) rats, with a significant increase in gut microbiota (Clostridia_UCG_014, NK4A214_group) and blood microbiome (Corynebacterium, Muribaculaceae). However, Cd exposure caused the translocation of Corynebacterium and Muribaculaceae from the gut into the blood. In addition, Cd exposure was associated with the up-regulation of serum indoxyl sulfate, phenyl sulfate, and p-cresol sulfate; down-regulation of δ-tocopherol and L-glutamine; and changes in blood microbiome and metabolites. In conclusion, we identified novel metabolic biomarkers for Cd toxicity, which will also expand our understanding of the role of blood microbiome in Cd-induced injury.
Collapse
Affiliation(s)
- Songqing Liu
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
- Sichuan Provincial Key Laboratory for Development and Utilization of Characteristic Horticultural Biological Resources, Chengdu Normal University, Chengdu, China
| | - Xin Deng
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Zheng Li
- North Sichuan Medical College, Nanchong, China
| | - Wenjing Zhou
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
| | - Gang Wang
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
- Sichuan Provincial Key Laboratory for Development and Utilization of Characteristic Horticultural Biological Resources, Chengdu Normal University, Chengdu, China
| | - Jiasui Zhan
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Binhong Hu
- College of Chemistry and Life Sciences, Chengdu Normal University, Chengdu, China
- Sichuan Provincial Key Laboratory for Development and Utilization of Characteristic Horticultural Biological Resources, Chengdu Normal University, Chengdu, China
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
23
|
Chen X, Ran J, Mazhar M, Zhu Y, Lin Y, Qin L, Miao S. The balanced unsaturated fatty acid supplement constituted by woody edible oils improved lipid metabolism and gut microbiota in high-fat diet mice. Front Nutr 2023; 10:1203932. [PMID: 37545586 PMCID: PMC10399753 DOI: 10.3389/fnut.2023.1203932] [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: 04/11/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
The dietary intervention has demonstrated effectiveness in improving hyperlipidemia and obesity. Woody edible oils are rich in unsaturated fatty acids (UFAs) that could positively affect lipid metabolism. In this study, the blended oil (BLO), a balanced UFA supplement, constituted by Zanthoxylum bungeanum (Chinese Red Pepper) seed oil, walnut (Juglans regia) oil, camellia (Camema oleifera) seed oil and perilla (Perilla frutescens) seed oil was established referring to the Chinese dietary reference intakes, in which the ratios of monounsaturated/polyunsaturated fatty acids and ω-6/ω-3 polyunsaturated fatty acids were 1:1 and 4:1, respectively. The BLO was administrated to KM mice fed a high-fat diet (HFD) by gavage every day at a dose of 3.0 mL/kg·bw for 10 weeks to assess its effects on serum lipid levels, liver antioxidant activities and gut microbial composition. The results showed that the BLO improved hepatic steatosis, liver oxidative stress, and serum lipid levels. Additionally, there was an increased abundance of Lactobacillus, Allobaculum, and Blautia, along with a decreased abundance of Staphylococcus in cecal contents. These changes were found to be positively correlated with the metabolic improvements, as indicated by Spearman's correlation analysis. These findings implied the practicality of the balanced unsaturated fatty acid consumption in preventing hyperlipidemia and obesity.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, China
- Department of Laboratory Medicine, Affiliated Jinyang Hospital of Guizhou Medical University, Guiyang, China
| | - Jingqi Ran
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Muhammad Mazhar
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, China
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Yong Zhu
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Yichen Lin
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | - Likang Qin
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-Bioengineering, Guizhou University, Guiyang, China
- School of Liquor and Food Engineering, Guizhou University, Guiyang, China
| | - Song Miao
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| |
Collapse
|
24
|
Lynch JB, Gonzalez EL, Choy K, Faull KF, Jewell T, Arellano A, Liang J, Yu KB, Paramo J, Hsiao EY. Gut microbiota Turicibacter strains differentially modify bile acids and host lipids. Nat Commun 2023; 14:3669. [PMID: 37339963 DOI: 10.1038/s41467-023-39403-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/07/2023] [Indexed: 06/22/2023] Open
Abstract
Bacteria from the Turicibacter genus are prominent members of the mammalian gut microbiota and correlate with alterations in dietary fat and body weight, but the specific connections between these symbionts and host physiology are poorly understood. To address this knowledge gap, we characterize a diverse set of mouse- and human-derived Turicibacter isolates, and find they group into clades that differ in their transformations of specific bile acids. We identify Turicibacter bile salt hydrolases that confer strain-specific differences in bile deconjugation. Using male and female gnotobiotic mice, we find colonization with individual Turicibacter strains leads to changes in host bile acid profiles, generally aligning with those produced in vitro. Further, colonizing mice with another bacterium exogenously expressing bile-modifying genes from Turicibacter strains decreases serum cholesterol, triglycerides, and adipose tissue mass. This identifies genes that enable Turicibacter strains to modify host bile acids and lipid metabolism, and positions Turicibacter bacteria as modulators of host fat biology.
Collapse
Affiliation(s)
- Jonathan B Lynch
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Erika L Gonzalez
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kayli Choy
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kym F Faull
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Pasarow Mass Spectrometry Laboratory, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | | | | | | | - Kristie B Yu
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jorge Paramo
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Elaine Y Hsiao
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
25
|
Chu C, Li T, Yu L, Li Y, Li M, Guo M, Zhao J, Zhai Q, Tian F, Chen W. A Low-Protein, High-Carbohydrate Diet Exerts a Neuroprotective Effect on Mice with 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-Induced Parkinson's Disease by Regulating the Microbiota-Metabolite-Brain Axis and Fibroblast Growth Factor 21. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37267589 DOI: 10.1021/acs.jafc.2c07606] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Parkinson's disease (PD) is closely linked to lifestyle factors, particularly dietary patterns, which have attracted interest as potential disease-modifying factors. Eating a low-protein, high-carbohydrate (LPHC) diet is a promising dietary intervention against brain aging; however, its protective effect on PD remains elusive. Here, we found that an LPHC diet ameliorated 1-methyl-4-phenyl-1,2,3,6-tetrathydropyridine (MPTP)-induced motor deficits, decreased dopaminergic neuronal death, and increased the levels of striatal dopamine, serotonin, and their metabolites in PD mice. Levels of fibroblast growth factor 21 (FGF-21), a member of the fibroblast growth factor family, were elevated in PD mice following LPHC treatment. Furthermore, the administration of FGF-21 exerted a protective effect on MPTP-induced PC12 cells, similar to the effect of an LPHC diet in MPTP-induced mice. Sequencing of the 16S rDNA from fecal microbiota revealed that an LPHC diet normalized the gut bacterial composition imbalance in PD mice, as evidenced by the increased abundance of the genera Bifidobacterium, Ileibacterium, Turicibacter, and Blautia and decreased abundance of Bilophila, Alistipes, and Bacteroides. PICRUSt-predicted fecal microbiome function revealed that an LPHC diet suppressed lipopolysaccharide biosynthesis and the citrate cycle (TCA cycle), biosynthesis of ubiquinone and other terpenoid-quinones, and oxidative phosphorylation pathways caused by MPTP, and enhanced the biosynthesis of amino acids, carbohydrate metabolism, and biosynthesis of other secondary metabolites. A nonmetabolomic analysis of the serum and feces showed that an LPHC diet significantly increased the levels of aromatic amino acids (AAAs), including tryptophan, tyrosine, and phenylalanine. In addition, an LPHC diet elevated the serum concentrations of bile acids (BAs), particularly tauroursodeoxycholic acid (TUDCA) and taurine. Collectively, our current findings point to the potential mechanism of administering an LPHC diet in attenuating movement impairments in MPTP-induced PD mice, with AAAs, microbial metabolites (TUDCA and taurine), and FGF-21 as key mediators along the gut-microbiota-brain axis.
Collapse
Affiliation(s)
- Chuanqi Chu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Tiantian Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yiwen Li
- Department of Food Science and Technology, The University of Georgia, Athens, Georgia 30602, United States
| | - Miaoyu Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Min Guo
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
26
|
Chu C, Yu L, Li Y, Guo H, Zhai Q, Chen W, Tian F. Lactobacillus plantarum CCFM405 against Rotenone-Induced Parkinson’s Disease Mice via Regulating Gut Microbiota and Branched-Chain Amino Acids Biosynthesis. Nutrients 2023; 15:nu15071737. [PMID: 37049578 PMCID: PMC10096885 DOI: 10.3390/nu15071737] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Recent studies have demonstrated that disturbances in the gut microbiota and microbiota -derived metabolites contribute to the pathogenesis of Parkinson’s disease (PD), suggesting that probiotic treatments that restore them may delay disease progression. This study aimed to examine the attenuating efficacy of L. plantarum CCFM405 and the potential mechanisms in mice with rotenone-induced PD. Our results indicate that L. plantarum CCFM405 ameliorated rotenone-induced motor deficits and constipation, decreased dopaminergic neuronal death, reduced intestinal inflammation and neuroinflammation, and raised dopamine levels, 5-HT, and associated metabolites in the striatal region of the brain in mice with PD. Sequencing of 16S rRNA from fecal microbiota revealed that L. plantarum CCFM405 normalized the gut bacterial composition in mice with PD, as evidenced by the increased relative abundance of the following genus, Bifidobacterium, Turicibacter, and Faecalibaculum, and decreased relative abundance of Alistipes, Bilophila, Akkermansia, and Escherichia-Shigella. The PICRUSt-predicted gut microbiota function revealed that L. plantarum CCFM405 enhanced the biosynthesis of amino acid pathways, particularly valine, leucine, and isoleucine (branched-chain amino acids, BCAAs). A non-metabolomic analysis of the serum and feces showed that L. plantarum CCFM405 markedly increased the levels of BCAAs. Pathway enrichment analysis based on the KEGG database further suggested that L. plantarum CCFM405 supplementation can promote BCAAs biosynthesis. Collectively, L. plantarum CCFM405 can help to prevent rotenone-induced PD by modulating the gut microbiota–metabolite axis. BCAAs may play a dominant role in L. plantarum CCFM405-associated neuroprotection in PD mice. This probiotic could be utilized as a potential food supplement in the management of PD.
Collapse
Affiliation(s)
- Chuanqi Chu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yiwen Li
- Department of Food Science and Technology, The University of Georgia, Athens, GA 30602, USA
| | - Hang Guo
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
27
|
Zhao N, Ma Y, Liang X, Zhang Y, Hong D, Wang Y, Bai D. Efficacy and Mechanism of Qianshan Huoxue Gao in Acute Coronary Syndrome via Regulation of Intestinal Flora and Metabolites. Drug Des Devel Ther 2023; 17:579-595. [PMID: 36855515 PMCID: PMC9968440 DOI: 10.2147/dddt.s396649] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Purpose To study the efficacy of Qianshan Huoxue Gao (QS) in treating acute coronary syndrome (ACS) and to explore the mechanism of action from the perspective of intestinal flora regulation. Methods Male Sprague-Dawley rats were divided into control, model, QS, and atorvastatin groups; except for the control group, rats underwent ligation of the left anterior descending branch of the coronary artery. Following treatment for 28 days, cardiac function was evaluated using an echocardiographic assay; ELISAs for serum creatine kinase isoenzyme (CK-MB), cardiac troponin I (cTnI), high-sensitivity C-reactive protein (hs-CRP), interleukin (IL)-2 (IL-2), IL-6, and tumor necrosis factor-α (TNF-α); assessment of cardiac enzymes and inflammatory response; hematoxylin and eosin (HE) staining for histopathological changes in the heart, skin, and viscera; 16S rRNA gene sequencing for intestinal flora diversity and structural differences analysis; and we further investigated intestinal contents using metabolomics. Results Compared with controls, CK-MB and cTnI were increased (P<0.01); ejection factor and fractional shortening were decreased (P<0.01); left ventricular internal end-diastolic dimension and left ventricular internal end-systolic dimension were increased (P<0.01); and IL-2, IL-6, TNF-α, and hs-CRP were increased in the model group. Myocardial damage and inflammation were also observed by HE staining. QS improved these indexes, similar to the atorvastatin group; therefore, QS could effectively treat ACS. QS modulates the structure and abundance of the intestinal flora in ACS model rats, among which Bacteroides, Lactobacillus, and Rikenellaceae_RC9_gut_group are associated with cardiovascular disease. Metabolomics revealed that the intestinal metabolite content changed in ACS, with ethanolamine (EA) being the most relevant metabolite for ACS treatment by QS. EA was significantly positively correlated with Eubacterium xylanophilum group, Ruminococcus, unclassified f__Oscillospiraceae, Intestinimonas, Eubacterium siraeum group, Lachnospiraceae NK4A136 group, and norank f__Desulfovibrionaceae. Conclusion QS can effectively treat ACS and can restore regulation of the intestinal flora. EA may be the primary metabolite of QS, exerting a therapeutic effect in ACS.
Collapse
Affiliation(s)
- Ning Zhao
- Department of Pharmacy, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China,Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yan Ma
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | - Xiaoxue Liang
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Yu Zhang
- Beijing Xiuzheng Pharmaceutical Company, Beijing, People’s Republic of China
| | - Dacheng Hong
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Ying Wang
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Dong Bai
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China,Correspondence: Dong Bai, Tel +86 13552343081, Fax +8610 64089002, Email
| |
Collapse
|
28
|
Jiang X, Yang Q, Qu H, Chen Y, Zhu S. Endogenous n-3 PUFAs Improve Non-Alcoholic Fatty Liver Disease through FFAR4-Mediated Gut-Liver Crosstalk. Nutrients 2023; 15:nu15030586. [PMID: 36771292 PMCID: PMC9919706 DOI: 10.3390/nu15030586] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
The gut-liver axis plays a key role in the development and progression of non-alcoholic fatty liver disease (NAFLD). Due to the complexity and incomplete understanding of the cross-talk between the gut and liver, effective therapeutic targets are largely unknown. Free fatty acid receptors (FFARs) may bridge the cross-talk between the gut and liver. FFAR4 has received considerable attention due to its important role in lipid metabolism. However, the role of FFAR4 in this cross talk in NAFLD remains unclear. In this study, mice with high endogenous n-3 PUFAs but FFAR4 deficiency were generated by crossbreeding Fat-1 and FFAR4 knockout mice. FFAR4 deficiency blocked the protective effects of high endogenous n-3 PUFAs on intestinal barrier dysfunction and hepatic steatosis. In addition, FFAR4 deficiency decreased gut microbiota diversity and enriched Rikenella, Anaerotruncus, and Enterococcus, and reduced Dubosiella, Ruminococcaceae UCG-010, Ruminococcaceae UCG-014, Coriobacteriaceae UCG-002, Faecalibaculum, Ruminococcaceae UCG-009, and Akkermansia. Notably, FFAR4 deficiency co-regulated pantothenic acid and CoA biosynthesis, β-alanine metabolism, and sphingolipid metabolism pathways in the gut and liver, potentially associated with the aggravation of NAFLD. Together, the beneficial effects of n-3 PUFAs on the gut and liver were mediated by FFAR4, providing insights on the role of FFAR4 in the treatment of NAFLD through the gut-liver axis.
Collapse
Affiliation(s)
- Xuan Jiang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qin Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Hongyan Qu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Yongquan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and School of Translational Medicine, Jiangnan University, Wuxi 214122, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and School of Translational Medicine, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
29
|
Zhao N, Wang Y, Ma Y, Liang X, Zhang X, Gao Y, Dong Y, Bai D, Hu J. Jia-Wei-Si-Miao-Yong-An decoction modulates intestinal flora and metabolites in acute coronary syndrome model. Front Cardiovasc Med 2023; 9:1038273. [PMID: 36684592 PMCID: PMC9845626 DOI: 10.3389/fcvm.2022.1038273] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Aims We assessed the efficacy of the traditional Chinese medicine formulation Jia-Wei-Si-Miao-Yong-An decoction (HJ11) in the treatment of acute coronary syndrome and evaluated its impact on the intestinal microbiota and their metabolites. Methods An acute coronary syndrome model was established in rats, which were randomly assigned to the model, HJ11 treatment, and atorvastatin treatment groups. Rats were then administered saline solution (model and sham operation control groups) or drugs by oral gavage for 28 d. Echocardiography was performed and serum creatine kinase-MB and cardiac troponin I levels were monitored to examine the cardiac function. Inflammation was evaluated using hematoxylin and eosin staining of heart tissue, and serum interleukin-2, interleukin-6, tumor necrosis factor alpha, and high-sensitivity C-reactive protein measurements. Gut microbiota composition was analyzed via 16S rRNA gene sequencing. Metabolomics was used to determine fecal metabolites and elucidate the modes of action of HJ11 in acute coronary syndrome treatment. Results HJ11 improved cardiac function and attenuated inflammation in rats with acute coronary syndrome. Relative to the untreated model group, the HJ11-treated group presented normalized Firmicutes/Bacteroidetes ratio and reduced abundances of the bacterial genera norank_f__Ruminococcaceae, Desulfovibrio, Clostridium_sensu_stricto_1, Adlercreutzia, Staphylococcus, Bacteroides, Prevotella, Rikenellaceae_RC9_gut_group, unclassified_o__Bacteroidales, and Ruminococcus_gauvreauii_group. We found 23 differentially expressed intestinal metabolites, and the enriched metabolic pathways were mainly related to amino acid metabolism. We also discovered that asymmetric dimethylarginine levels were strongly associated with cardiovascular disease. Correlation analyses revealed strong associations among intestinal microflora, their metabolites, proinflammatory factors, and cardiac function. Hence, the therapeutic effects of HJ11 on acute coronary syndrome are related to specific alterations in gut microbiota and their metabolites. Conclusion This work demonstrated that HJ11 effectively treats acute coronary syndrome. HJ11 seems to increase the abundance of beneficial bacterial taxa (Bacteroides and Rikenellaceae_RC9_gut_group), mitigate the risk factors associated with cardiovascular disease, alter bacterial metabolites, lower asymmetric dimethylarginine levels, and effectively treat acute coronary syndrome.
Collapse
Affiliation(s)
- Ning Zhao
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Pharmacy, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Wang
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan Ma
- Department of Pathophysiology and Allergy Research, Vienna General Hospital, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Xiaoxue Liang
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xi Zhang
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuan Gao
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingying Dong
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dong Bai
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingqing Hu
- Formula-Syndrome Research Center, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
30
|
Wang L, Zhang Z, Luo G, Wang Y, Du K, Gao X. Metabolome combined with gut microbiome revealed the lipid-lowering mechanism of Xuezhiping capsule on hyperlipidemic hamster induced by high fat diet. Front Mol Biosci 2023; 10:1147910. [PMID: 36891237 PMCID: PMC9986548 DOI: 10.3389/fmolb.2023.1147910] [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: 01/19/2023] [Accepted: 02/08/2023] [Indexed: 02/22/2023] Open
Abstract
Introduction: Hyperlipidemia is a common metabolic disorder with presence of excess fat or lipids in the blood, may induce liver injury, oxidative stress and inflammatory. Xuezhiping capsule (XZP) is a famous Chinese patent medicine clinically used for anti-hyperlipidemia. However, the regulation mechanism of XZP on hyperlipidemia has not been elucidated so far. Methods: This study aimed to explore the effects of XZP on hypolipidemic, antioxidant and anti-inflammatory effects, and the potential mechanism by a combination of untargeted metabolomics and 16S rRNA sequencing. Results: The results indicated that XZP reduced the level of total cholesterol (TC), triglyceride (TG), low density lipoprotein cholesterol (LDL-C), increased the level of high density liptein cholesterol (HDL-C), alleviated excessive accumulation of lipid droplets in liver. Biochemical indexes of liver function including gamma glutamyl transferase (GGT) and glutamic oxaloacetic transaminase (GOT) in liver were remarkably decreased. Meanwhile, XZP increased the level of oxidative stress biochemical indexes including superoxide dismutase (SOD) and glutathione (GSH). In addition, XZP increased the level of peroxisome proliferators-activated receptors α (PPARα), acetyl CoA carboxylase 1 (ACOX1) and cholesterol 7-alpha hydroxylase (CYP7A1) in liver, and improved lipid metabolism in serum, liver and fecal lipid metabolism. XZP increased diversity index and the ratio of Firmicutes and Bacteroidetes, regulated seventeen genera, and illustrated strong correlations with liver lipid metabolism and phenotypic indicators. Discussion: These findings suggest that XZP reduced blood lipid and liver lipid, protected liver function, anti inflammation and anti-oxidation, ameliorate lipid metabolic disorders by modulating alpha linolenic acid and linoleic acid metabolism, bile acid metabolism, arachidonic acid metabolism, and regulated gut microbiota composition of high-fat diet (HFD) hamsters.
Collapse
Affiliation(s)
- Li Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhixin Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Gan Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ke Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyan Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
31
|
Li H, Wang Y, Shao S, Yu H, Wang D, Li C, Yuan Q, Liu W, Cao J, Wang X, Guo H, Wu X, Wang S. Rabdosia serra alleviates dextran sulfate sodium salt-induced colitis in mice through anti-inflammation, regulating Th17/Treg balance, maintaining intestinal barrier integrity, and modulating gut microbiota. J Pharm Anal 2022; 12:824-838. [PMID: 36605573 PMCID: PMC9805946 DOI: 10.1016/j.jpha.2022.08.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 02/01/2023] Open
Abstract
Rabdosia serra (R. serra), an important component of Chinese herbal tea, has traditionally been used to treat hepatitis, jaundice, cholecystitis, and colitis. However, the chemical composition of R. serra and its effect against colitis remain unclear. In this study, the chemical composition of the water extract of R. serra was analyzed using ultra performance liquid chromatography coupled with a hybrid linear ion trap quadrupole-orbitrap mass spectrometer (UPLC-LTQ-Orbitrap-MS). A total of 46 compounds, comprising ent-kaurane diterpenoids, flavonoids, phenolic acids, and steroids, were identified in the water extract of R. serra, and the extract could significantly alleviate dextran sulfate sodium salt-induced colitis by improving colon length, upregulating anti-inflammatory factors, downregulating proinflammatory factors, and restoring the balance of T helper 17/T regulatory cells. R. serra also preserved intestinal barrier function by increasing the level of tight junction proteins (zonula occludens 1 and occludin) in mouse colonic tissue. In addition, R. serra modulated the gut microbiota composition by increasing bacterial richness and diversity, increasing the abundance of beneficial bacteria (Muribaculaceae, Bacteroides, Lactobacillus, and Prevotellaceae_UCG-001), and decreasing the abundance of pathogenic bacteria (Turicibacter, Eubacterium_fissicatena_group, and Eubacterium_xylanophilum_group). Gut microbiota depletion by antibiotics further confirmed that R. serra alleviated colitis in a microbiota-dependent manner. Overall, our findings provide chemical and biological evidence for the potential application of R. serra in the management of colitis.
Collapse
Affiliation(s)
- Hongyi Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Yi Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shumin Shao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Hui Yu
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou, 510000, China
| | - Deqin Wang
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou, 510000, China
| | - Chuyuan Li
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou, 510000, China
| | - Qin Yuan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Wen Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Jiliang Cao
- College of Pharmacy, Shenzhen Technology University, Shenzhen, Guangdong, 518118, China
| | - Xiaojuan Wang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Haibiao Guo
- Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou, 510000, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Corresponding author.
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
- Macau Centre for Research and Development in Chinese Medicine, University of Macau, Macao, 999078, China
- Corresponding author. State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China.
| |
Collapse
|
32
|
Oleic Acid Facilitates Cd Excretion by Increasing the Abundance of Burkholderia in Cd-Exposed Mice. Int J Mol Sci 2022; 23:ijms232314718. [PMID: 36499044 PMCID: PMC9741113 DOI: 10.3390/ijms232314718] [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: 10/28/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
As a global pollutant, cadmium (Cd) can easily enter the body through food chains, threatening human health. Most Cd is initially absorbed in the gut, with the gut microbiota playing a pivotal role in reducing Cd absorption and accumulation. This study assessed the effects of three fatty acids on Cd accumulation and toxicity in Cd-exposed mice. The results showed that oleic acid (OA) was the most effective in facilitating Cd excretion in mice among these fatty acids. The use of OA led to reduced Cd accumulation in the organs and increased Cd content in the feces. The metagenomic analysis of the gut microbiota showed that the genus Burkholderia was the most significantly restored by OA in Cd-exposed mice. Burkholderia cepacia, as the type species for the genus Burkholderia, also exhibited strong Cd tolerance after treatment with OA. Furthermore, the electron microscopy analysis showed that most of the Cd was adsorbed on the surface of B. cepacia, where the extracellular polymeric substances (EPSs) secreted by B. cepacia play a key role, displaying a strong capacity for Cd adsorption. The peak at 2355 cm-1 and the total sulfhydryl group content of EPSs showed significant increases following co-treatment with Cd and OA. The results demonstrated the potential roles that gut Burkholderia may play in OA-mediated Cd excretion in mice.
Collapse
|
33
|
Puerariae lobatae Radix Alleviates Pre-Eclampsia by Remodeling Gut Microbiota and Protecting the Gut and Placental Barriers. Nutrients 2022; 14:nu14235025. [PMID: 36501055 PMCID: PMC9738998 DOI: 10.3390/nu14235025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Pre-eclampsia (PE) is a serious pregnancy complication, and gut dysbiosis is an important cause of it. Puerariae lobatae Radix (PLR) is a medicine and food homologous species; however, its effect on PE is unclear. This study aimed to investigate the efficacy of PLR in alleviating PE and its mechanisms. We used an NG-nitro-L-arginine methyl ester (L-NAME)-induced PE mouse model to examine the efficacy of preventive and therapeutic PLR supplementation. The results showed that both PLR interventions alleviated hypertension and proteinuria, increased fetal and placental weights, and elevated the levels of VEGF and PlGF. Moreover, PLR protected the placenta from oxidative stress via activating the Nrf2/HO-1/NQO1 pathway and mitigated placental damage by increasing intestinal barrier markers (ZO-1, Occludin, and Claudin-1) expression and reducing lipopolysaccharide leakage. Notably, preventive PLR administration corrected gut dysbiosis in PE mice, as evidenced by the increased abundance and positive interactions of beneficial bacteria including Bifidobacterium, Blautia, and Turicibacter. Fecal microbiota transplantation confirmed that the gut microbiota partially mediated the beneficial effects of PLR on PE. Our findings revealed that modulating the gut microbiota is an effective strategy for the treatment of PE and highlighted that PLR might be used as an intestinal nutrient supplement in PE patients.
Collapse
|
34
|
Ramsay TG, Arfken AM, Summers KL. Enteroendocrine peptides, growth, and the microbiome during the porcine weaning transition. Anim Microbiome 2022; 4:56. [DOI: 10.1186/s42523-022-00206-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 09/28/2022] [Indexed: 11/19/2022] Open
Abstract
Abstract
Background
Growth rate in pigs can be affected by numerous factors that also affect feeding behavior and the microbiome. Recent studies report some communication between the microbiome and the enteroendocrine system. The present study examined if changes in the piglet microbiome between birth and during the weaning transition can be correlated either positively or negatively with growth rate and plasma concentrations of enteroendocrine peptides.
Results
During the post-weaning transition, a 49% reduction in average daily gain was observed at day 24 (P < 0.05) relative to day 21. Pigs recovered by day 28 with body weight and average daily gain increases of 17% and 175%, respectively relative to day 24 and the highest rate of gain was measured at day 35 (462 g/day). The time interval between day 21–24 had the highest number of correlations (n = 25) between the relative abundance differences in taxa over time and corresponding percent weight gain. Amplicon sequence variants with the greatest correlation with percent weight gain between day 21–24 belonged to families Prevotellaceae NK3B31 (ρ = 0.65, P < 0.001), Veillonellaceae (ρ = 0.63, P < 0.001) and Rikenellaceae RC9 (ρ = 0.62, P < 0.001). Seven taxa were positively correlated with percent weight gain between day 24–28. Eight taxa were positively correlated with percent weight gain between day 28–35, of which four were Clostridia. Only Lactobacillus reuteri was positively correlated across both day 24–28 and day 28–35 analyses. Insulin-like growth factor 1 (IGF-1; R2 = 0.61, P < 0.001), glucose-dependent insulinotropic polypeptide (GIP; R2 = 0.20, P < 0.001), glucagon-like peptide 1 (GLP-1; R2 = 0.51, P < 0.001), and glucagon-like peptide 2 (GLP-2; R2 = 0.21, P < 0.001) were significantly associated with the piglet fecal community NMDS, while serotonin showed no significant association (R2 = 0.03, P = 0.15). Higher concentrations of GLP-1 and GLP-2 characterized day 1 fecal communities, while GIP levels had the strongest relationship primarily with samples ordinated with the day 21 cluster.
Conclusions
Demonstration of an association of certain taxa with individual gut peptides at specific ages suggests the potential for the microbiome to elicit changes in the gut enteroendocrine system during early postnatal development in the pig.
Collapse
|
35
|
Gao X, Hu Y, Tao Y, Liu S, Chen H, Li J, Zhao Y, Sheng J, Tian Y, Fan Y. Cymbopogon citratus (DC.) Stapf aqueous extract ameliorates loperamide-induced constipation in mice by promoting gastrointestinal motility and regulating the gut microbiota. Front Microbiol 2022; 13:1017804. [PMID: 36267178 PMCID: PMC9578511 DOI: 10.3389/fmicb.2022.1017804] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/09/2022] [Indexed: 12/02/2022] Open
Abstract
Slow transit constipation (STC) is the most common type of functional constipation. Drugs with good effects and few side effects are urgently needed form the treatment of STC. Cymbopogon citratus (DC.) Stapf (CC) is an important medicinal and edible spice plant. The wide range of biological activities suggested that CC may have laxative effects, but thus far, it has not been reported. In this study, the loperamide-induced STC mouse model was used to evaluate the laxative effect of the aqueous extract of CC (CCAE), and the laxative mechanism was systematically explored from the perspectives of the enteric nervous system (ENS), neurotransmitter secretion, gastrointestinal motility factors, intestinal inflammation, gut barrier and gut microbiota. The results showed that CCAE not only decreased the serum vasoactive intestinal polypeptide (VIP), induced nitric oxide synthases (iNOS), and acetylcholinesterase (AchE) in STC mice but also increased the expression of gastrointestinal motility factors in colonic interstitial cells of Cajal (ICCs) and smooth muscle cells (SMCs), thereby significantly shortening the defecation time and improving the gastrointestinal transit rate. The significantly affected gastrointestinal motility factors included stem cell factor receptor (c-Kit), stem cell factor (SCF), anoctamin 1 (Ano1), ryanodine receptor 3 (RyR3), smooth muscle myosin light chain kinase (smMLCK) and Connexin 43 (Cx43). Meanwhile, CCAE could repair loperamide-induced intestinal inflammation and intestinal barrier damage by reducing the expression of the pro-inflammatory factor IL-1β and increasing the expression of the anti-inflammatory factor IL-10, chemical barrier (Muc-2) and mechanical barrier (Cldn4, Cldn12, Occludin, ZO-1, and ZO-2). Interestingly, CCAE could also partially restore loperamide-induced gut microbial dysbiosis in various aspects, such as microbial diversity, community structure and species composition. Importantly, we established a complex but clear network between gut microbiota and host parameters. Muribaculaceae, Lachnospiraceae and UCG-010 showed the most interesting associations with the laxative phenotypes; several other specific taxa showed significant associations with serum neurotransmitters, gastrointestinal motility factors, intestinal inflammation, and the gut barrier. These findings suggested that CCAE might promote intestinal motility by modulating the ENS-ICCs-SMCs network, intestinal inflammation, intestinal barrier and gut microbiota. CC may be an effective and safe therapeutic choice for STC.
Collapse
Affiliation(s)
- Xiaoyu Gao
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
| | - Yifan Hu
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming, China
- College of Agronomy and Biotechnology, Yunnan Agricultural University, Kunming, China
| | - Yafei Tao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Shuangfeng Liu
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Haowen Chen
- College of Agronomy and Biotechnology, Yunnan Agricultural University, Kunming, China
| | - Jiayi Li
- College of Agronomy and Biotechnology, Yunnan Agricultural University, Kunming, China
| | - Yan Zhao
- Department of Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jun Sheng
- Yunnan Provincial Engineering Research Center for Edible and Medicinal Homologous Functional Food, Yunnan Agricultural University, Kunming, China
- *Correspondence: Jun Sheng,
| | - Yang Tian
- Yunnan Provincial Engineering Research Center for Edible and Medicinal Homologous Functional Food, Yunnan Agricultural University, Kunming, China
- Yang Tian,
| | - Yuanhong Fan
- Yunnan Plateau Characteristic Agricultural Industry Research Institute, Yunnan Agricultural University, Kunming, China
- Yuanhong Fan,
| |
Collapse
|
36
|
Chen G, Peng Y, Huang Y, Xie M, Dai Z, Cai H, Dong W, Xu W, Xie Z, Chen D, Fan X, Zhou W, Kan X, Yang T, Chen C, Sun Y, Zeng X, Liu Z. Fluoride induced leaky gut and bloom of Erysipelatoclostridium ramosum mediate the exacerbation of obesity in high-fat-diet fed mice. J Adv Res 2022:S2090-1232(22)00239-9. [PMID: 36341987 PMCID: PMC10403698 DOI: 10.1016/j.jare.2022.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/18/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Fluoride is widely presented in drinking water and foods. A strong relation between fluoride exposure and obesity has been reported. However, the potential mechanisms on fluoride-induced obesity remain unexplored. Objectives and methods The effects of fluoride on the obesity were investigated using mice model. Furthermore, the role of gut homeostasis in exacerbation of the obesity induced by fluoride was evaluated. Results The results showed that fluoride alone did not induce obesity in normal diet (ND) fed mice, whereas, it could trigger exacerbation of obesity in high-fat diet (HFD) fed mice. Fluoride impaired intestinal barrier and activated Toll-like receptor 4 (TLR4) signaling to induce obesity, which was further verified in TLR4-/- mice. Furthermore, fluoride could deteriorate the gut microbiota in HFD mice. The fecal microbiota transplantation from fluoride-induced mice was sufficient to induce obesity, while the exacerbation of obesity by fluoride was blocked upon gut microbiota depletion. The fluoride-induced bloom of Erysipelatoclostridium ramosum was responsible for exacerbation of obesity. In addition, a potential strategy for prevention of fluoride-induced obesity was proposed by intervention with polysaccharides from Fuzhuan brick tea. Conclusion Overall, these results provide the first evidence of a comprehensive cross-talk mechanism between fluoride and obesity in HFD fed mice, which is mediated by gut microbiota and intestinal barrier. E. ramosum was identified as a crucial mediator of fluoride induced obesity, which could be explored as potential target for prevention and treatment of obesity with exciting translational value.
Collapse
|
37
|
High fat diet-induced hyperlipidemia and tissue steatosis in rabbits through modulating ileal microbiota. Appl Microbiol Biotechnol 2022; 106:7187-7207. [PMID: 36173452 DOI: 10.1007/s00253-022-12203-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/02/2022]
Abstract
High-fat diet (HFD) and overnutrition are important starting factors that may alter intestinal microbiota, lipid metabolism, and systemic inflammation. However, there were few studies on how intestinal microbiota contributes to tissue steatosis and hyperlipidemia. Here, we investigated the effect of lipid metabolism disorder-induced inflammation via toll-like receptor 2 (TLR-2), toll-like receptor 4 (TLR-4), and nuclear factor-κB (NF-κB) pathways at the intestinal level in response to HFD. Twenty 80-day-old male New Zealand White rabbits were randomly divided into the normal diet group (NDG) and the high-fat diet group (HDG) for 80 days. Growth performance, blood biochemical parameters, lipid metabolism, inflammation, degree of tissue steatosis, and intestinal microbial composition were measured. HFD increased the relative abundance of Christensenellaceae_R_7_group, Marvinbryantia, Akkermansia etc., with a reduced relative abundance of Enterorhabdus and Lactobacillus. Moreover, HFD caused steatosis in the liver and abdominal fat and abnormal expression of some genes related to lipid metabolism and tight junction proteins. The TLR-2, TLR-4, NF-κB, TNF-α, and IL-6 were confirmed by overexpression with downregulation of IL-10. Serum biochemical indices (TG, TCHO, LDL-C, and HDL-C) were also increased, indicating evidence for the development of the hyperlipidemia model. Correlation analysis showed that this microbial dysbiosis was correlated with lipid metabolism and inflammation, which were associated with the intestinal tract's barrier function and hyperlipidemia. These results provide an insight into the relationship between HFD, the intestinal microbiota, intestinal barrier, tissue inflammation, lipid metabolism, and hyperlipidemia. KEY POINTS: • High-fat diet leads to ileal microbiota disorders • Ileal microbiota mediates local and systemic lipid metabolism disorders and inflammation • There is a specific link between ileal microbiota, histopathology, and hyperlipidemia.
Collapse
|
38
|
Oyeyinka SA, Gbashi S, A. Onarinde B, Adebo OA, Njobeh PB. Metabolite profile of raw and cooked pasta from whole wheat grain enriched with Bambara groundnut. J FOOD PROCESS PRES 2022. [DOI: 10.1111/jfpp.17152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Samson A. Oyeyinka
- Centre of Excellence in Agr‐food Technologies, National Centre for Food Manufacturing University of Lincoln Holbeach, PE12 7PT UK
- Department of Biotechnology and Food Technology, Faculty of Science University of Johannesburg P.O. Box 17011, Doornfontein Campus Gauteng South Africa
| | - Sefater Gbashi
- Department of Biotechnology and Food Technology, Faculty of Science University of Johannesburg P.O. Box 17011, Doornfontein Campus Gauteng South Africa
| | - Bukola A. Onarinde
- Centre of Excellence in Agr‐food Technologies, National Centre for Food Manufacturing University of Lincoln Holbeach, PE12 7PT UK
| | - Oluwafemi A. Adebo
- Department of Biotechnology and Food Technology, Faculty of Science University of Johannesburg P.O. Box 17011, Doornfontein Campus Gauteng South Africa
| | - Patrick B. Njobeh
- Department of Biotechnology and Food Technology, Faculty of Science University of Johannesburg P.O. Box 17011, Doornfontein Campus Gauteng South Africa
| |
Collapse
|
39
|
Alteration of fecal microbiome and metabolome by mung bean coat improves diet-induced non-alcoholic fatty liver disease in mice. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.04.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
40
|
Zhou C, Zhang W, Lin H, Zhang L, Wu F, Wang Y, Yu S, Peng X, Cheng W, Li M, Pan X, Huang Z, Zhang W. Effect of theaflavin-3,3′-digallate on leptin-deficient induced nonalcoholic fatty liver disease might be related to lipid metabolism regulated by the Fads1/PPARδ/Fabp4 axis and gut microbiota. Front Pharmacol 2022; 13:925264. [PMID: 36105184 PMCID: PMC9464872 DOI: 10.3389/fphar.2022.925264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), one of the risk factors for hepatitis, cirrhosis, and even hepatic carcinoma, has been a global public health problem. The polyphenol compound theaflavin-3,3′-digallate (TF3), mainly extracted from black tea, has been reported to produce an effect on hypoglycemic and antilipid deposition in vitro. In our study, we further investigated the function and novel mechanisms of TF3 in protecting NAFLD in vivo. By using leptin-deficient obese (ob/ob) mice with NAFLD symptoms, TF3 treatment prevented body weight and waistline gain, reduced lipid accumulation, and alleviated liver function injury, as well as decreased serum lipid levels and TG levels in livers in ob/ob mice, observing no side effects. Furthermore, the transcriptome sequencing of liver tissue showed that TF3 treatment corrected the expression profiles of livers in ob/ob mice compared with that of the model group. It is interesting to note that TF3 might regulate lipid metabolism via the Fads1/PPARδ/Fabp4 axis. In addition, 16S rRNA sequencing demonstrated that TF3 increased the abundance of Prevotellaceae_UCG-001, norank_f_Ruminococcaceae, and GCA-900066575 and significantly decreased that of Parvibacter. Taken together, the effect of TF3 on NAFLD might be related to lipid metabolism regulated by the Fads1/PPARδ/Fabp4 axis and gut microbiota. TF3 might be a promising candidate for NAFLD therapy.
Collapse
Affiliation(s)
- Cheng Zhou
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Wenji Zhang
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Hui Lin
- Department of Radiation Oncology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Luyun Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Fan Wu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Yan Wang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Susu Yu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xinyue Peng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Wenli Cheng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Min Li
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xiaoying Pan
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Zhenrui Huang
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- *Correspondence: Zhenrui Huang, ; Wenjuan Zhang,
| | - Wenjuan Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
- *Correspondence: Zhenrui Huang, ; Wenjuan Zhang,
| |
Collapse
|
41
|
Bai Y, Ma K, Li J, Ren Z, Zhang J, Shan A. Lactobacillus rhamnosus GG ameliorates DON-induced intestinal damage depending on the enrichment of beneficial bacteria in weaned piglets. J Anim Sci Biotechnol 2022; 13:90. [PMID: 35962456 PMCID: PMC9375241 DOI: 10.1186/s40104-022-00737-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/01/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Deoxynivalenol (DON) is one of the most common environmental pollutants that induces intestinal inflammation and microbiota dysbiosis. Lactobacillus rhamnosus GG (LGG) is a probiotic that not only has anti-inflammatory effects, but also shows protective effect on the intestinal barrier. However, it is still unknown whether LGG exerts beneficial effects against DON-induced intestinal damage in piglets. In this work, a total of 36 weaned piglets were randomized to one of four treatment groups for 21 d. The treatment groups were CON (basal diet); LGG (basal diet supplemented with 1.77 × 1011 CFU/kg LGG); DON (DON-contaminated diet) and LGG + DON (DON-contaminated diet supplemented with 1.77 × 1011 CFU/kg LGG). RESULT Supplementation of LGG can enhance growth performance of piglets exposed to DON by improving intestinal barrier function. LGG has a mitigating effect on intestinal inflammation induced by DON exposure, largely through repression of the TLR4/NF-κB signaling pathway. Furthermore, supplementation of LGG increased the relative abundances of beneficial bacteria (e.g., Collinsella, Lactobacillus, Ruminococcus_torques_group and Anaerofustis), and decreased the relative abundances of harmful bacteria (e.g., Parabacteroides and Ruminiclostridium_6), and also promoted the production of SCFAs. CONCLUSIONS LGG ameliorates DON-induced intestinal damage, which may provide theoretical support for the application of LGG to alleviate the adverse effects induced by DON exposure.
Collapse
Affiliation(s)
- Yongsong Bai
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Kaidi Ma
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Jibo Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China
| | - Zhongshuai Ren
- College of Animal Sciences, Jilin University, Key Laboratory of Zoonosis Research, Ministry of Education, Changchun, 130062, P. R. China
| | - Jing Zhang
- College of Animal Sciences, Jilin University, Key Laboratory of Zoonosis Research, Ministry of Education, Changchun, 130062, P. R. China.
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, 150030, P. R. China.
| |
Collapse
|
42
|
Traditional Chinese Medicine Formula Jian Pi Tiao Gan Yin Reduces Obesity in Mice by Modulating the Gut Microbiota and Fecal Metabolism. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9727889. [PMID: 35979004 PMCID: PMC9377893 DOI: 10.1155/2022/9727889] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/10/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022]
Abstract
The current study employed the high-fat diet (HFD) induced murine model to assess the relationship between the effect of Jian Pi Tiao Gan Yin (JPTGY) and the alterations of gut microbiota and fecal metabolism. C57BL/6 mice were used to establish an animal model of obesity via HFD induce. Serum biochemical indicators of lipid metabolism were used to evaluate the pharmacodynamics of JPTGY in obese mice. Bacterial communities and metabolites in the feces specimens from the controls, the Group HFD, and the JPTGY-exposed corpulency group were studied by 16s rDNA genetic sequence in combination with liquid chromatography-mass spectrometry (LC-MS) based untargeted fecal metabolomics techniques. Results revealed that JPTGY significantly decreased the levels of total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and elevated high-density lipoprotein cholesterol (HDL-C). Moreover, JPTGY could up-regulate the abundance and diversity of fecal microbiota, which was characterized by the higher phylum of proteobacteria. Consistently, at the genus levels, JPTGY supplementation induced enrichments in Lachnospiraceae NK4A136 group, Oscillibacter, Turicibacter, Clostridium sensu stricto 1, and Intestinimonas, which were intimately related to 14 pivotal fecal metabolins in respond to JPTGY therapy were determined. What is more, metabolomics further analyses show that the therapeutic effect of JPTGY for obesity involves linoleic acid (LA) metabolism paths, alpha-linolenic acid (ALA) metabolism paths, glycerophospholipid metabolism paths, arachidonic acid (AA) metabolism paths, and pyrimidine metabolism paths, which implied the potential mechanism of JPTGY in treating obesity. It was concluded that the linking of corpulency phenotypes with intestinal flora and fecal metabolins unveils the latent causal link of JPTGY in the treatment of hyperlipidemia and obesity.
Collapse
|
43
|
Yang B, Huang S, Zhao G, Ma Q. Dietary supplementation of porcine bile acids improves laying performance, serum lipid metabolism and cecal microbiota in late-phase laying hens. ANIMAL NUTRITION 2022; 11:283-292. [PMID: 36263401 PMCID: PMC9576542 DOI: 10.1016/j.aninu.2022.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 12/23/2022]
Abstract
Due to the exceptional laying performance of hens, the demand on lipid metabolism and oxidation in vivo is vigorous, resulting in excessive lipid accumulation in late-phase hens, which lowers the production performance. Bile acids regulate lipid metabolism and gut microbiota in humans and animals. However, the effect of porcine bile acids on lipid metabolism and cecal microbiota in laying hens in the late phase is still unclear. A total of 360 healthy 45-week-old laying hens were chosen for a 24-week feeding trial, where 0, 30, 60 and 90 mg/kg porcine bile acids were added to a basal diet, respectively. The results showed that dietary supplementation of 60 mg/kg bile acids increased egg production and feed conversion (P < 0.05). Also, 60 and 90 mg/kg porcine bile acids reduced abdominal fat percentage and body weight (P < 0.05). The levels of serum total cholesterol, triglyceride, and low-density lipoprotein cholesterol of hens decreased (P < 0.05) in bile acids supplement groups. As for cecal microbiota, bile acids supplementation did not affect the alpha diversity of cecal microbiota at the genus level. Moreover, dietary supplementation of 90 mg/kg bile acids resulted in an increase in the abundance of beneficial bacteria in the cecum, such as Lactobacillus, Bifidobacterium and Turicibacter. The changes in the cecal microbiota caused by bile acids supplementation correlated with serum lipid indexes. According to KEGG pathway analysis, dietary supplementation of 60 and 90 mg/kg bile acids promoted structural transformation of the cecal microbiota to down-regulate steroid biosynthesis, up-regulate fatty acid degradation and up-regulate unsaturated fatty acid biosynthesis. Meanwhile, bile acids bio-isomerization function of cecal microbiota was enhanced in 60 and 90 mg/kg bile acids treatment, and the short-chain fatty acid metabolism was also affected. In conclusion, the present study revealed dietary supplementation of porcine bile acids enriched probiotics in the gut and improved serum lipid metabolism of laying hens. These findings demonstrate that porcine bile acids can be a potential gut beneficial promoter for late-phase laying hens.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Guoxian Zhao
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Qiugang Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Corresponding author.
| |
Collapse
|
44
|
Zhao W, Wang J, Latta M, Wang C, Liu Y, Ma W, Zhou Z, Hu S, Chen P, Liu Y. Rhizoma Gastrodiae Water Extract Modulates the Gut Microbiota and Pathological Changes of P-TauThr231 to Protect Against Cognitive Impairment in Mice. Front Pharmacol 2022; 13:903659. [PMID: 35910384 PMCID: PMC9335362 DOI: 10.3389/fphar.2022.903659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/09/2022] [Indexed: 11/30/2022] Open
Abstract
Gastrodiae Rhizoma and its active constituents are known to exhibit neuroprotective effects in Alzheimer’s disease (AD). However, the effect of Rhizoma Gastrodiae water extract (WERG) on AD and the detailed mechanism of action remain unclear. In this study, the mechanism of action of WERG was investigated by the microbiome–gut–brain axis using a D-galactose (D-gal)/AlCl3-induced AD mouse model. WERG improved the cognitive impairment of D-gal/AlCl3-induced mice. The expression level of p-Tauthr231 in the WERG-H treatment group was decreased, and p-Tauthr231 was found negative in hippocampal DG, CA1, and CA3 regions. Here, the diversity and composition of the gut microbiota were analyzed by 16sRNA sequencing. WERG-H treatment had a positive correlation with Firmicutes, Bacilli, Lactobacillus johnsonii, Lactobacillus murinus, and Lactobacillus reuteri. Interestingly, the Rikenellaceae-RC9 gut group in the gut increased in D-gal/AlCl3-induced mice, but the increased L. johnsonii, L. murinus, and L. reuteri reversed this process. This may be a potential mechanistic link between gut microbiota dysbiosis and P-TauThr231 levels in AD progression. In conclusion, this study demonstrated that WERG improved the cognitive impairment of the AD mouse model by enriching gut probiotics and reducing P-TauThr231 levels.
Collapse
Affiliation(s)
- Wenbin Zhao
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Jianhui Wang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Maria Latta
- School of Pharmacy, University of Connecticut, Mansfield, CT, United States
| | - Chenyu Wang
- School of Basic Medical Science, Lanzhou University, Lanzhou, China
| | - Yuheng Liu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Wantong Ma
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Zhongkun Zhou
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Shujian Hu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Peng Chen
- School of Pharmacy, Lanzhou University, Lanzhou, China
- *Correspondence: Peng Chen, ; Yingqian Liu,
| | - Yingqian Liu
- School of Pharmacy, Lanzhou University, Lanzhou, China
- *Correspondence: Peng Chen, ; Yingqian Liu,
| |
Collapse
|
45
|
Li J, Lian H, Zheng A, Zhang J, Dai P, Niu Y, Gao T, Li M, Zhang L, Fu T. Effects of Different Roughages on Growth Performance, Nutrient Digestibility, Ruminal Fermentation, and Microbial Community in Weaned Holstein Calves. Front Vet Sci 2022; 9:864320. [PMID: 35903131 PMCID: PMC9315432 DOI: 10.3389/fvets.2022.864320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
This study aimed to assess the effects of feeding with different forage sources and starter concentrations on growth performance, nutrient digestibility, ruminal fermentation, and the microbial community in weaned Holstein calves. A total of 54 Holstein calves (body weight (BW) = 77.50 ± 5.07 kg; age = 70 ± 2.54 days) were assigned to 1 of 3 treatment groups (n = 18/group) that were offered diets with different forages: (1) peanut vine (PV), (2) oat hay (OH), or (3) an alfalfa hay + oat hay combination (alfalfa hay:oat hay =1:1, AO). Starter and forage intakes were recorded daily, while BW and growth parameters were assessed at 15-day intervals. The apparent digestibility of nutrients was determined. Ruminal fluid samples were collected and used to detect relevant indicators. A difference was observed for the forage × age interaction for all feed, nutrient intake, BW, ADG, and body structure parameters (P < 0.05). The final BW, average daily feed intake (ADFI), and average daily gain of the PV calves were higher than those of calves from the other groups (P < 0.05). The ruminal propionate concentration evidently increased in calves of the AO group (P < 0.05). The abundances of Rikenellaceae_RC9_gut_group and Shuttleworthia showed distinct responses to feeding with different forages (P < 0.05) at the genus level. The relative abundance of Shuttleworthia was negatively related to rumen pH and acid detergent fiber digestibility (P < 0.05) and strongly positively related to propionate concentration (P < 0.01). A positive correlation was found between Ruminococcus_1 abundance and butyrate concentration and neutral detergent fiber digestibility (P < 0.05). The relative abundances of Succiniclasticum and Prevotella_7 were negatively related to butyrate concentration (P < 0.05). In conclusion, there was an interaction between the factors (forage × age). The peanut vine used as a forage source promoted a higher starter concentrate intake compared to other diets and increased with the calves' age. The growth performance and rumen bacterial community of the calves were further improved. These results indicate that peanut vine can be used as the main source of forage in the diets of weaned calves.
Collapse
Affiliation(s)
- Jichao Li
- Henan International Joint Laboratory of Nutrition Regulation and Ecological Raising of Domestic Animal, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Hongxia Lian
- Henan International Joint Laboratory of Nutrition Regulation and Ecological Raising of Domestic Animal, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Airong Zheng
- Henan Forage Feeding Technology Extension Station, Zhengzhou, China
| | - Jiangfan Zhang
- Henan International Joint Laboratory of Nutrition Regulation and Ecological Raising of Domestic Animal, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Pengfei Dai
- Henan International Joint Laboratory of Nutrition Regulation and Ecological Raising of Domestic Animal, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Yan Niu
- Henan Forage Feeding Technology Extension Station, Zhengzhou, China
| | - Tengyun Gao
- Henan International Joint Laboratory of Nutrition Regulation and Ecological Raising of Domestic Animal, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Ming Li
- Henan International Joint Laboratory of Nutrition Regulation and Ecological Raising of Domestic Animal, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Liyang Zhang
- Henan International Joint Laboratory of Nutrition Regulation and Ecological Raising of Domestic Animal, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- *Correspondence: Liyang Zhang
| | - Tong Fu
- Henan International Joint Laboratory of Nutrition Regulation and Ecological Raising of Domestic Animal, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
- Tong Fu
| |
Collapse
|
46
|
Ferulic acid improves intestinal barrier function through altering gut microbiota composition in high-fat diet-induced mice. Eur J Nutr 2022; 61:3767-3783. [PMID: 35732902 DOI: 10.1007/s00394-022-02927-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 05/31/2022] [Indexed: 12/16/2022]
Abstract
PURPOSE A high-fat diet (HFD) induces gut microbiota (GM) disorders, leading to intestinal barrier dysfunction and inflammation. Ferulic acid (FA) has shown anti-obesity effects, e.g., reducing body weight and food intake. However, the mechanism linking the anti-obesity effects of FA and GM modulation remains obscure. The present study aimed to clarify the mechanism underlying the anti-obesity effects of FA and modulation of the GM. METHODS C57BL/6 J mice were fed by a low-fat diet (LFD) and HFD with or without FA at a dose of 100 mg/kg of body weight by oral gavage for 12 weeks. Using high-throughput sequencing, gas chromatography, real-time fluorescence quantitative PCR and immunohistochemical staining, the attenuation of obesity by FA were assessed via intestinal barrier integrity, inflammation, and the GM. RESULTS FA reduced weight gain, improved HFD-induced GM imbalance, significantly enhanced intestinal short-chain fatty acid (SCFA)-producing bacteria (e.g., Olsenella, Eisenbergiella, Dubosiella, Clostridiales_unclassified, and Faecalibaculum) along with SCFA accumulation and its receptors' expression, decreased endotoxin-producing bacteria or obesity-related bacterial genera, and serum endotoxin (lipopolysaccharides), and inhibited the colonic TLR4/NF-κB pathway. Thus, FA can mitigate colonic barrier dysfunction and intestinal inflammation, induce the production of SCFAs and inhibit endotoxins by modulating the GM. CONCLUSION These results indicate that enhancement of intestinal barrier by altering the GM may be an anti-obesity target of FA and that FA can be used as a functional compound with great developmental values.
Collapse
|
47
|
Zhang R, Zhang Q, Chen Y, Zhao Q, Zhang B, Wang L, Zhou C, Zhang Q, Chen K, Zhang Y, Hou X, Chen H, Liu X, Ni M, Jiang B. Combined treatment with Rg1 and adipose-derived stem cells alleviates DSS-induced colitis in a mouse model. Stem Cell Res Ther 2022; 13:272. [PMID: 35729638 PMCID: PMC9210677 DOI: 10.1186/s13287-022-02940-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/13/2021] [Indexed: 11/27/2022] Open
Abstract
Background Inflammatory bowel diseases, consisting of Crohn’s disease and ulcerative colitis constitute chronic inflammatory conditions that may compromise the whole gastrointestinal tract as well as the colonic mucosa. Currently, there are no curative interventions for IBD, and all available treatments have side effects that limit their use. Adipose-derived stem cell (ADSC) treatment is a prospective treatment option for IBD. Previous findings indicated that ginsenoside (Rg1) dampened inflammatory diseases like colitis by inhibiting the binding of LPS to TLR4 on macrophages and restoring the Th17/Treg ratio. The purpose of this work was to investigate whether Rg1 can increase the influence of ADSC in a mouse model of colitis triggered by dextran sulfate sodium (DSS). Methods ADSC was intravenously inoculated into mice with DSS-triggered colitis, while Rg1 was delivered via oral gavage. Colon inflammation was assessed via body weight, colon length along with H&E staining. Serum cytokine levels were measured using ELISA. Besides, flow cytometry was adopted to determine the percentage, as well as FMI of immune cells in the spleen. The effects of simultaneous Rg1 and ADSC treatment on TLR4-MyD88 signaling were assessed via immunofluorescence. Results Rg1 and ADSC effectively alleviated the impacts of colon inflammation, weight loss, and colon length reduction along with histological score. Treatment with Rg1 and ADSC reduced serum levels of the proinflammatory cytokines, IL-1β, TNF-α, IL-6, IL-4, and IL-17A and upregulated the level of immunosuppressive cytokine, IL-10. Compared with ADSC or Rg1 alone, combined treatment with Rg1 and ADSC significantly improved the structure of microbial community. Additionally, treatment with Rg1 plus ADSC selectively elevated the level of splenic regulatory T (Treg) cells and downregulated the proportion of T helper type 17 (Th17) cells, indicating restoration of intestinal homeostasis. Besides, we established that the combination of ADSC + Rg1 restored immunological balance more effectively than either ADSC or Rg1 alone, illustrating that Rg1's modulatory function on the gut microbiota may boost the impact of ADSCs in restoration of the immune balance. ADSC combined with Rg1 might downregulate the expression of TLR4 and MyD88, thereby suppressing TLR4-MyD8 signaling. The immunofluorescence results also suggested that co-therapy with Rg-1 and ADSC may optimize treatment strategies of IBD. Conclusions Here, we find that the combination of Rg1 and ADSC alleviates DSS-induced colitis in a mouse model more efficiently than ADSC alone, indicating that Rg1 enhances the effect of ADSC against colitis. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02940-x.
Collapse
Affiliation(s)
- Rui Zhang
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China.,Graduate School of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Qingqing Zhang
- Department of Pathogen Biology, Key Laboratory of Pathogen of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China
| | - Yanni Chen
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China
| | - Qing Zhao
- Department of Pathogen Biology, Key Laboratory of Pathogen of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China
| | - Bo Zhang
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China
| | - Ling Wang
- Graduate School of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Chungen Zhou
- Colorectal Disease Center of Nanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China
| | - Qi Zhang
- Graduate School of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Kun Chen
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China
| | - Yuqing Zhang
- Graduate School of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Xiaotao Hou
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China
| | - Hao Chen
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China
| | - Xingyin Liu
- Department of Pathogen Biology, Key Laboratory of Pathogen of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China.
| | - Min Ni
- Colorectal Disease Center of Nanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China.
| | - Bin Jiang
- Colorectal Disease Center of Nanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China.
| |
Collapse
|
48
|
Cui J, Sachaphibulkij K, Teo WS, Lim HM, Zou L, Ong CN, Alberts R, Chen J, Lim LHK. Annexin-A1 deficiency attenuates stress-induced tumor growth via fatty acid metabolism in mice: an Integrated multiple omics analysis on the stress- microbiome-metabolite-epigenetic-oncology (SMMEO) axis. Theranostics 2022; 12:3794-3817. [PMID: 35664067 PMCID: PMC9131274 DOI: 10.7150/thno.68611] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/28/2022] [Indexed: 11/22/2022] Open
Abstract
Background: High emotional or psychophysical stress levels have been correlated with an increased risk and progression of various diseases. How stress impacts the gut microbiota to influence metabolism and subsequent cancer progression is unclear. Methods: Feces and serum samples from BALB/c ANXA1+/+ and ANXA1-/- mice with or without chronic restraint stress were used for 16S rRNA gene sequencing and GC-MS metabolomics analysis to investigate the effect of stress on microbiome and metabolomics during stress and breast tumorigenesis. Breast tumors samples from stressed and non-stressed mice were used to perform Whole-Genome Bisulfite Sequencing (WGBS) and RNAseq analysis to construct the potential network from candidate hub genes. Finally, machine learning and integrated analysis were used to map the axis from chronic restraint stress to breast cancer development. Results: We report that chronic stress promotes breast tumor growth via a stress-microbiome-metabolite-epigenetic-oncology (SMMEO) axis. Chronic restraint stress in mice alters the microbiome composition and fatty acids metabolism and induces an epigenetic signature in tumors xenografted after stress. Subsequent machine learning and systemic modeling analyses identified a significant correlation among microbiome composition, metabolites, and differentially methylated regions in stressed tumors. Moreover, silencing Annexin-A1 inhibits the changes in the gut microbiome and fatty acid metabolism after stress as well as basal and stress-induced tumor growth. Conclusions: These data support a physiological axis linking the microbiome and metabolites to cancer epigenetics and inflammation. The identification of this axis could propel the next phase of experimental discovery in further understanding the underlying molecular mechanism of tumorigenesis caused by physiological stress.
Collapse
Affiliation(s)
- Jianzhou Cui
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, NUS, Singapore
- NUS Immunology Program, Life Sciences Institute, NUS, Singapore
| | - Karishma Sachaphibulkij
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, NUS, Singapore
- NUS Immunology Program, Life Sciences Institute, NUS, Singapore
| | - Wen Shiun Teo
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, NUS, Singapore
- NUS Immunology Program, Life Sciences Institute, NUS, Singapore
| | - Hong Meng Lim
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, NUS, Singapore
- NUS Immunology Program, Life Sciences Institute, NUS, Singapore
| | - Li Zou
- Saw Swee Hock School of Public Health, NUS, Singapore
| | - Choon Nam Ong
- Saw Swee Hock School of Public Health, NUS, Singapore
- NUS Environmental Research Institute, NUS, Singapore
| | - Rudi Alberts
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, NUS, Singapore
| | - Jinmiao Chen
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Lina H. K. Lim
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, NUS, Singapore
- NUS Immunology Program, Life Sciences Institute, NUS, Singapore
| |
Collapse
|
49
|
Du L, Wang Q, Ji S, Sun Y, Huang W, Zhang Y, Li S, Yan S, Jin H. Metabolomic and Microbial Remodeling by Shanmei Capsule Improves Hyperlipidemia in High Fat Food-Induced Mice. Front Cell Infect Microbiol 2022; 12:729940. [PMID: 35573781 PMCID: PMC9094705 DOI: 10.3389/fcimb.2022.729940] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Hyperlipidemia refers to a chronic disease caused by systemic metabolic disorder, and its pathophysiology is very complex. Shanmei capsule (SM) is a famous preparation with a long tradition of use for anti-hyperlipidemia treatment in China. However, the regulation mechanism of SM on hyperlipidemia has not been elucidated so far. In this study, a combination of UPLC-Q-TOF/MS techniques and 16S rDNA gene sequencing was performed to investigate the effects of SM treatment on plasma metabolism-mediated change and intestinal homeostasis. The results indicated that SM potently ameliorated high-fat diet-induced glucose and lipid metabolic disorders and reduced the histopathological injury. Pathway analysis indicated that alterations of differential metabolites were mainly involved in glycerophospholipid metabolism, linolenic acid metabolism, α-linoleic acid metabolism, and arachidonic acid metabolism. These changes were accompanied by a significant perturbation of intestinal microbiota characterized by marked increased microbial richness and changed microbiota composition. There were many genera illustrating strong correlations with hyperlipidemia-related markers (e.g., weight gains, GLU, and total cholesterol), including the Lachnospiraceae NK4A136 group and the Lachnospiraceae NK4B4 group. Overall, this study initially confirmed that hyperlipidemia is associated with metabolic disturbance and intestinal microbiota disorders, and SM can be employed to help decrease hyperlipidemia risk, including improving the abnormal metabolic profile and maintaining the gut microbial environment.
Collapse
Affiliation(s)
- Lijing Du
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Wang
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shuai Ji
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuanfang Sun
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjing Huang
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yiping Zhang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shasha Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shikai Yan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- *Correspondence: Shikai Yan, ; Huizi Jin,
| | - Huizi Jin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Shikai Yan, ; Huizi Jin,
| |
Collapse
|
50
|
Xiao M, Huang M, Huan W, Dong J, Xiao J, Wu J, Wang D, Song L. Effects of Torreya grandis Kernel Oil on Lipid Metabolism and Intestinal Flora in C57BL/6J Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4472751. [PMID: 35464771 PMCID: PMC9023180 DOI: 10.1155/2022/4472751] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 02/05/2023]
Abstract
Background Recent experimental studies have shown that vegetable oil supplementation ameliorates high-fat diet- (HFD-) induced hyperlipidemia and oxidative stress in mice via modulating hepatic lipid metabolism and the composition of the gut microbiota. The aim of this study was to investigate the efficacy of the Torreya grandis kernel oil (TKO) rich in unpolysaturated fatty acid against hyperlipidemia and gain a deep insight into its potential mechanisms. Methods Normal mice were randomly divided into three groups: ND (normal diet), LO (normal diet supplement with 4% TKO), and HO (normal diet supplement with 8% TKO). Hyperlipidemia mice were randomly divided into two groups: HFN (normal diet) and HFO (normal diet supplement with 8% TKO). Blood biochemistry and histomorphology were observed; liver RNA-seq, metabolomics, and gut 16S rRNA were analyzed. Results Continuous supplementation of TKO in normal mice significantly ameliorated serum total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and free fatty acid (FFA) accumulation, decreased blood glucose and malondialdehyde (MDA), and enhanced superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) levels. According to GO and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, most differentially expressed genes (DEGs) were significantly enriched in the biosynthesis of unsaturated fatty acid pathways, and significantly changed metabolites (SCMs) might be involved in the metabolism of lipids. High-dose TKO improved gut alpha diversity and beta diversity showing that the microbial community compositions of the five groups were different. Conclusion Supplementation of TKO functions in the prevention of hyperlipidemia via regulating hepatic lipid metabolism and enhancing microbiota richness in normal mice. Our study is the first to reveal the mechanism of TKO regulating blood lipid levels by using multiomics and promote further studies on TKO for their biological activity.
Collapse
Affiliation(s)
- Minghui Xiao
- The Nurturing Station for the State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, China
| | - Minjie Huang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Weiwei Huan
- College of Chemistry and Materials Engineering, Zhejiang A&F University, Hangzhou 311300, China
| | - Jie Dong
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Jianbo Xiao
- Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo-Ourense Campus, E-32004 Ourense, Spain
| | - Jiasheng Wu
- The Nurturing Station for the State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, China
| | - Deqian Wang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Lili Song
- The Nurturing Station for the State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, China
| |
Collapse
|